cognitive enhancers (nootropics). part 3: drugs ... · 2 w. froestl et al. / cognitive enhancers...

114
Journal of Alzheimer’s Disease 34 (2013) 1–114 DOI 10.3233/JAD-121729 IOS Press 1 Review Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes. Disease-modifying Drugs Wolfgang Froestl , Andrea Pfeifer and Andreas Muhs AC Immune SA, EPFL, Lausanne, Switzerland Accepted 7 October 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. The review covers the evolution of research in this field over the last 25 years. Keywords: Amyloid- aggregation inhibitors, antibodies, antioxidants, cognitive enhancers, metal chelators, natural products, peptides, psychostimulants, tau, vaccines INTRODUCTION As of September 30, 2012, there are 26,788 entries in PubMed under the term cognitive enhancers, 26,781 entries under the term nootropic, and 245 entries under the term cognition enhancers. Scifinder lists 5,133 references under the research topic nootropic, 541 references under the term cognitive enhancer, and 9,853 references for cognition enhancers. The Thom- son Reuters Pharma database lists 1,111 drugs as 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]. 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. 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 (ADHD), or aging. Mark J. Millan and 24 eminent researchers [3] presented an excellent overview on cognitive dysfunction in psychiatric disorders in the February 2012 issue of Nature Reviews Drug Discovery and define cognition as “a suite of interrelated conscious ISSN 1387-2877/13/$27.50 © 2013 – IOS Press and the authors. All rights reserved

Upload: lamanh

Post on 08-Jun-2019

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Journal of Alzheimer’s Disease 34 (2013) 1–114DOI 10.3233/JAD-121729IOS Press

1

Review

Cognitive Enhancers (Nootropics). Part 3:Drugs Interacting with Targets other thanReceptors or Enzymes. Disease-modifyingDrugs

Wolfgang Froestl∗, Andrea Pfeifer and Andreas MuhsAC Immune SA, EPFL, Lausanne, Switzerland

Accepted 7 October 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. The review covers the evolution of research in this field over the last 25 years.

Keywords: Amyloid-� aggregation inhibitors, antibodies, antioxidants, cognitive enhancers, metal chelators, natural products,peptides, psychostimulants, tau, vaccines

INTRODUCTION

As of September 30, 2012, there are 26,788 entriesin PubMed under the term cognitive enhancers, 26,781entries under the term nootropic, and 245 entries underthe term cognition enhancers. Scifinder lists 5,133references under the research topic nootropic, 541references under the term cognitive enhancer, and9,853 references for cognition enhancers. The Thom-son Reuters Pharma database lists 1,111 drugs as

∗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].

nootropic 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.

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 (ADHD), oraging. Mark J. Millan and 24 eminent researchers[3] presented an excellent overview on cognitivedysfunction in psychiatric disorders in the February2012 issue of Nature Reviews Drug Discovery anddefine cognition as “a suite of interrelated conscious

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

Page 2: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

(and unconscious) mental activities, including pre-attentional sensory gating, attention, learning andmemory, problem solving, planning, reasoning andjudgment, understanding, knowing and represent-ing, creativity, intuition and insight, spontaneousthought, introspection, as well as mental time travel,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 ofthe 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 compo-nent binding (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, drugs interacting with enzymeswere described [1696]. Here, in Part 3, we give anoverview on drugs interacting with targets 3 to 10 andcompounds and preparations of categories 11 to 19.

Disease modifying drugs are aimed to counteract theprogression of a disease. In particular for AD, manyexcellent reviews discuss this subject (in chronolog-ical order) [6–27]. �-secretase activators and �- and�-secretase inhibitors (and modulators) were presentedin Part 2 of this review. Drugs interacting with amyloid-� (A�) and tau including immunotherapy are describedin Part 3.

DRUGS INTERACTING WITH CYTOKINES

There is abundant evidence that inflammatory mech-anisms within the central nervous system (CNS)contribute to cognitive impairment via cytokine-mediated interactions between neurons and glial cells.A current hypothesis is that an extracellular insultto neurons could trigger the production of inflam-matory cytokines by astrocytes and microglia [28].Conversely, A� has been shown to induce the expres-sion of interleukin (IL)-1�, tumor necrosis factor-�(TNF-�) and IL-6 in astrocytes and microglia in cul-ture [29]. One genome-wide analysis in 691 subjects ofmean age of 72.6 years showed that raised chemokine(C-C motif) receptor 2 (CCR2) expression was themost strongly associated transcript with lower Mini-Mental Status Exam scores and accelerated decline inscore over a period of 9 years [30]. Decline in scoreover a period of 9 years [30]. The expression profiles ofcytokines in the brains of Alzheimer’s disease patientswere compared to the brains of non-demented patients[1697]. Also see 11. Antioxidants, Also see 11. Antiox-idants, because inflammation is tightly connected withthe oxidative cascade.

TT-301 (MW01-6-189WH; MW-189; TransitionTherapeutics, Toronto following its acquisition of Neu-roMedix under license from Northwestern University)is an inhibitor of microglial cell activation and pro-inflammatory cytokine production for the potential i.v.treatment of CNS diseases including AD. Preclinicalcharacterization of the suppression of brain proinflam-matory cytokine upregulation was carried out [31]. Adouble-blind, randomized, placebo-controlled Phase Iclinical trial in healthy male subjects (n = 16) startedin the US in May 2011 (Thomson Reuters Pharma,update of September 14, 2012). The structure was notcommunicated.

Page 3: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 3

Fig. 1. Drugs interacting with cytokines and with gene expression.

TT-302 (MW01-7-084WH, MW-084; TransitionTherapeutics, Toronto; Fig. 1) is an inhibitor of pro-inflammatory cytokine production by activated glia forthe potential oral treatment of CNS disorders includingAD, traumatic brain injury and inflammatory diseasessuch as arthritis [32]. Phase I clinical trials are expectedfor 2012 (Thomson Reuters Pharma, update of Septem-ber 29, 2012).

Minozac (MW01-2-151SRM, MW-151, TransitionTherapeutics, Toronto following its acquisition of Neu-roMedix, under license from Northwestern University)(Fig. 1) is an inhibitor of pro-inflammatory cytokineproduction by activated glia for the potential treatmentof CNS diseases including AD, Parkinson’s disease(PD), multiple sclerosis, and traumatic brain injury[33–36] (Thomson Reuters Pharma, update of August13, 2012).

AD-16 (Guangzhou Institutes of Biomedicine andHealth) is the thiophene analogue of Minozac. Itreduced amyloid-� induced spatial learning and mem-ory impairment as potently as donepezil in anAlzheimer’s mouse model [1698]. (Thomson ReutersPharma, update of October 19, 2012).

SEN-1176 (Senexis, Cambridge, UK) (Fig. 1) is apyrrolo[3,2-e][1,2,4]triazolo[1,5-a]pyrimidine, whichsuppresses A�42-induced macrophage production ofnitric oxide, TNF-�, IL-1�, and IL-6 in a dose-dependent fashion, an activity profile consistent with aneuroinflammation inhibitor [1711] (Thomson ReutersPharma, update of June 24, 2011).

Infliximab (Janssen Biotech, Horsham, PA),a launched monoclonal antibody against TNF-�,improved cognition after intrathecal administration ina woman with AD [38].

The University of Zurich is investigating an antibodyagainst the interleukin-12 subunit p40 for the poten-tial treatment of Alzheimer’s disease [1699] (ThomsonReuters Pharma, update of November 26, 2012).

The development of REN-1189 (formerly CPI-1189; Centaur Pharmaceuticals), a TNF-� releaseinhibitor, was terminated. Also the development ofSemapimod (CNI-1493; Cytokine PharmaSciences,CPSI, formerly Cytokine Networks), a cytokineinhibitor, which inhibited A� production, plaque for-mation, and cognitive deterioration in an animal modelof AD was suspended [39, 40].

Page 4: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

4 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

DRUGS INTERACTING WITH GENEEXPRESSION

Gene therapy in AD, a potential for disease modi-fication, was discussed [1715]. An interesting reviewon genes and the parsing of cognitive processes wascommunicated [42]. Bi-phasic change in brain-derivedneurotropic factor (BDNF) gene expression follow-ing antidepressant drug treatment was described [43,44]. The modulation of Nur77 and Nor-1 expressionby dopaminergic drugs was elucidated [45]. The reg-ulation of GABAA receptor subunit expression bypharmacological agents was investigated [46]. Genetherapy in mouse models of Huntington’s disease (HD)was reviewed [47].

Beperminogene perplasmid (AMG-0001; DS-992; Collategene; AnGes MG, formerly MedGeneBioscience, Osaka, in collaboration with DaiichiSankyo) is a hepatocyte growth factor (HGF)plasmid-based gene therapy for i.m. injection. Itshowed nootropic properties with a potential for thetreatment of PD. In November 2011, a Phase III trialfor peripheral arterial disease was expected to be initi-ated in 2012. In April 2012, the Phase III studies werestill in preparation [48]. A long-term follow-up eval-uation of results from the clinical trial TREAT-HGFwas reported [49] (Thomson Reuters Pharma, updateof September 19, 2012).

RVX-208 (Resverlogix, Calgary) (Fig. 1) is anapolipoprotein A1 (ApoA1) gene expression stim-ulator for the potential prevention of A� plaqueaccumulation in AD. In January 2011, data from ananalysis of AD biomarkers in the Phase II ASSERTtrial in 299 patients showed that after 12 weeksof treatment with 150 mg/day, a positive effect onA�40 was seen. A compilation of data on RVX-208was published in Drugs in R & D [50]. The ratio-nale for the SUSTAIN study (172 patients) and theASSURE study (310 patients) was presented [51].Preclinical evaluations were reported [52, 53] andclinical data were provided [54–56]. RVX-208 sig-nificantly increased high-density lipoprotein (HDL)-C(p = 0.001), the primary endpoint of the SUSTAIN trial,a Phase 2b clinical study. SUSTAIN also successfullymet secondary endpoints, showed increases in levels ofApo-AI (p = 0.002) and large HDL particles (p = 0.02),both believed to be important factors in enhancingreverse cholesterol transport activity. In July 2012, thecompany was planning to initiate a Phase II trial ofRVX-208 in patients with mild cognitive impairment(MCI) in the second half of 2013 (Thomson ReutersPharma, update of September 27, 2012).

There are several drugs interacting with gene expres-sion in preclinical evaluation (in alphabetical order):

AAV-CYP46A1 (INSERM in collaboration withsanofi) is an adeno-associated virus (AAV) 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 APP23 mice. Significantimprovements in cognitive function assessed by theMorris water maze were seen in Tau22 mice [57](Thomson Reuters Pharma, update of June 6, 2012).See also Part 2, Chapter 2.12. Drugs interacting withcholesterol 24-hydroxylase (CYP46A1).

AZ-AAV9 (RegenX Biosciences, Washington DC)is an AAV vector-9 that carries neuroprotective genesfor the potential injectable treatment of AD (ThomsonReuters Pharma, update of July 27, 2012).

HSD17B10 is a gene, which encodes HSD10, amitochondrial multifunctional enzyme that plays asignificant part in the metabolism of neuroactivesteroids and the degradation of isoleucine. Elevatedlevels of HSD10 were observed in the hippocampi ofAD patients [58].

PRO-289 (Prosensa Therapeutics, Leiden, theNetherlands) is the lead from a series of single-strandedRNA-based antisense oligonucleotide-based therapeu-tics to inhibit the CTG trinucleotide repeat expansionof the huntingtin gene (HTT) by preventing the cellu-lar production of aberrantly expanded HTT mRNA andmutant huntingtin protein (Thomson Reuters Pharma,update of February 10, 2012).

SynCav (Raft Therapeutics, San Diego CA) isa gene therapy that upregulates synaptic drivenCaveolin-1 to promote regeneration of neurons forthe potential treatment of neurodegenerative diseasessuch as AD, PD, HD, and amyotrophic lateral sclerosis(ALS) (Thomson Reuters Pharma, update of August10, 2012).

Intrahippocampal gene transfer of F-spondin, anactivator of the reelin pathway, improved spatial learn-ing/memory in the Morris water maze and increasedthe exploration of the novel object in the Novel ObjectRecognition test in wild-type mice [59].

lnventiva (Daix, Bourgogne, France) is investigat-ing compounds that increase expression of a target geneunder epigenetic control resulting in increased levels ofa secreted neuronal protein for the potential treatmentof Alzheimer’s disease. (Thomson Reuters Pharma,update of November 23, 2012). Structures were notcommunicated.

Neurologix under license from Keio University wasinvestigating colivelin, a hybrid peptide composed of

Page 5: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 5

activity-dependent neurotrophic factor [60–64] and ahumanin derivative for the potential gene therapyof AD [65, 66]. The program was terminated. Thedevelopment of LX-6171 (Lexicon Pharmaceuticals,formerly Lexicon Genetics), a small molecule inhibitorof the SLC6A gene and the LG617 receptor, was dis-continued as well.

DRUGS INTERACTING WITH HEATSHOCK PROTEINS

Binding of heat shock protein 90 (HsP90) to taufacilitates a conformational change that results inits phosphorylation by glycogen synthase kinase 3�(GSK-3�) and its aggregation into filamentous struc-tures [67]. HsP90 regulates tau pathology throughco-chaperone complexes [68].

HsP90 inhibitors, such as PU-H71, PU-3, PU24FCl(Fig. 2) and PU-DZ8 of the purine class of HsP90inhibitors from the Memorial Sloan-Kettering Instituteof Cancer Research, New York caused an eliminationof aggregated tau [69–75].

PU-H71 is in Phase I clinical trials since July 2011(n = 40) in the US. The study was expected to be com-plete by July 2013 (Thomson Reuters Pharma, updateof April 24, 2012).

ALS Biopharma (Doylestown, PA) is investigatingsmall-molecule brain-penetrant modulators of HsP70for the potential treatment of neurological disordersincluding AD (Thomson Reuters Pharma update ofJanuary 17, 2012).

Conforma Therapeutics (San Diego, CA) presenteda new class of HsP90 inhibitors [76–78] and showedthat E102 (structure not disclosed) promoted selec-tive decrease of the P-tau species in a mouse modelof tauopathy.

KU-32 (University of Kansas, Fig. 2) is a novo-biocin derivative acting as an inhibitor of Hsp90 and aninducer of Hsp70 for the potential treatment of neuro-logical diseases. In transgenic mice expressing humanP301L-mutant tau KU-32 (10 mg/kg for 15 weeks)reduced cortical levels of CP13-Iabeled tau [1700].(Thomson Reuters Pharma, update of May 31, 2012).

Lundbeck follows up another series of HsP90inhibitors (Thomson Reuters Pharma update of March31, 2011).The structures were not disclosed.

Fig. 2. Heat shock protein 90 inhibitors and a thyrotropin-releasing hormone analogue.

Page 6: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

6 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

A� accumulation decreased expression of the heatshock-protein 70-interacting protein (CHIP), whichfunctions as a tau ubiquitin ligase [79]. CHIP is akey molecular link between A� and tau patholo-gies. Increasing CHIP levels may have beneficialeffects to decrease tau pathology, because CHIP canpolyubiquitinate tau and may play a crucial role inpreventing accumulation of phospho-tau and neurofib-rillary tangles. Heat shock protein 70 prevented bothtau aggregation and the inhibitory effect of preexist-ing tau aggregates on fast axonal transport [80]. Anexchange of HsP70 for HsP90 is involved in tau degra-dation [1701].

Excellent reviews on heat shock proteins were pre-viously published [81, 82].

The development of AEG-33773 (Aegera Thera-peutics), an allosteric modulator of HsP90 leading toHsP70 upregulation, was terminated.

DRUGS INTERACTING WITH HORMONES

Sustained efforts went into the exploration of thecognition enhancing effects of thyrotropin-releasinghormone and its analogues since it had been recog-nized as an activator of brain cholinergic systems [83].Pilot studies of intravenous thyrotropin-releasing hor-mone in AD were undertaken [84, 85]. Improvement ofcognitive deficits in depressed patients were reported[86].

Taltirelin (Ceredist, TA-0910; Mitsubishi TanabePharma) (Fig. 2), an orally active synthetic thyrotropin-releasing hormone analogue, was launched in Japan in2000 for the treatment of neurodegenerative disease, inparticular spinocerebellar degeneration [87–89]. Thesales in 2011 were USD 226.8 million (ThomsonReuters Pharma, update of August 30, 2012).

KPS-0373 (Kissei under license from Shionogi)is a thyrotropin-releasing hormone derivative for thepotential oral treatment of spinocerebellar ataxia inPhase II clinical trials in Japan (Thomson ReutersPharma, update of July 31, 2012). The structure wasnot communicated.

Leuprolide acetate implant (VP-4896, Memryte;Curaxis Pharmaceuticals, Raleigh NC, formerly Voy-ager Pharmaceutical and DURECT) is a biodegradableimplant formulation of the gonadotropin-releasinghormone agonist 1-9-luteinizing hormone-releasingfactor-6D-leucine-9-(N-ethyl-L-prolinamide), a non-apeptide, for the treatment of AD in women. APhase IIb clinical study plan is currently evalu-ated. The effects of treatment with leuprolide acetate

depot on working memory in women were described[90]. Luteinizing hormone modulated the process-ing of amyloid-� protein precursor (A�PP) andA� deposition [91] and of cognition in transgenicmice [92]. Several reviews were published [93–96](Thomson Reuters Pharma, update of August 24,2012).

Tesamorelin (Egrifta; TH-9507; Theratechnolo-gies, Quebec and US commercialization partner EMDSerono and licensee Sanofi) is a stabilized, trun-cated growth hormone releasing factor (GRF1-44;ThGRF1-44). In a controlled study of 152 adults (66with MCI) participants self-administered daily subcu-taneous injections of tesamorelin or placebo for 20weeks, which had favorable effects on cognition in bothadults with MCI and healthy older adults [97] (Thom-son Reuters Pharma, update of August 15, 2012).

The development of azetirelin (YM-14673;Yamanouchi, now Astellas [98, 99]), of cal-citriol (Neurocalc; Apollo Biopharmaceuticals,now MitoKor), of JTP-2942 (Japan Tobacco[100–102]), of montirelin (CG-3703; CNK-602A;NS-3; Grunenthal [103]), of orotirelin (CG-3509;Grunenthal [104–106]), of posatirelin (RGH-2202;Gedeon Richter [107–109]), of protirelin (DN-1417;Takeda [110]), of thyrotropin-releasing hormoneanalogs (Roche), and of thymoliberin (RX-77368;Reckitt & Colman [111]) was terminated.

DRUGS INTERACTING WITH IONCHANNELS (NON-RECEPTORS)

Dysregulation of Ca2+ homeostasis plays a crucialrole in the pathogenesis of AD. In the pathogene-sis of AD [1702]. Oligomeric A� proteins directlyincorporated into neuronal membranes, formed cation-sensitive ion channels (“amyloid channels”) andcaused disruption of calcium homeostasis [112, 113].A� oligomers directly and dose-dependently mod-ulated P/Q-type calcium channels [114]. In tripletransgenic AD mice, upregulated ryanodine receptoractivity led to a reduction of long-term potentiation[115]. Mutations of presenilin can affect the intra-cellular calcium levels via the endoplasmic reticulumcalcium stores [116]. The levels of the calcium-sensingenzyme hippocalcin were elevated in AD brains [117].Interesting reviews were published [118–121].

Drug repositioning for the treatment of Alzheimer’sdisease was described recently [1703].

ARC-029 (Archer Pharmaceuticals, Roskamp Insti-tute, Sarasota FL) is a blood-brain barrier crossing

Page 7: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 7

Fig. 3. Two ion channel blocker and three NGF and BDNF stimulators.

formulation of nilvadipine (Fig. 3), a blocker ofL-type voltage-gated calcium channels. The drug hasbeen approved for a multisite Phase III clinical trialenrolling more than 500 AD patients in Europe. InFebruary 2012, the study was expected to begin in thesecond half of 2012 (Thomson Reuters Pharma, updateof April 16, 2012).

Nilvadipine (FR-34235; Fujisawa; launched inJapan in 1989) was extensively characterized in vitroand in vivo for the potential treatment of AD. It facili-tated the clearance of A� across the blood-brain barrier[122, 123]. It prevented the impairment of spatial mem-ory induced by cerebral ischemia combined with A�in rats [124, 125]. It also antagonized A� vasoactiv-ity [126]. First positive clinical results on nilvadipinein AD patients were communicated. It was safe andprovided short-term cognitive benefits in AD patients[127–131].

ARC-031 and ARC-031-SR (Archer Pharmaceuti-cals; sustained release formulation) is a non-calciumchannel blocking and soluble A� reducing nilvadipinederivative in Phase I clinical trials (Thomson ReutersPharma, update of January 3, 2012). The structure wasnot communicated.

RNS-60 (Revalesio Corporation, Tacoma WA) actson voltage-gated ion channels. It is formulated as

an inhalant anti-inflammatory charge-stabilized nanos-tructure. The company investigates also the potentialtreatment in AD and PD. A Phase I/IIa asthma trialbegan in May 2010. In October 2011, data from themulti-dose stage of the Phase I study were reported.RNS-60 showed a therapeutic benefit and was safe atall doses (Thomson Reuters Pharma, update of April27, 2012). The structure was not communicated.

ZSET-1446 (ST-101, Sonexa Therapeutics, SanDiego CA under license from Zenyaku Kogyo) (Fig. 3)is in Phase II clinical trials for AD since February 2009.A second Phase II clinical trial for the treatment ofessential tremor was initiated in the US in May 2011.It was recognized that ZSET-1446 (ST-101) targetedT-type voltage-gated calcium channels in mediatingimproved cognition in the CNS [132]. In vivo data werepublished previously [133, 134] (Thomson ReutersPharma, update of July 17, 2012).

AD-N02 (Adamed, Mazowieckie, Poland) is asodium channel blocker and dopamine/5-HT stabilizerfor the potential treatment of schizophrenia and bipolardisorder (Thomson Reuters Pharma, update of August22, 2012). The structure was not communicated.

Isradipine (Novartis, launched in 1997) is aL-type voltage-gated calcium channel blocker pro-tecting MC65 neuroblastoma cells from A�PP

Page 8: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

8 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

C-terminal fragment (A�PP-CTF)-induced neurotox-icity [135–137, 1704].

The development of many drugs interacting withion channels was terminated (in alphabetical order):DMP-543 (Bristol-Myers Squibb; an inhibitor of M-type K+ channels), enecadin (PAION under licensefrom Nippon Shinyaku; a sodium and calcium channelblocker), LB-101, LB-102, LB-217, LB-218, LB-253,LB-269, LB-301, and LB-302 (Lifelike Biomatic;modulators of two ionic channels, i.e., ionokines),linopirdine (DuP 996; Du Pont Merck, Bristol-MyersSquibb; an inhibitor of M-type K+ channels; despitepromising EEG brain mapping data [138] and anincrease of parietal regional cerebral blood flow inAD patients [139], the clinical results were disap-pointing [140]), MEM-1003 (BAY-Z-4406, MemoryPharmaceuticals under license from Bayer; a L-typevoltage-gated calcium channel [141]), nerispirdine(HP-184, Hoechst, now sanofi; an inhibitor of M-type K+ channels), nicardipine (YC-93, Yamanouchi[142]), nifedipine (BAY-A-1040, Adalat, Bayer[143]), nimodipine (BAY-E-9736, Nimotop, Bayer[144–152]), nitrendipine (BAY-E-5009; Bayotensin,Bayer; all L-type voltage-gated calcium channels[153, 154]), NP-34 (NP-04634; Noscira, formerlyNeuropharma; a dual calcium channel blocker andacetylcholinesterase inhibitor [155]), NS-649 (Neu-roSearch; a neuron-specific calcium channel blocker[156]), NSD-761 (NeuroSearch; a selective ion chan-nel modulator); RGH-2716 (TDN-345; a nootropicneuroselective ion channel blocker; Takeda in collab-oration with Gedeon Richter [157–159]), SNX-482(R-type calcium channel blocker of Neurex, nowElan Pharmaceuticals [160–162]), SPI-017 (Sucampo;selective type-2 chloride channel activator), tamo-larizine (NC-1100, calcium channel blocker, NipponChemiphar [163–165]), VRX-698 (Valeant Pharma-ceuticals International; a potassium channel opener),and XE-991 (Bristol-Myers Squibb; a KCNQ potas-sium channel blocker [166, 167]).

DRUGS INTERACTING WITH NERVEGROWTH FACTORS

The role of neurotrophic factors in AD was dis-cussed [168] as was a neurotrophic rationale forthe therapy of neurodegenerative diseases [169]. Thepotential therapeutic use of BDNF, a key regulatorfor protein-synthesis dependent long-term potentiationand long-term memory, in neurological and psychiatricdisorders was presented [170, 171], in particular for

AD [172, 173]. Interestingly, genetic knockdown ofBDNF in triple transgenic AD mice did not alter A�or tau pathology [174]. Nerve growth factor (NGF)-cholinergic dependency in brain aging, MCI, and ADwas discussed [175–181]. NGF treatment in demen-tia was described [182]. NGF and AD, new factsfor an old hypotheses were communicated recently[183]. Hippocampal ProNGF signaling pathways andamyloid-� levels in mild cognitive impairment andAlzheimer’s disease were elucidated [1705]. It wasclaimed that NGF promoted long-term memory for-mation by activating poly(ADP-ribose) polymerase-1(PARP-1) [184].

NeuroAid (MLC-601; Danqi Plantan Jiaonang;Moleac Pte Ltd, Singapore) is a BDNF stimulatorderived from Radix Astragali, Radix Salvia Miltior-rhizae, Radix Paeoniae Rubra, Rhizoma Chuanxiong,Radix Angelicae Sinensis, Carthamus Tinctorius,Prunus Persica, Radix Polygalae, Rhizoma AcoriTatarinowii, Buthus Martensii, Hirudo, EupolyphagaSeu Steleophaga, Calculus Bovis Synthetic or Arti-factus and Cornu Saigae Tataricae, for the potentialoral prevention of stroke including cerebral infarctionand ischemic stroke and for the potential treatmentof traumatic brain injury in a randomized, double-blind, placebo-controlled, multicenter, Phase III trial(n = 1100) since November 2007 in Singapore and thePhilippines. The study was expected to be completedin 2012 (Thomson Reuters Pharma, update of October3, 2012).

CERE-110 (AAV2-NGF; NeuroRescue AD; Cere-gene, San Diego CA) is an AAV2 vector based genedelivery system containing cDNA for NGF. A Phase IIclinical trial in mild-to-moderate AD patients (n = 50)started in May 2009 in the US. The therapeutic poten-tial of CERE-110 was described in detail [185, 186](Thomson Reuters Pharma, update of July 20, 2012).

GM-607 (Genervon Biopharmaceuticals, PasadenaCA) is a motoneuronotrophic factor analogue for thepotential treatment of ischemic stroke, spinal cordinjuries, ALS, AD, HD, and PD. By April 2011, aPhase II ischemic stroke trial was initiated. By Febru-ary 2012, further Phase II IND applications werebeing prepared for spinal cord injuries, ALS, PD, andAD (Thomson Reuters Pharma, update of August 28,2012).

MIM-D3 (Mimetogen Pharmaceutics, Quebec,Fig. 3) is a small molecule NGF peptidomimetic forthe treatment of AD and dry eye (xerophthalmia). APhase II trial for dry eye was initiated in November2010 [187] (Thomson Reuters Pharma, update of May17, 2012).

Page 9: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 9

PYM-50028 (Cogane, P58, P63; Phytopharm, UK)(Fig. 3), a steroidal saponin, stimulated BDNFrelease and may be beneficial for the treatmentof PD, AD, glaucoma, and ALS. PYM-50028reversed neuronal damage induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MTPT) in a mousemodel of PD [188]. In July 2011, FDA grantedPYM-50028 orphan drug status for ALS. In Novem-ber 2010, recruitment of patients (n = 400) beganin the randomized, double-blind, proof-of-concept,placebo-controlled, dose-ranging Phase II studyCONFIDENT-PD (Thomson Reuters Pharma, updateof August 16, 2012).

T-817MA (Toyama Chemical, Fujifilm Holding)(Fig. 3) is a neurotrophic small molecule in PhaseII clinical trials for AD in the US since April 2008.T-817MA attenuated cognitive impairments in P301Ltau transgenic mice [189], prevented memory deficitscaused by i.c.v. A� administration [190, 191], andattenuated A�-induced neurotoxicity [192] (ThomsonReuters Pharma, update of July 30, 2012).

NsG-0202 (ECB-AD, ECT-AD, NsGene A/s, Den-mark) is an encapsulated cell biodelivery system todeliver cells expressing NGF. A Phase Ib trial startedin December 2007 in Sweden. First clinical data werepresented in July 2011. In January 2012, the trial wasexpected to be completed in 2012 [193] (ThomsonReuters Pharma, update of January 17, 2012).

There are several drugs interacting with neu-rotrophic factors in preclinical evaluation (in alpha-betical order):

AL-209 (ADNF-9, SAL; Allon Therapeutics, Van-couver) is an activity-dependent neurotrophic factorderived nine amino acid sequence peptide and tubulin-binding agent that stimulated PARP-1 for the potentialtreatment of CNS and ocular disorders (ThomsonReuters Pharma, update of March 6, 2012).

AL-309 (Allon Therapeutics, Vancouver) is ananalogue of AL-209 and PARP stimulator for oral,intranasal and s.c. administration. By October 2012preclinical development had been completed (Thom-son Reuters Pharma, update of October 17, 2012).

AMRS-001 (CNS-001; Amarantus BioSciences,Sunnyvale CA, formerly CNS Protein Therapeuticshaving acquired the relevant IP from Prescient Neu-ropharma) is a program of therapeutics based onmesencephalic astrocyte-derived neurotrophic factor[194], which promoted the survival of dopaminergicneurons for the potential treatment of PD, AD, andbrain injury. It also protected the heart from ischemicdamage [195] (Thomson Reuters Pharma, update ofOctober 2, 2012).

Catecholamine derivatives (Emory University,Atlanta, GA) acted like BDNF to activate the tyrosinekinase B (TrkB) receptor. They have a potential for thetreatment of neurological diseases such as ALS, PD,and AD (Thomson Reuters Pharma, update of July 27,2012). Structures were not communicated.

CB-1, CB-2, and CB-3 (Molcode, Tartu, Estonia)are BDNF mimetics, which activated the TrkB recep-tor leading to its phosphorylation (Thomson ReutersPharma, update of December 27, 2011). The structureswere not communicated.

7,8-Dihydroxy-flavone, a TrkB receptor agonistand BDNF mimic, reversed memory deficits in a mousemodel of AD [196]. The compound and derivativesthereof are also evaluated at Emory University in col-laboration with the University of Wisconsin [197–199](Thomson Reuters Pharma, update of August 10,2012).

FC29 peptide (University of Queensland, Brisbane,Australia) is a fragment of the p75 neurotophin recep-tor that inhibited p75NTR-mediated neuronal deathvia regulation of the Trk receptor function for thepotential treatment of AD, PD, and motor neuron dis-ease (Thomson Reuters Pharma, update of May 17,2012).

Gambogic amine and gambogic amide (EmoryUniversity, Atlanta GA), TrkA receptor agonists withrobust neurotrophic activity, prevented neuronal celldeath [200] (Thomson Reuters Pharma, update ofAugust 28, 2012).

Gedunin (Emory University, Atlanta GA) and itsderivatives including deoxygedunin are TrkB receptoragonists for the potential treatment of PD, HD, AD,HIV-related dementia, ALS, stroke, and multiple scle-rosis [201] (Thomson Reuters Pharma, update of April26, 2012).

JRP-655 (Prous Institute for Biomedical Research,Barcelona) is a BDNF secretagogue and neuroplastic-ity modulator (Thomson Reuters Pharma, update ofSeptember 5, 2012). The structure was not communi-cated.

4-methylcatechol, which increased BDNF contentand stimulated BDNF mRNA expression and synthe-sis, effectively improved spatial learning and memoryin rats [202, 203].

NeuroAiD II (MLC-901, Moleac Pte Ltd., Singa-pore), a BDNF stimulator, is a derivative of NeuroAiDfor the potential treatment of global ischemia inpreclinical development (Thomson Reuters Pharma,update of September 2, 2011).

ND-602 (Neurodyn, Charlottetown, Canada) is aneuroprotective progranulin (PC cell-growth factor)-

Page 10: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

10 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

expressing lentiviral vector for the potential treatmentof PD, AD, and ALS. ND-602 was found to increaseactivity of neprilysin and significantly reduced A� andplaque burden in the hippocampus and entorhinal cor-tex of Tg2576 mice (Thomson Reuters, update of May25, 2012).

The development of many drugs interacting withneurotrophic factors was terminated. The mostadvanced drug, xaliproden (SR-57746A, Xaprila;sanofi), a 5-HT1A agonist and stimulator of endoge-nous neurotrophin synthesis, did not meet the cognitionendpoints in Phase III trials in AD patients, whichapplies also to paliroden (SR-57667B; with a biphenylinstead of the �-naphtyl in xaliproden; sanofi, [204]).The lessons learnt from the xaliproden clinical trialswere discussed [205]. The biological characterizationwas reviewed [206].

Also the development of other drugs inter-acting with neurotrophic factors (in alphabeticalorder) was terminated: ABS-200 (American Bio-genetic Sciences), ADNF-14 (NIH, activity-dependentneurotrophic factor-14), AGT-120, and AGT-140(ArmaGen Technologies); AK-30-NGF (a mono-clonal antibody as carrier for the delivery of NGF;Alkermes), arundic acid (MK-0724, ONO-2506, Aro-cyte, Ono Pharmaceuticals [207–210]), beta NGF(tethered to a molecular shuttle; Apollo Life Sci-ences), CEP-427 (a neurotrophic factor enhancingsmall molecule; Cephalon, now Teva), coleneuramide(MCC-257; Mitsubishi Tanabe Pharma), CX-438(Cortex Pharmaceuticals), dekafin-1 (a fibroblastgrowth factor receptor, agonist and tyrosine kinasemodulator; Enkam Pharmaceuticals); dFGF (dimer-ized fibroblast growth factor; Massachusetts Instituteof Technology (MIT)/ViaCell Inc.), GDF-1 (CreativeBiomolecules, now Curis; a growth and differen-tiation factor-1 agonist), glial maturation factor(Rhone-Poulenc/Regeneron), huIM-13 (a humanizedmonoclonal antibody that binds to the TrkA receptor,Lay Line Genomics), inosine (an NGF, Alseres Phar-maceuticals, formerly Boston Life Sciences (BLSI),under license from the Children’s Hospital of Boston),KP-66, KP-447, and KP-546 (Krenitsky), leteprinim(AIT-082; SPI-205, Neotrofin; Spectrum Pharmaceu-ticals, formerly NeoTherapeutics), which induced theexpression of NGF, neurotrophin-3, and of basicfibroblast growth factor [211–216], LM11A-31 (a p75neurotrophin receptor ligand; Elan), nerve growthfactor agonists (Chiron), neuregulin-2 (cerebellum-derived growth factor, NRG2; Acorda Therapeuticsunder license from CeNeS and Harvard Univer-sity), NGF therapy (Lay Line Genomics), NP-901

(Noscira), NS-521 (NeuroSearch), p75 NGF recep-tor antagonist (Circadian Technologies under licensefrom the Walter & Eliza Hall Institute), NT-3 (Genen-tech and StemCells), ReN-1820 (ReNeuron underlicense from the University of Bristol), ST-857 (acetyl-L-carnitine arginine amide; Sigma-Tau [217–219]),T-588 (Toyama [220–227]), and trofexin (a peptideneurotrophic factor; Protarga).

DRUGS INTERACTING WITH RE-UPTAKETRANSPORTERS (PSYCHOSTIMULANTS)

Psychostimulants exert behavioral-calming andcognition-enhancing actions in the treatment of ADHD[228]. Cognition-enhancing doses of psychostimulantsexert regionally restricted actions elevating extra-cellular catecholamine levels and enhancing neuralsignal processing preferentially in the prefrontal cor-tex. Additional evidence suggests a prominent role ofprefrontal cortex �2 and D1 receptors in the behav-ioral and electrophysiological actions of low-dosepsychostimulants [229, 230]. Excellent reviews onattention-modulating effects of cognitive enhancerswere published [231–236]. An analysis of studentuse (and abuse) of cognitive enhancers was presented[237]. A review on glycine transporter type 1 (GlyT1)inhibitors was disclosed [238].

Methylphenidate (Ritalin, Ciba, now Novar-tis, launched 1956) is a psychostimulant drug,which increased the levels of dopamine and nora-drenaline in the brain through re-uptake inhibition ofthe monoamine transporters [239]. Methylphenidatepreferentially increased catecholamine neurotrans-mission within the prefrontal cortex at low dosesthat enhance cognitive functions [240]. Cognition-enhancing doses of methylphenidate preferentiallyincreased prefrontal cortex neuronal responsiveness[241]. Methylphenidate decreased the amount of glu-cose needed by the brain to perform a cognitive task[242]. The improvement of memory functions bymethylphenidate (and modafinil) in healthy individualswas described [243].

Most of these experiments were made withthe mixture of erythro and threo stereoisomers.The active principle of methylphenidate is the(+)-(�R,2R)-threo-methylphenidate hydrochloride(Dexmethylphenidate, Ritadex; Novartis Pharmaunder license from Celgene launched 2002) (Fig. 4),also available as an extended-release oral formulationFocalin XR [244]. The spheroidal oral drug absorp-tion system dexmethylphenidate was described [245](Thomson Reuters Pharma, update of July 27, 2012).

Page 11: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 11

Fig. 4. Psychostimulant cognition enhancers.

Modafinil (Provigil, Lafon Laboratories, laterCephalon, now Teva, launched 1998) is an inhibitor ofdopamine, noradrenaline, and serotonin re-uptake andan �1 adrenoceptor agonist [246]. Modafinil showedimprovement of cognition and attention in patientswith chronic schizophrenia ADHD [247, 248], in par-ticular in the first episode of psychosis [249, 250]. Theneuronal mechanism by which modafinil affects cog-nitive and emotional function in schizophrenic patients

was reviewed [251]. Experiments using BOLD func-tional magnetic resonance imaging (fMRI) in healthyvolunteers showed that modafinil enhanced the effi-ciency of prefrontal cortical cognitive informationprocessing, while dampening reactivity to threateningstimuli in the amygdala, the brain area implicated inanxiety [252]. Modafinil improved attention in well-rested individuals [243]. Modafinil reliably enhancedperformance on several cognitive tests of planning

Page 12: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

12 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

and working memory, but did not improve learningand delayed recall performance in healthy volunteers[253]. It may be useful in the treatment of substanceabuse [254]. It improved working memory and sus-tained attention in cocaine users [255].

The active principle of modafinil is the (R)-enantiomer Armodafinil (Nuvigil; CEP-10953;Cephalon, now Teva launched 2009) (Fig. 4). Thesales of armodafinil in 2011 amounted to USD 266mil (Thomson Reuters Pharma, update of September21, 2012).

Atomoxetine (tomoxetine, Strattera, LY-139603;Lilly; launched 2003) (Fig. 4) is a selective nora-drenaline re-uptake inhibitor for the treatment ofADHD in children and adults [256]. Atomoxetineinhibited noradrenaline and serotonin transporters withKi values of 2.6 and 48 nM, respectively [257]. Ato-moxetine improved response inhibition in ADHDpatients and in healthy volunteers via increased acti-vation in the right frontal gyrus measured via fMRI[258, 259]. Sales in 2011 were USD 620 million(Thomson Reuters Pharma, update of September 26,2012).

Lisdexamfetamine (Vyvanse; Shire Pharmaceuti-cals and New River Pharmaceuticals, launched 2007)(Fig. 4) is a drug for the treatment of ADHD inchildren, adolescents, and adults [260]. Lisdexam-fetamine itself is inactive and acts as a prodrug todextroamphetamine upon cleavage of the lysine por-tion of the molecule. Dextroamphetamine inhibitednoradrenaline, serotonin and dopamine transporterswith respective Ki values of 39, 3,830, and 34 nM[261]. The use of lisdexamfetamine in patients withdementia was described [231]. Sales in 2011 wereUSD 805 million (Thomson Reuters Pharma, updateof September 28, 2012).

Indeloxazine (YM-08054; Elen, Noin,Yamanouchi, Schering-Plough, now Merck andJeil Pharmaceuticals, launched in China and SouthKorea) (Fig. 4) blocked the reuptake of noradrenalinewith IC50 of 3.2 �M and of serotonin with IC50 of0.71 �M. It is also a MAO inhibitor. It showed learningand memory improving effects in many animal tests[262–267].

NS-2359 (NeuroSearch, Fig. 4) is a blocker ofdopamine, noradrenaline, and serotonin re-uptake cur-rently in Phase I clinical trials in Denmark forthe potential treatment of CNS diseases. GSK waspreviously developing NS-2359 for the treatmentfor depression but discontinued the development(Thomson Reuters Pharma, update of September 26,2012).

There are several drugs interacting with re-uptaketransporters in preclinical evaluation (in alphabeticalorder):

AM-285 (Cyclocreatine, CincY; Lumos Pharma,Austin TX based on University of Cincinnati technol-ogy) (Fig. 4) is evaluated for the potential treatmentof creatine transporter deficiency, an X-linked autismspectrum disorder characterized by severe cognitiveimpairment [268]. In June 2012, the FDA awardedcyclocreatine Orphan designation for the treatmentof creatine transporter deficiency (Thomson ReutersPharma, update of August 24, 2012).

AS-1522489-00 (Astellas Pharma) is a glycinetransporter-1 inhibitor, a 1,2,4-triazole derivative, ofwhich only the (S)-atropisomer is biologically active[269, 270] (Thomson Reuters Pharma, update of Jan-uary 27, 2012). The structure was not communicated.

Lu-AA42202 (Lundbeck) (Fig. 4) is a tripledopamine, noradrenaline, and serotonin re-uptakeinhibitor for the potential treatment of major depres-sive disorders and ADHD (Thomson Reuters Pharma,update of December 1, 2010).

MLR-1017 (mesocarb, sydnocarb, sidnocarb,Melior Pharmaceuticals, Exton, PA) (Fig. 4) isa dopamine transporter inhibitor for the potentialtreatment of ADHD and levodopa-induced side effectsin PD. The drug was previously launched in Russia[271, 272] (Thomson Reuters Pharma, update of April12, 2012).

PD-2005 (P2D Bioscience, Cincinnati, OH, identi-cal with AHN 2-005, National Institute of Drug Abuseand US Naval Medical Research Center) (Fig. 4) is aninhibitor of the dopamine transporter for the potentialtreatment of ADHD and for cognitive impairments intraumatic brain injury. It dose-dependently improvedperformance of rats in a delayed-alternation task ofspatial working memory [273] (Thomson ReutersPharma, update of June 12, 2012).

RO-4543338 (Roche) is a well-tolerated glycinetransporter inhibitor, which facilitated drug cue extinc-tion [274, 275]. The structure was not communicated.

Selective Serotonin Re-uptake Inhibitors (SSRIs)showed cognitive enhancing effects [276]. Patientstreated with citalopram showed significant improve-ments on a cognitive subscale. Paroxetine and fluoxe-tine improved most of the tested cognitive functions ina study of 242 depressed patients during one year [277].Other studies with citalopram, fluoxetine, sertraline,and zimelidine did not report improvement of memoryand/or reaction time in demented patients [278].

Thiethylperazine (Torecan) (Fig. 4), a drugapproved by the FDA to relieve nausea and vomiting,

Page 13: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 13

Fig. 5. VMAT2 and DA-reuptake PET ligands.

is an activator of ABCC1 transporters. In a mousemodel of AD expressing the ABCC1 transporter, amarkedly reduced load of A� was measured. Defi-ciency of ABCC1 transporter substantially increasedcerebral A� levels [279] (Thomson Reuters Pharma,update of April 26, 2012).

The development of ASP-2535 (Astellas; aglycine transporter-1 inhibitor [280]), coluracetam(BCI-540, MKC-231; BrainCells Inc. under licensefrom Mitsubishi Tanabe Pharma; an enhancer ofhigh-affinity choline uptake and K+-induced releaseof acetylcholine [281–288]), CP-101 (CogniPharm,an i.v. formulation of modafinil), D-serine re-uptakeinhibitors (Memory Pharmaceuticals, now Roche),flufenoxina (FAES Farma; a dual serotonin and nore-pinephrine re-uptake inhibitor), glycine transporter-1inhibitors (Helicon Therapeutics), JNJ-17305600(NFPS; NPS Allelix; a glycine transporter inhibitor[289]), PD-2007 (P2D Bioscience, a dopamine trans-porter inhibitor), SCH-900435 (Org-25935; Organon,Schering-Plough, now Merck, a glycine transporter-1inhibitor), SSR-103800 [290–292]) and SSR-504734(both sanofi; both glycine transporter-1 inhibitors[293–297]), teniloxazine (sufoxazine, Lucelan,

Metatone, Y-8894, Yoshitomi, Mitsubishi Pharma, anoradrenaline uptake inhibitor [298],[299–301]), andtesofensine (NS-2330, NeuroSearch, a monoaminere-uptake inhibitors of serotonin, dopamine, and nora-drenaline [302]) was terminated. The developmentof tesofensine for the once-daily oral treatment ofobesity in Phase II trials was also suspended [303].

The vesicular monoamine transporter 2(VMAT2) located on the membrane of vesiclesis responsible for storing and packaging neuro-transmitters into monoamine vesicles or granules.Imaging VMAT2 in the brain provides a measurementreflecting the integrity of dopaminergic, noradrenergicand serotonergic neurons [304]. Positron emissiontomography (PET) ligands for the VMAT2 havebecome valuable diagnostic tools.

(+)-[11C]-dihydrotetrabenazine (DTBZ) (Fig. 5)allows the determination of the striatal monoaminergicpresynaptic terminal density. In a study enrolling 20patients with dementia with Lewy bodies (DLB), 25AD patients, 7 patients with frontotemporal dementia,and 19 elderly controls clinicians could accuratelydifferentiate between the different forms of dementia[305–307]. The uptake of (+)-11C-DTBZ in the stria-

Page 14: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

14 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

tum of AD patients mainly reflecting the presynapticdopaminergic system was unchanged compared tocontrols. The striatal uptake in DLB and PD patientswas significantly decreased [308].

18F-Florbenazine (18F-AV-133; 18F-FP-DTBZ;Avid Radiopharmaceuticals, Philadelphia PA, a sub-sidiary of Lilly, under license from University ofMichigan) (Fig. 5) is in Phase III clinical evaluation.In May 2012, an open-label, single-blind, Phase II/IIIstudy (NCT01550484; 18F-AV-133-B04) was initi-ated in patients with undiagnosed movement disorders(n = 150) in the US to assess the safety and efficacyof florbenazine in differentiating PD. At that time, thestudy was scheduled to complete in September 2014.For the synthesis, see [309]; for the binding character-istics in rat striatum and hypothalamus homogenates(Kd = 0.19 and 0.25 nM, respectively), see [310, 311].The whole body biodistribution was studied [312].18F-AV-133 was used for noninvasive assessment ofthe vesicular monoamine transporter type 2 in 17 PDpatients and 6 healthy controls. VMAT2 binding poten-tial was decreased by 81% in the posterior putamen,70% in the anterior putamen, and 48% in the caudatenucleus of PD patients [313].

This ligand is a valid measure of dopaminergicneuron integrity. The in vivo assessment in patientswith DLB and AD was reported [314]. The optimalscanning time window was elucidated [315]. For PETimaging in a MPTP-induced mouse model of PD, see[316] (Thomson Reuters Pharma, update of August 7,2012).

11C-�-CFT and 18F-�-CFT (Fig. 5) proved tobe valuable PET ligands due to their high affinityinteraction with dopamine re-uptake sites. AD patientsshowed a reduction of 11C-�-CFT in putamen and cau-date of about 20%. Thus the putamen and the caudatenucleus were equally affected in contrast to PD, whichshowed predominantly putaminal reduction [317]. InPD, the mean uptake of 18F-�-CFT in the contralat-eral putamen was reduced to 31% and in the ipsilatralputamen to 45%. In the caudate nucleus, the uptakewas reduced contralaterally to 67% and ipsilaterally to77% [318]. For an exhaustive review, see [319].

123I-Ioflupane (123I-FP-CIT; Amersham, now GEHealthcare) (Fig. 5) is a SPECT ligand launched in2000. It is the ligand of choice to differentiate betweenAD and DLB patients [320, 321]. It is valuable forthe diagnosis of Parkinsonian syndromes [322]. SERTdependent 123I-ioflupane uptake allows a more com-prehensive assessment of neurochemical disturbancesin degenerative Parkinsonism [323–325] (ThomsonReuters Pharma, update of August 14, 2012).

DRUGS INTERACTING WITHTRANSCRIPTION FACTORS

cAMP response element-binding (CREB) proteinand the discovery of cognitive enhancers was reviewed[326–328]. CREB phosphorylation as a mechanis-tic marker in the development of memory enhancingAD therapeutics was described [329]. The currentknowledge of key calcium signal-regulated transcrip-tion factors, namely CREB, nuclear factor of activatedT-cells, and downstream regulatory element antago-nist modulator (DREAM) and memory formation wassummarized [330]. A� disrupted activity-dependentgene transcription required for memory through theCREB-regulated transcription co-activators CRTC1[331]. The GABAB receptor antagonist SGS742improved spatial memory and reduced protein bind-ing to CREB2 in the hippocampus [332] (see Part1, Chapter 1.10.2 GABAB receptors). CREB anti-sense oligonucleotide administration into the dorsalhippocampal CA3 region impaired long- but not short-term spatial memory in mice [333].

The lack of DREAM protein enhanced learning andmemory and slowed brain aging [334].

SRF/MYOCD inhibitors (Socratech LLC,Rochester NY) are small molecule inhibitors againstserum response factor and myocardin, which areinteractive transcription factors in vascular smoothmuscle cells for the potential treatment of AD[335]. For a review on the imbalance of vascularmolecules in AD, see [336] (Thomson ReutersPharma, update of February 15, 2012). Structureswere not communicated.

The development of CREB modulators (HeliconTherapeutics) and of EPAC inhibitors (exchange pro-tein directly activated by cAMP, a guanine nucleotide-exchange factor; Scottish Biomedical [337]) wasterminated.

ANTIOXIDANTS

Soluble A� oligomers localize to mitochondriaand interfere with their normal functioning caus-ing an overproduction of reactive oxygen species(ROS), inhibiting respiration and ATP production anddamaging the structure of mitochondria [338–340].Some authors, however, claim that the early impair-ments of mitochondrial dysfunction and oxidativestress may precede A� overproduction and depo-sition [341–343]. Excellent reviews were presentedover years [344–347]. Possibilities of intervention ofantioxidant pathways in AD were discussed [348, 349].

Page 15: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 15

Fig. 6. Mitochondria-targeting antioxidants.

Also see Chapter 3. Cytokines, as inflammation istightly connected with the oxidative cascade.

Over the years, clinicians have explored treatmentof AD patients with antioxidants. Free radicals canbe scavenged by dietary means. Compounds such asacetyl-L-carnitine, curcumin [350], Gingko bilobaextracts such as EGb 716, (R)-α-lipoic acid, mela-tonin, morin, trolox, vitamin C [351], and vitamin E[352–355] were tested in clinical trials in AD patients.The results were disappointing [356–372]. As a conse-quence of a landmark study, clinicians even advise ADpatients not to take large doses of vitamin E or �-lipoicacid [373].

Many clinical trials were also carried out withsynthetic antioxidants such as edaravone (MCI-186,Mitsubishi [374],[375–377]), idebenone (CV-2619,Avan, Catena, Noben, Sovrima; Takeda [378–380]),and the derivatives of 21-aminosteroids (Upjohn’slazaroids, such as tirilazad or U-74006F, U-74389G,U-74500A, U-75412E, U-78518F, and U-83836E[381–384]). None of these compounds produced statis-tically significant therapeutic benefits for AD patients.

MitoQ (redox mixture of mitoquinone and mito-quinol; Antipodean Pharmaceuticals, Menlo Park

CA, under license from the University of Otago;Fig. 6 shows the oxidized form) is a brain penetrat-ing mitochondria-targeting antioxidant [385–388]. Itselectively blocked mitochondrial oxidative damageand prevented cell death [389–394]. It is evaluated inPhase II clinical trials for the treatment of PD and liverdamage. Clinical trials for the treatment of Friedreich’sataxia and sunburn were abandoned (Thomson ReutersPharma, update of June 18, 2012).

A potential follow-up compound is MitoVitE(Fig. 6) [395, 396]. The development of MitoPBN(Antipodean Pharmaceuticals, Menlo Park CA)(Fig. 6) consisting of the spin trapping agent �-phenyl-N-tert.-butyl-nitrone [397, 398] conjugated totriphenyl-phosphonium bromide was terminated [399](Thomson Reuters Pharma, update of June 18, 2012).

MTP-131 (Bendavia, SS-31, Szeto-Schiller pep-tide; Stealth Peptides Inc., Newton Centre MA) (Fig. 7)is a cell-permeable mitochondria-targeting antioxidanttetrapeptide (NH2-D-Arg-Dmt-Lys-Phe-NH2), whichis in Phase II clinical trials for ischemia reperfusioninjury since September. 2010 (EMBRACE–STEMItrial). It will further be investigated for the treatmentof eye, neurological, mitochondrial, and metabolic

Page 16: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

16 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 7. The Szeto-Schiller peptide, a manganoporphyrin antioxidant and CNB-001.

diseases [400–402],[403] (Thomson Reuters Pharma,update of June 15, 2012).

VP-20629 (OX1; IN-OX1; OX1; OXIGON, Indole-3-propionic acid; ViroPharma, Exton PA, under licensefrom Intellect Neurosciences, New York Universityand Mindset BioPharmaceuticals) is an antioxidant andA� aggregation/deposition inhibitor. The prevention ofA� induced neurotoxicity by indole-3-propionic acidwas described [404]. The rationale behind the selectionof OXIGON as a potential disease-modifying therapyfor Alzheimer’s disease was presented [1706]. Pre-liminary data from a multiple-dose Phase Ib trial forAD were reported in October 2010. It appears thatViroPharma wants to pursue the drug for the indicationFriedreich’s ataxia only (Thomson Reuters Pharma,update of August 17, 2012).

There are several antioxidants in preclinical evalua-tion (in alphabetical order):

Catalytic manganoporphyrine antioxidants, suchas AEOL-10113, AEOL-10150, AEOL-10201 andAEOL-11207 (Aeolus Pharmaceuticals, MissionViejo CA; Fig. 7) may be promising for the treatment of

PD and ALS [405–407] and ischemic stroke [408–412](Thomson Reuters Pharma, update of April 23, 2012).

CNB-001 (The Salk Institute for Biological Studies)(Fig. 7) is a curcumin derivative for potential treatmentof traumatic brain injury and stroke (Thomson ReutersPharma, update of November 9, 2011).

DL-3-n-butylphtalide (NBP, Hebei, China) is a nat-ural antioxidant extracted from seeds of Apium anda powerful free radical scavenger [413]. It protecteddopamine neurons in a rotenone model for PD [414].

FRP-0924 (gemifloxacin, Neuron BioPharma,Granada, Spain) is an antioxidant and neuroprotectantfor the potential treatment of neurodegenerative dis-eases including AD. FRP-924 crossed the blood-brainbarrier and prevented neuronal death. As fluoro-quinolones are generally well tolerated with most sideeffects being mild and serious adverse effects beingrare, it is expected that the drug will enter Phase IIrapidly (Thomson Reuters Pharma, update of August14, 2012).

IAC (Cerebricon, Finland and Medestea Research,Torino, Italy) (Fig. 8) is a novel radical scavenger react-

Page 17: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 17

Fig. 8. Antioxidants in preclinical evaluation.

ing with most carbon-, nitrogen-, and oxygen-centeredradicals of biological interest [415–418]. Daily treat-ment with IAC (3–30 mg/kg i.p.) decreased mortality,enhanced cognitive functions in the water maze, andreduced the A� plaque burden in hA�PP transgenicmice [419].

Lipid soluble antioxidants (OXIS International,Beverley Hills CA) mimic the activity of the body’snatural cell membrane-protecting antioxidant vitaminE for the potential treatment of cardiovascular diseases,diabetes, AD, and PD (Thomson Reuters Pharma,update of June 12, 2012). The structures were notcommunicated.

Lipocrine and Memoquin, dual acetylcholines-terase inhibitors and antioxidants (University ofBologna), were described in Part 2, Chapter 2.1.1.3.(formulae in Part 2 in Fig. 6).

NPS-0155 (Neuron BioPharma, Granada, Spain) isan antioxidant and neuroprotective compound for thepotential treatment of AD and other neurodegenerativediseases (Thomson Reuters Pharma, update of April27, 2012). The structure was not communicated.

PAN-811 (3-AP; NSC-663249; OCX-191; Triapine,Panacea Pharmaceuticals, Gaithersburg MD) (Fig. 8) isa ribonucleotide reductase inhibitor, calcium ion chela-tor, and radical scavenger for the potential treatmentof AD and ischemia [420, 421] (Thomson ReutersPharma, update of February 25, 2011).

S-52 (Shanghai Institute of Materia Medica) (Fig. 8)is a sinomenine derivative, an active natural product

from the Chinese herb Sinomenum acutum. It is a scav-enger of free radicals. It attenuated the toxicity of A�to energy metabolism, mitochondrial membrane struc-ture, and key enzymes in the electron transport chain[422].

There are numerous preclinical reports of drugsand natural products, which, in addition to their radi-cal scavenging properties, also prevented A�-inducedneurotoxicity, such as peoniflorin [423], 2,2′-pyridoin[424], quetiapine [425]), stemazole (Fig. 8) [426], andzeatin (Fig. 8) [427].

Dual free radical scavengers and A� binding lig-ands were described [428, 429].

The development of many antioxidants was ter-minated (in alphabetical order) of AN-808 (AthenaNeurosciences, now Elan Pharmaceuticals), AO-2 andAO-3 antioxidants (Antoxis), CNSB-002 (AM-36;Relevare; AMRAD, formerly Zenyth Therapeutics[430] [431, 432]), CPI-1189 (Centaur [433, 434]), dis-ufenton sodium (Cerovive, NXY-059, AstraZeneca,which was in Phase III clinical trials for the treatmentof acute ischemic stroke [435–445]), FR-210575 (Fuji-sawa [446]), MDL-101002 (Hoechst Marion Roussel,now sanofi [447, 448]), and raxofelast (IRFI-016,Biomedica Foscama Industria [449–452]).

METAL CHELATORS

The metallobiology of AD was investigated in greatdetail over the last fifteen years. Excellent reviews were

Page 18: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

18 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

published (in chronological order) 2001: [453], 2002:[454], 2003: [455], 2006: [456], 2007: [457–459],2008: [460–462], 2009: [463–467], 2010: [468–472],2011: [473–478], 2012: [479–485], 2012: [1707].

DP-b99 (D-Pharm, Rehovot, Israel) (Fig. 9) is ani.v. prodrug of the calcium and zinc chelator BAPTAfor the potential treatment of stroke and traumaticbrain injury. It is in Phase III clinical trials for strokesince October 2009. First reports on clinical resultswere communicated [486–488]. Also preclincial datawere disclosed [489–491] (Thomson Reuters Pharma,update of February 3, 2012).

PBT-2 (Prana Biotechnology, Parkville, Australia)(Fig. 9) is an oral zinc ionophore metal-protein attenu-ating compound that reduced levels of soluble A� andtau hyperphosphorylation [492]. A Phase IIa study inAD patients (n = 78), started in December 2006 withdaily doses of 50 or 250 mg or placebo over threemonths, was completed in January 2008. A signifi-cant reduction of A�42 levels in the cerebrospinal fluid(CSF) was seen in the 250 mg group. This dose alsosignificantly improved executive function performancein the category fluency and the trail making cogni-tive tests compared to placebo [493, 494]. In January2012, the FDA approved an IND to initiate a random-ized, multicenter, double-blind, placebo-controlled,parallel-group, safety, tolerability, and efficacy PhaseIIa trial in early- to mid-stage HD patients (n = 100) inthe US and Australia. In April 2012, the first patientwas dosed in this trial. By June 2012 the trial hadbeen initiated in Australia. Data are expected in thesecond half of 2013. The compound was extensivelycharacterized in preclinical studies [495–497] (Thom-son Reuters Pharma, update of October 2, 2012). Seealso Chapter 17.1. Small molecules preventing tauaggregation.

AEN-100 (Synthetic Biologics, formerly AdeonaPharmaceuticals, Ann Arbor, MI) is a once-daily, gas-troretentive, sustained-release, oral tablet formulationof zinc acetate for the potential treatment of ALS andAD. By November 2011, a Phase I study for ALSpatients was underway (Thomson Reuters Pharma,update of September 19, 2012).

Other metal chelators are currently in preclinicalevaluation (in alphabetical order):

Aom-0937 (Hangzhou Adamerck Pharmlabs,China) is a prodrug of DP-b99 (Thomson ReutersPharma, update of July 28, 2011). The structure wasnot communicated.

DP-460 (D-Pharm, Rehovot Israel) (Fig. 9) is amembrane active chelator derivative of the calcium-specific chelator BAPTA that modulates copper and

zinc homeostasis to inhibit oxygen radicals and plaqueformation. It is in preclinical evaluation for the poten-tial oral treatment of AD and ALS (Thomson ReutersPharma, update of November 1, 2011). It appears thatDP-460 was preferred to DP-109 [498]. Both com-pounds have been evaluated as neuroprotectants ina transgenic mouse model of ALS [499] (ThomsonReuters Pharma, update of August 7, 2012).

Deferoxamine (SAN-121; Sanomune, a subsidiaryof DiaMedica under license from HealthPartnersResearch Foundation, Winnipeg, Canada) (Fig. 10) is anasally-delivered iron chelator improving performancein a radial arm water maze in P3301L tau trans-genic mice [500] (Thomson Reuters Pharma, updateof December 23, 2011).

HLA20A (Technion Haifa) (Fig. 10) is a com-bination of an 8-hydroxy-quinoline metal chelator,which was carbamoylated at the 8-hydroxy functionto interact with acetylcholinesterase as in rivastigmine[501, 502]. See Part 2, Chapter 2.1.1.10. Dual acetyl-cholinesterase inhibitors and metal chelators.

PA-1637 (Palumed, France) is an A� plaqueproduction-inhibiting copper chelating agent (Thom-son Reuters Pharma, update of May 28, 2012). Thestructure was not communicated.

PBT-3 and PBT-4 (Prana Biotechnology, Parkville,Australia) are non-8-hydroxyquinoline metal pro-tein attenuating compounds, potential follow-ups ofPBT-2. (For both Thomson Reuters Pharma, updateof November 2, 2011). The structures were notcommunicated.

Triazole-pyridine derivatives as inhibitors ofmetal-induced A� aggregation were described [503].

VAR-10200 (HLA-20; Varinel, West Chester, PA)(Fig. 10) is a dual iron-chelating agent and MAO-Binhibitor for the potential treatment of age-related mac-ular degeneration [501] (Thomson Reuters Pharma,update of February 24, 2012). See Part 2, Chapter 2.26.Drugs interacting with Monoamine Oxidase.

VAR-10300 (M-30; Varinel, West Chester, PA,Technion and the Weizmann Institute) (Fig. 10)combines the iron-chelating properties of 8-hydroxy-quinoline with the MAO inhibitor moiety of rasaglinine[504–517] (Thomson Reuters Pharma, update ofSeptember 24, 2012). See also Part 2, Chapter 2.26.Drugs interacting with Monoamine Oxidase.

The development of many metal chelators wasterminated (in alphabetical order): desferrioxam-ine [518–520], Gero-46 (clioquinol, Gerolymatos);NG-1 and PBT-1 (clioquinol, Prana Biotechnol-ogy [521, 522]); phanquinone (PN Gerolymatos);tetrathiomolybdate (a copper chelating agent of

Page 19: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 19

Fig. 9. Metal chelators.

Fig. 10. Metal chelators II.

Adeona Pharmaceuticals, formerly Pipex under licensefrom the University of Michigan); the iron chelatorVAR-10100 (VK-28) [523–526] and VK-11 and VK-12 (Prana Biotechnology).

NATURAL PRODUCTS

Excellent reviews on naturally occurring phyto-chemicals for the prevention and treatment of AD have

Page 20: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

20 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

been presented [527–532]. An overview on “naturalsubstances and AD: from preclinical studies to evi-dence based medicine” was published recently [533].

Mentat (BR-16A; Himalaya Drug Co.) is a stan-dardized mixture of herbal extracts, which has beenlaunched in India and the US (as MindCare) inNovember 2000 for the treatment of cognitive deficits(Thomson Reuters Pharma, update of November 6,2000).

YY-280 (Yucrid; Yuyu, South Korea) is a combi-nation therapy of ticlopidine and EGb-761 (tanamin,a Ginkgo biloba extract) administered as a tabletfor the treatment of apoplexy and myocardial infarc-tion. The drug was registered in Korea in May 2008[534] (Thomson Reuters Pharma, update of April 11,2012).

SK-PC-B70M (SK Chemicals Life Science, SouthKorea) is derived from the dried root of Pulsatellakoreana (baekduong). A randomized, double-blind,placebo-controlled Phase III clinical trial for the treat-ment of mild-to-moderate AD patients (n = 256) wasinitiated in November 2010 in South Korea. SK-PC-B70M improved scopolamine-induced impairments ofmemory consolidation in rats [535, 536]. It had antiox-idant activity and reduced A� levels in the brains ofTg2576 mice [537, 538]. It alleviated the neurologicsymptoms in G93A-SOD1 ALS mice [538] (ThomsonReuters Pharma, update of February 28, 2012).

Circadin (KI-1001, NSC-56423; Neurim Phar-maceuticals, Tel Aviv) is an oral controlled-releaseformulation of melatonin (i.e., melatonin acetamide)and was launched in 2007 for the treatment of primaryinsomnia. A Phase II clinical trial was initiated in Israelin patients with MCI (n = 50) in October 2007. AnotherPhase II clinical trial was initiated in the US, UK, andIsrael in patients with mild-to-moderate AD (n = 140)treated with an acetylcholinesterase inhibitor to evalu-ate the effects of add-on Circadin (2 mg) on decline incognitive skills, global functioning, and daytime som-nolence in September 2009 (Thomson Reuters Pharma,update of September 26, 2012).

KD-501 (Kwang Dong Pharmaceutical Co., Seoul)is an extract from the Scrophulariae radix. The drug isin Phase II clinical trials for AD since March 2009(Thomson Reuters Pharma, update of February 11,2011).

PPLs (NatureWise Biotech, Taipeh, Taiwan) areprenylflavanone compounds extracted from Taiwanesepropolis in Phase II clinical trials. A pilot AD programof PPLs in combination with Aricept was initiated inNovember 2010 (Thomson Reuters Pharma, update ofJuly 10, 2012).

PTX-200 (Phytrix, Munich, Germany) is a plant-derived neuroprotectant for the potential treatment ofPD. A Phase I/IIa clinical evaluation in the UK wassuccessfully completed (Thomson Reuters Pharma,update of February 1, 2012).

Resveratrol is a natural phyto compound, whichactivated Sirtuin-1 [539–544]. It reduced A� accu-mulation [545–548]. It remodeled soluble oligomersand fibrils of A� into off-pathway conformers [549].Resveratrol is not a direct activator of SIRT1 enzymeactivity [550]. Resveratrol improved memory deficitsin mice fed a high-fat diet [551]. Subchronic oraltoxicity and cardiovascular safety pharmacology stud-ies were carried out [552]. The biosynthesis ofresveratrol in yeast and in mammalian cells wasachieved [553]. The Georgetown University MedicalCenter in Washington DC started at Phase II, ran-domized, double-blind, placebo-controlled study inpatients with mild-to-moderate AD (n = 120) in the USin May 2012 (NCT01504854). A novel application inHD was discussed recently [554]. See also in Part 2,Chapter 2.40, Drugs interacting with Sirtuin, formulain Part 2, Fig. 23 (Thomson Reuters Pharma, update ofJune 22, 2012).

RPh-201 (Regenera Pharma, Rehovot, Israel) is asubcutaneous formulation of an agent derived froma traditional medicinal plant. A Phase I/IIa random-ized, double-blind, placebo-controlled trial in healthyvolunteers and adults with AD (n = 56) started in Jan-uary 2012 in Canada. A topical formulation is currentlyevaluated in a Phase II study in patients with chroniculcerated wounds (n = 15) in Israel (both ThomsonReuters Pharma, update of January 31, 2012).

VR-040 (apomorphine as dry powder inhaledformulation, Vectura, Chippenham, UK) is beingdeveloped in Phase II clinical trials for the potentialtreatment of PD since June 2006 (Thomson ReutersPharma, update of October 3, 2012).

Exebryl-1 (ProteoTech, Kirkland, WA and Chineselicensee Tasly Pharmaceuticals) is a synthetic com-pound with molecular weight of about 300 Da, one ofthe components of an Amazonian vine Uncaria tomen-tosa extract, which reduced aggregation of both A�and tau. A Phase I clinical trial for AD was initiated inJuly 2008 (Thomson Reuters Pharma, update of June8, 2012).

Taisi (Beijing SL Pharmaceutical under license fromXuanwu Hospital of Capital Medical University), acapsule formulation of a stilbene analogue isolatedfrom an unspecified organism, is in clinical studies inChina since February 2011 (Thomson Reuters Pharma,update of June 29, 2012).

Page 21: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 21

There are many natural products in preclinical eval-uation as potential drugs for the treatment of AD (inalphabetical order):

Alpha-mangostin, a polyphenolic xanthone deriva-tive from mangosteen, concentration-dependentlyattenuated neurotoxicity induced by A�1-40 or A�1-42oligomers (EC50 = 3.89 nM and 4.14 nM, respectively)[555].

Anatabine (Star Scientific, Glen Allen VI throughits subsidiary Rock Creek Pharmaceuticals in col-laboration with the Roskamp Institute) reduced A�when applied to cells [556] (Thomson Reuters Pharma,update of January 3, 2012).

Andrographis paniculata leaves extract showedcerebroprotective and nootropic activities in rats [557].

Apomorphine is an inhibitor of A� fibril formation[558]. Apomorphine treatment of AD mice promotedA� degradation [559]. Roles of apomorphine for reg-ulated �-cleavage, autophagy, and antioxidation werediscussed [560].

AX-00111 (Axonal Consultoria Tecnologica Ltda,Sao Paulo Brazil) is a plant-derived compound for thepotential treatment of AD (Thomson Reuters Pharma,update of June 1, 2012).

Axona (Accera Inc., Broomfield, CO) is a newmedical food therapy for AD patients [561, 562].It contains the proprietary formulation of medium-chain triglycerides, mostly caprylic triglyceride. Therationale for the use of Axona is based on the find-ing that the cerebral hypometabolism (i.e., impairedglucose metabolism) is an early sign of AD. Medium-chain triglycerides are metabolized in the liverresulting in the production of the ketone body beta-hydroxybutyrate, which is transported to the brainto provide an alternative fuel source for cerebralmetabolism [563, 564]. See also [565].

Bacopa monnieri has shown memory free recallenhancing effects in adult humans [566]. Neuroprotec-tive effects in experimental models of dementia weredescribed [567].

Baicalein (5,6,7-trihydroxyflavone) protected corti-cal neurons from A�25-35-induced toxicity [568] andin a one dose pre-treatment at 5 and 10 mg/kg i.p.attenuated A�25-35-induced amnesia in mice in a step-through passive avoidance paradigm. Post-treatmentfor 7 or 13 days (10–15 mg/kg i.p.) also attenuatedA�25-35-induced amnesia [569]. Baicalin preventedthe production of hydrogen peroxide and oxidativestress induced by A� aggregation in SH-SY5Y cells[570]. Apigenin (4′,5,7-trihydroxyflavone), baicalein,and nordihydroguaiaretic acid were potent inhibitorsof liposome permeabilization by A�42 oligomers

[571]. Baicalein inhibited the formation of �-synucleinoligomers within living cells and prevented A� peptidefibrillization and oligomerization [572].

Beta-asarone improved cognitive functions in ratsafter injection of A� into the hippocampus [573] prob-ably via JNK signaling and modulation of bcl-2 familyproteins [574] and attenuation of neuronal apoptosis[575, 1708].

BT-11 is an extract of the dried root of Poly-gala tenuifolia (Willdenow) and effectively enhancedcognitive functions in elderly humans [576–578].

BV-7003 (Bioved Pharmaceuticals) is a naturalproduct for the potential treatment of memory loss(Thomson Reuters Pharma, update of September 11,2012). The structure was not communicated.

Cabernet Sauvignon attenuated A� neuropathol-ogy in a mouse model of AD [579].

Carvacrol showed cognition enhancing activity intwo rat models of dementia [580].

Catechins showed potent anti-amyloidogenic andfibril-destabilizing effects in vitro [581–583].

Celastrol, a triterpenoid antioxidant compound iso-lated from the Chinese Thunder of God vine (T.wilfordii) reduced A� pathology in a transgenic mousemodel of AD [584].

Celastrus paniculatus seeds showed nootropicactivity [585].

Chelerythrine showed promising cholinesteraseinhibition, good inhibition of amyloid-� aggregationand the ability to disaggregate preformed amyloid-�aggregates [1709].

Cinnamon extract reduced A� oligomerization andcorrected cognitive impairment in animal models ofAD [586].

Coumarins are naturally occurring �-secretaseinhibitors [587] and acetylcholinesterase inhibitors[588].

Cryptotanshinone (CTS), an active componentof the medicinal herb Salvia miltiorrhiza, inhibitedA� aggregation and protected SH-SY5Y cells fromdamage by A� [589]. It upregulated �-secretase byactivation PI3K pathway in cortical neurons [590].

Curcumin showed potent anti-amyloidogeniceffects for AD A� fibrils in vitro and in vivo [591–605].Clinical trials in AD patients showed disappointingresults [350, 600, 606]. A novel nanoparticle for-mulation of curcumin (NanoCurc) was developed atthe Indiana University School of Medicine [607].For curcumin-decorated nanoliposomes see [608].Structure-activity relationships of A� aggregationinhibitors based on the curcumin scaffold were pre-sented [609–611].

Page 22: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

22 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

DL-3-n-butylphtalide (NBP Pharmaceuticals,Hebei China) is a natural antioxidant extracted fromseeds of Apium and a powerful free radical scavenger[413]. It protected dopamine neurons in a rotenonemodel for PD [414].

DX-9386 is a traditional Chinese medicinalprescription, which improved thymectomy-inducedimpairment of learning behaviors in mice [612].

Ecdysterones, steroidal hormones in insects andterrestrial plants, inhibited A� aggregation, disaggre-gated preformed fibrils, and inhibited A�42-inducedcytotoxicity [613].

(-)-Epigallocatechin-3-gallate (EGCG; Sunphe-non) present in green tea reduced A�-mediatedcognitive impairment presumably via flavonoid-mediated presenilin-1 phosphorylation, whichreduced A� production [614–616]. EGCG preventedlipopolysaccharide-induced elevation of A� gener-ation [617]. EGCG remodeled mature �-synucleinand A� fibrils and reduced cellular toxicity [618].The cell signaling pathways and iron chelation weredescribed [582, 619–624]. Also ERK and NF- κBpathways are involved [617]. The structural propertiesof EGCG-induced, nontoxic AD A� oligomers weredescribed [625]. EGCG functions through estrogenreceptor mediated activation of ADAM10 in thepromotion of non-amyloidogenic processing of A�PP[626, 627]). A special formulation of EGCG innanolipidic particles to improve its bioavailability waspresented [628]. See also Part 1, Chapter 1.9. Drugsinteracting with estrogen receptors and Part 1, Fig. 10.

ESP-102, a standardized combined extract ofAngelica gigas, Saururus chinensis, and Schizandrachinenis, significantly improved scopolamine-inducedmemory impairment [629] and A�1-42-induced mem-ory impairment in mice [630].

An aqueous extract of Eucommia ulmoidesOliv. Bark (EUE) showed beneficial effects on learn-ing and memory impairments in mice [631].

Flavonoids in AD and neuroinflammation werediscussed in depth [621, 632–635]. Some act as acetyl-cholinesterase inhibitors [636]. Biflavanoids weresuperior to monoflavonoids in inhibiting A� toxicity[637].

Fortasyn Connect is a multi-nutrient diet com-prising docosahexanoic acid (DHA), eicosapentenoicacid, uridine-mono-phosphate, choline, phospholipids,folic acid, vitamins B6, B12, C, and E and sele-nium. It reduced AD-like pathology in young adultA�PPSWE/PS1dE9 mice [638].

Fulvic acid inhibited aggregation and promoted dis-assembly of tau fibrils associated with AD [639].

Gallic acid from grape seed polyphenol extract maybe useful for the treatment of AD [583].

Garlic extract attenuated the cytotoxicity of A� onundifferentiated PC12 cells [640] and suppressed lipidperoxidation induced by A� in PC12 cells [641]. Itprotected against A�-induced apoptosis [642]. Garlicextract inhibited A� fibril formation and defibrillatedA� preformed fibrils [643]. Aged garlic extract ame-liorated the early cognitive deficits in Tg2576 mice[644, 645]. A review on the “aged garlic extract” waspublished [646].

Gastrodin protected primary cultured rat hip-pocampal neurons against amyloid-� peptide-inducedneurotoxicity via ERK1/2-Nrf2 pathway [1710].

Ginger root extracts (Zingiber officinale, Cog-nition Therapeutics) effected inhibition of A�42aggregation (Thomson Reuters Pharma, update of May23, 2012).

Gingko biloba (EGb 761, Tanakan, Tanamin) treat-ment prevented age-related spatial memory deficits ina transgenic mouse model of AD [647]. It enhancedadult hippocampal neurogenesis and phosphorylationof CREB [648]. For reviews on Ginkgo biloba extractand CNS functions, see [649, 650]. For results of clin-ical studies, see (in chronological order) [651–665].

Grape-derived polyphenolics from Vitis viniferagrape seeds attenuated cognitive deterioration in amouse model of AD [666]. Ultrastructural alterationsof AD paired helical filaments by grape seed-derivedpolyphenols was studied [667].

Guanosine protected human neuroblastoma cellsfrom oxidative stress and toxicity induced by A� pep-tide oligomers [668].

Hederacolchidide-E from Pulsatilla koreanashowed cognition-enhancing and neuroprotectiveeffects by reversing scopolamine-induced cognitiveimpairments in rats. It increased viability of humanneuroblastoma SK-N-SH cells incubated with A�42[536].

Heme prevented A�1-40 aggregation and its cyto-toxicity [669].

Hopeahainol A attenuated memory deficits by tar-geting amyloid-� in APP/PS1 transgenic mice [1711].

HSH-971 (Ocean University of China, Shandong)is a marine sulfated oligosaccharide for the potentialtreatment of AD [670]. (Thomson Reuters Pharma,update of November 7, 2011).

HX-106 (HX-106N; ViroMed Co., Seoul) is anatural product inhibiting acetyl-cholinesterase forthe potential treatment of AD (Thomson ReutersPharma, update of June 13, 2012). The structure wasnot disclosed.

Page 23: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 23

Hyperoside protected primary rat cortical neu-rons from neurotoxicity induced by A� via thePI3K/Akt/Bad/Bcl(XL)-regulated mitochondrialapoptotic pathway [671].

IB-10C179(Instituto Biomar, Leon, Spain) is acompound derived from marine organisms which pro-tected primary neurons from apoptosis and reducedfree radical production (Thomson Reuters Pharma,update of March 7, 2012). The structure was notcommunicated.

Icariin is a flavonoid isolated from Epimediiherba. It inhibited A�25-35 induced expression of�-secretase in rat hippocampus [672]. It attenuatedA�-induced neurotoxicity by inhibition of tau pro-tein hyperphosphorylation in PC12 cells [673]. Itattenuated lipopolysaccharide-induced microglial acti-vation and resultant death of neurons by inhibitingTAK1/IKK/NF-κB and JNK/p38 MAPK pathways[674]. It improved memory impairment in AD modelmice and attenuated A�-induced neurite atrophy [675].There seems to be a synergistic effect to improve learn-ing and memory deficits in rats by co-administrationof Icariin and Panax notoginseng saponins [676].

IDN-5706, a hyperforin derivative, decreased thecontent of acetylcholinesterase associated with dif-ferent types of A� plaques in 7-month-old doubleA�PPSWE/PS1 transgenic mice after treatment withIDN 5706 for 10 weeks [677].

Kaempferol protected PC12 and T47D cells fromA� toxicity [581, 678, 679].

Loganin isolated from Cornus officinalis showedcognitive-enhancing activity in scopolamine-inducedamnesic mice [680].

Luteolin exerted ameliorating effects on A�-induced impairment of water maze performance andpassive avoidance in rats [681–684].

Medical Food Cocktail consisting of the dietarysupplements curcumin, piperine, epigallocatechin gal-late, �-lipoic acid, N-acetylcysteine, B vitamins,vitamin C, and folate administered for 6 months toTg2576 mice resulted in improvement of cognitivefunctioning [685].

Melatonin inhibited AD �-fibrillogenesis[686–689]. The neuro-protective activities ofmelatonin against the A� are not mediated by mela-tonin membrane receptors. The role of melatonin inAD-like neurodegeneration was described [690–693].Melatonin facilitated short-term memory [694].

Menthol exerted a protective effect on A�-inducedcognitive deficits in mice [695].

Morin destabilized A�42 protofibrils [696]. It inhib-ited the early stages of A� aggregation [697]. It

attenuated tau hyperphosphorylation by inhibitingGSK-3� [698].

Myrcetin is a naturally occurring regulator of metal-induced A� aggregation and neurotoxicity [699, 700].

Naringin showed memory enhancing activity inmice [701].

NeurocentRX Pharma is investigating a naturalproduct-based compound for the treatment of cognitivediseases (Thomson Reuters Pharma, update of Febru-ary 17, 2012).

Nordihydroguaiaretic acid inhibited growth aris-ing from direct A� protofibril association [599, 702,703].

O4 (an orcein-related small molecule) was able toconvert toxic A� oligomers to nontoxic �-sheet-richamyloid fibrils [704].

Obovatol, a biphenolic compound isolated fromMagnolia obovata, attenuated scopolamine-inducedcognitive dysfunctions [705], improved cognitivefunctions in animal models of AD [706], andattenuated LPS-induced memory impairments in micevia inhibition of NF-κB signaling pathway [707, 708].

Oleuropein and derivatives from olives were recog-nized as tau aggregation inhibitors [709].

Oren-gedoku-to exerts its potential use for thetreatment of AD as a weak, reversible inhibitor ofindoleamine-2,3-dioxygenase [710].

Oroxylin A is a flavonoid compound that is found inthe root of Scutellaria baicalensis Georgi. It attenuatedthe memory impairment induced by transient bilateralcommon carotid artery occlusion in mice [711].

1,2,3,4,6-penta-O-galloyl-beta-D-glucopyranoseis the active constituent of the traditional medicinalherb Paeonia suffruticosa. It showed potent A�anti-aggregation effects in vitro and in vivo [712].

Piceatannol showed protective effects against A�-induced neuronal cell death [713].

Pinocembrin, a flavonoid abundant in propolis, pro-tected against A�-induced toxicity in neurons [714].

Polyphenol derivatives (Pharma Eight, Nagoya)probably interacted with A� through � – � stackinginteractions via the aromatic amino acids of A� [715].For a review, see [716] (Thomson Reuters Pharma,update of January 13, 2011).

Procyanidins extracted from the lotus seedpodreversed memory impairment in cognitively impairedaged rats associated with decreased hippocampalCREB phosphorylation [717, 718].

Prosopis cineraria (L.) Druce (Leguminosae), aplant of the Thar Desert of India and Pakistan, is usedtraditionally by local people for the treatment of mem-ory disorders and to arrest wandering of the mind.

Page 24: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

24 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

The extract exerted significant nootropic activity in theMorris water maze test, which may be attributed to theinhibition of brain acetylcholinesterase [719].

Puerarin, a phytoestrogen isolated from Puerarialobata, attenuated A�-induced cognitive impairment[720–723, 1712].

Quercetin-O-glucuronide significantly reduced thegeneration of amyloid-� peptide [1713].

Pycnogenol protected neurons from A�-inducedapoptosis [724].

Quercetin showed protective effects against A�42in primary neurons [586, 725]).

Rosmarinic acid protected PC12 cells fromA�-induced neurotoxicity [599, 726]. Subchronicadministration of rosmarinic acid, a natural prolyloligopeptidase inhibitor, enhanced cognitive perfor-mance [727].

Rutin inhibited A� aggregation and cytotoxicity,attenuated oxidative stress, and decreased the produc-tion of nitric oxide and proinflammatory cytokines[728].

Saffron from Crocus sativus had an inhibitory effecton A� aggregation [729, 730]. It was administeredto 46 patients with probable AD in a 16-week dou-ble blind study (15 mg twice a day). Saffron was safeand effective [731]. A 22 week study corroborated thefirst results [732]. Saffron may be a source of novelacetylcholinesterase inhibitors [733].

Salidroside showed neuroprotective effects againstA�-induced oxidative stress in SH-SY5Y human neu-roblastoma cells [734].

S-allyl-L-cysteine, the main constituent of garlic,protected against A�-induced apoptosis and attenuatedcaspase-3 activation, DNA fragmentation and PARPcleavage [648]. It exerted protective effects on A�-induced cell death in NGF-differentiated PC12 cells[735] and protected against A�-induced neurotoxic-ity in organotypic hippocampal cultures [736]. For areview on garlic extract and one of its active ingredi-ents, S-allyl-L-cysteine, see [646].

Silibinin, a flavonoid derived from Silybum mar-ianum, prevented memory impairment induced byA�25-35 in the Y-maze and novel object recognitiontests in mice [737].

Sinapic acid is a phenylpropanoid compound withanti-inflammatory and neuroprotective effects in amouse model of A�1-42 protein-induced AD [738].

Souvenaid (Nutricia, Chatel-St-Denis, Switzer-land) is a mix of nutrients including the omega-3fatty acid docosahexanoic acid, uridine monophos-phate, and choline, dietary precursors for the synthesisof phospholipids. Two clinical trials, Souvenir I, which

lasted 12 weeks, and Souvenir II, which lasted 24weeks, were concluded [739–741].

Substance P, the tachykinin undecapeptide, pro-tected cerebellar granule cells against A�-inducedapoptosis [742].

Syringin from the dried stem barks of Fraxinusrhynchophylla protected against A�-induced toxicityin neuronal cells [743].

Tannic acid destabilized A� fibrils in vitro [744,745]. It is a natural �-secretase inhibitor [746].

�-Tocopherol quinone inhibited A� aggregationand cytotoxicity, disaggregated preformed fibrils anddecreased the production of ROS, nitric oxide, andinflammatory cytokines [747].

Thymol showed cognitive-enhancing activity in tworat models of dementia [580].

Total coptis alkaloids produced a protective effecton A�25-35-induced learning and memory dysfunctionin rats [748].

Ursolic acid attenuated A�-induced apoptosis in adose dependent manner [749].

Vitamin A has anti-oligomerization effects on A�in vitro [750].

Vitamin B12 deficieny is associated with cognitiveimpairment [1714]. Vitamin B12 supplements admin-istered orally or parenterally at high dose (1 mg daily)were effective in correcting biochemical defiency, butimproved cognition only in patients with pre-existingvitamin B12 deficiency [1715].

Vitamin D defiency is associated with increasedodds of cognitive impairment [1716–1718].

Withania somnifera (also known as Ashwagandha,an Indian ginseng) is a nootropic agent promotingcognition including memory [751]). W. somniferaadministered once daily over 30 days reversed behav-ioral deficits, plaque pathology, accumulation of A�peptides and oligomers in the brains of middle-agedand old A�PP/PS1 AD mice. Enhanced expression oflow-density lipoprotein receptor-related protein (LRP)in brain microvessels and the A� degrading proteaseneprilysin occurred 14–21 days after a substantialdecrease in brain A� levels [752].

Wuzi Yanzong Granule improved memory func-tions in patients with MCI [753].

Yokukansan (TJ-54), a traditional Japanese herbalmedicine, was tested in clinics for potential anti-dementia effects [754]. Spatial memory in a rat modelof early AD was improved [755].

Zokumei-to (ZMT) attenuated A�25-35-inducedmemory impairment [756].

The Central Drug Research Institute (India) wasdeveloping bacosides A and B, which are saponin

Page 25: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 25

nootropic agents. The compounds were in Phase IIclinical evaluation, but it appears that the developmentwas terminated. Also the development of CBNU-06 (Chungbuk University), DCB-AD1 (DevelopmentCenter for Biotechnology), dysiherbaine (DaiichiSuntory Biomedical Research Co., a compound iso-lated from the Micronesian sponge Dysidea herbacea[757]), of the nordihydroguaiaretic acid derivativeEM-4232 (Eromos Pharmaceutical LLC), HSS-808, HSS-818, HSS-838, HSS-848 and HSS-888(HerbalScienceLLC Singapore; standardized turmericcurcuma longa extracts), of LHM-123 (Mazal PlantPharmaceuticals), MDF-004 and MDF-005 (NeuronBioPharma), and of YY-1224 and YY-1824 (Yuyu;orally available terpene trilactones extracted fromGinkgo biloba [758]) was terminated.

NOOTROPICS (“DRUGS WITHOUTMECHANISM”)

In this chapter compounds are described, whosemechanism(s) of action are unknown. Initiation of clin-ical trials was based on positive effects on impairedbrain functions in experimental animals after proof ofgood tolerability.

LSL-001 (Laboratoire S. Lasnier, Etaules, France)is a nootropic compound in Phase II clinical trials sinceJune 2011 for the potential treatment of AD (ThomsonReuters Pharma, update of June 5, 2012). The structurewas not disclosed.

N-251 (Neurokos Inc., Palo Alto, CA) is a com-pound of an undisclosed mechanism of action forthe potential treatment of AD. Phase II clinical trialsstarted in May 2009 (Thomson Reuters Pharma, updateof January 16, 2012). The structure of N-251 was notcommunicated.

PF-03049423 (Pfizer) is a neurorestorative com-pound for the potential treatment of neurologicaldisorders including stroke recovery. In December2010, a randomized, double-blind, placebo-controlledPhase II trial was initiated in the US and in South Korea(n = 240) (Thomson Reuters Pharma, update of August13, 2012). The structure was not communicated.

TPM-189 (Teikoku Pharma USA, San Jose, CA) isa transdermal formulation of a small molecule ther-apeutic for the potential treatment of AD in PhaseII development (Thomson Reuters Pharma, update ofMay 17, 2012). The structure was not communicated.

VI-1121 (VIVUS Inc., Mountain View, CA) is anootropic agent in Phase II clinical trials (n = 50) sinceAugust 2011 in the US (NCT01428362) (Thomson

Reuters Pharma, update of September 9, 2011). Thestructure of VI-1121 was not communicated.

ASP-0777 (Astellas Pharma) is in Phase I clini-cal trials since May 2009 (Thomson Reuters Pharma,update of September 19, 2012). The structure was notdisclosed.

Lilly is developing a small molecule for the treat-ment of cognitive impairment in schizophrenia inPhase I clinical trials since July 2012. (ThomsonReuters Pharma, update of November 13, 2012). Thestructure was not disclosed.

RO-5508887 (Roche) is a nootropic agent inPhase I clinical trials in healthy male volunteers inFrance since October 2011 (Thomson Reuters Pharma,update of August 16, 2012). The structure was notdisclosed.

SEP-363856 (Sunovion Pharmaceuticals, Marlbor-ough MA and Sumitomo Chemical) is an orally activecompound with novel mechanism of action for thetreatment of schizophrenia. The drug is in Phase I in theUS. (Thomson Reuters Pharma, update of November5, 2012). The structure was not disclosed.

TAK-357 (Takeda) is a cognitive enhancer withunspecified drug target in Phase I clinical development(Thomson Reuters Pharma, update of July 30, 2012).The structure was not communicated.

There are currently many nootropic agents in pre-clinical evaluation (in alphabetical order):

AC-0523 (Neuera, a subsidiary of Accera, Broom-field, CO) (Fig. 11) is indicated for the treatmentof mitochondrial dysfunction-related HD (ThomsonReuters Pharma, update of February 15, 2012).

AFX-929 (Afecta Pharmaceuticals; Irvine, CA) is anootropic agent in preclinical development (ThomsonReuters Pharma, update of November 10, 2011). Thestructure was not disclosed.

Alzheimer’s disease therapeutics (Berg Pharma,Nashville TN) are nootropic compounds for the poten-tial treatment of AD (Thomson Reuters Pharma,update of April 17, 2012). The structures were notdisclosed.

Alzheimer’s disease therapeutics (SienaBiotech/Roche) are neuroprotectant compounds for the poten-tial treatment of AD (Thomson Reuters Pharma,update of February 8, 2012). The structures were notdisclosed.

Alzheimer’s disease therapy (Reyon Pharmaceu-ticals, Seoul) is a therapeutic program for the potentialtreatment of AD (Thomson Reuters Pharma, update ofJune 20, 2012). The structures were not disclosed.

AVN-457, AVN-458, and AVN-492 (Avineuro Phar-maceuticals, San Francisco, CA) are nootropic agents

Page 26: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

26 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 11. Nootropics and one peptide.

in preclinical development (All three drugs in Thom-son Reuters Pharma, update of August 19, 2011). Thestructures were not disclosed.

CPC-001 (Chase Pharmaceuticals, Washington,DC) is a compound for the palliative treatment of AD(Thomson Reuters Pharma, update of July 12, 2012).The structure was not communicated.

CWF-0804 (JW Pharmaceutical Corp., ChoongwaeHoldings, Seoul) is a nootropic agent in develop-ment for the Korean and Japanese markets (ThomsonReuters Pharma, update of May 11, 2012). The struc-ture was not disclosed.

D-130 (Envoy Therapeutics, Jupiter, FL) is a com-pound targeting the cortex region of the brain usingbacTRAP technology for the potential treatment ofcognition deficits (Thomson Reuters Pharma, updateof June 1, 2012). The structure was not disclosed.

D-180 (Envoy Therapeutics, Jupiter, FL) is a com-pound targeting the striatum region of the brain usingbacTRAP technology for the potential oral treatmentof PD (Thomson Reuters Pharma, update of September12, 2012). The structure was not disclosed.

GSK-2647544 (GSK) is a neuroprotectant for thepotential treatment of Alzheimer’s disease. In October2012, a single-blind, randomized, placebo-controlled,Phase I trial was expected to begin later that month inAustralia in healthy male subjects (expected n = 16). Atthat time, the trial was expected to complete in April

2013 (Thomson Reuters Pharma, update of November20, 2012). The structure was not disclosed.

J-147 (Salk Institute for Biological Studies)(Fig. 11) is a drug that improved memory in normalrodents and prevented cognitive decline in transgenicmice of AD (Thomson Reuters Pharma, update ofDecember 22, 2011).

JAY 2-22-33 (Medical College of Georgia) (Fig. 11)significantly reduced A� toxicity and improved cogni-tive performances in transgenic AD mice [759].

JWB1-84-1 (Medical College of Georgia) (Fig. 11)is a tertiary amine analogue of choline from the labora-tory of the late Prof. Jerry J. Buccafusco. It produced adose-dependent decrease in the number of errors madeby well-trained AD-transgenic mice in the radial armwater maze test [759, 760].

KD-901 (Kwang Dong Pharmaceutical Co., Seoul)is a nootropic drug in preclinical evaluation (Thom-son Reuters Pharma, update of March 1, 2011). Thestructure was not disclosed.

KU-046 (Kareus Therapeutics and Connexios LifeSciences, Atlanta, GA) is a combination drug, whichdemonstrated significant improvement of cognition(Thomson Reuters Pharma, update of May 29, 2012).The structure was not disclosed.

LNK-3186 and LNK-3248 (AstraZeneca after itsacquisition of Link Medicine Corp., Waltham, MA)are nootropic compounds (For both Thomson Reuters

Page 27: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 27

Pharma, update of July 13, 2012). The structures werenot disclosed.

Maltoyl p-coumarate attenuated cognitive deficitsin rats treated with scopolamine or with A�42 [761].

MeN061016-1 (Lijun International Pharmaceuti-cal, Hong Kong) is a small molecule neuroprotectant(Thomson Reuters Pharma, update of June 26, 2012).The structure was not disclosed.

MPP-26 (M et P Pharma, formerly Mattern Pharma-ceuticals, Emmetten, Switzerland) is an intranasal gelformulation of pregnenolone for the potential enhance-ment of memory (Thomson Reuters Pharma, update ofJanuary 27, 2012).

NNZ-2591 (Neuren Pharmaceuticals, Auckland,NZ) (Fig. 11) is an orally active neuroprotectant dike-topiperazine derivative for the treatment of brain injuryand PD [762] (Thomson Reuters Pharma, update ofAugust 31, 2012).

NXD-9062 (Nymox Pharmaceutical Corp., Quebec)is a neuroprotectant for the potential treatment of AD(Thomson Reuters Pharma, update of February 22,2012). The structure was not disclosed.

NXT-182 (Inception Sciences Inc., San Diego) isa novel small molecule neuroprotectant for the poten-tial treatment of CNS disorders, including Alzheimer’sdisease, Parkinson’s disease and age-related cognitivedecline (Thomson Reuters Pharma, update of October29, 2012). The structure was not disclosed.

OG-635 (Oryzon Genomics, Barcelona) is anootropic agent for the potential treatment of AD andPD (Thomson Reuters Pharma, update of May 31,2012). The structure was not disclosed.

Pentylenetetrazole (Balance Therapeutics, Hills-borough, CA) is investigated for the potential treatmentof cognitive impairment in Down’s syndrome (Thom-son Reuters Pharma, update of September 4, 2012).

PNB-03, PNB-04, and PNB-05 (PharmaNeuro-Boost NV, Limburg, Belgium) are nootropic agents forthe potential treatment of PD (PNB-03), AD (PNB-04),and obsessive compulsive disorders (PNB-05) (Thom-son Reuters Pharma, update of February 1, 2012). Thestructures were not communicated.

PTI-125 (Pain Therapeutics Inc., Austin, TX)reduced A�-related AD pathogenesis by targeting fil-amin A [763] (Thomson Reuters Pharma, update ofAugust 3, 2012). The structure was not communicated.

RP-4000 (Reviva Pharmaceuticals, San Jose, CA)is a small molecule nootropic agent (Thomson ReutersPharma, update of December 27, 2011). The structurewas not communicated.

SEL-103 (Selvita Life Sciences Solutions, Krakow,Poland and Orion, Espoo, FL) is a program inves-

tigating small molecule nootropic agents (ThomsonReuters Pharma, update of July 4, 2012). Structureswere not communicated.

SKL-A4R (SK Biopharmaceuticals, formerly SKLife Science, Fair Lawn, NJ) is a small moleculenootropic agent (Thomson Reuters Pharma, update ofMarch 19, 2012). The structure was not communicated.

TYP-1 (NOBEL ILAC, Istanbul, Turkey) is a smallmolecule neuroprotectant (Thomson Reuters Pharma,update of July 30, 2011). The structure was not com-municated.

The development of ABIO-08-01 (BTG-1640;Abiogen under license from British TechnologyGroup, BTG), ADS-8703 (Adamas Pharmaceuticals),AIP-002 (American Home Products, Wyeth, nowPfizer), AIT-034 (Spectrum Pharmaceuticals, formerlyNeoTherapeutics), Alzheimer’s disease therapeutics(DuPont/Scios), ALE-26015 (Allelix Pharm-Eco LP),aloracetam (Hoechst, now sanofi), Alzene (Bar IlanUniversity and Ivax); AQW-051 (Novartis; Phase IItrials for treatment of schizophrenia and PD are ongo-ing), ASP-2535 and ASP-2905 (Astellas), AVN-397(Avineuro Pharmaceuticals; in Phase II trials for thetreatment of anxiety), AWD-52-39 (Alzneimittel-werke Dresden, elbion), AZD-2858 (AstraZeneca),BCE-001 (4-chloro-phenoxy-acetic acid-1,3-bis(dimethylamino)-2-propylester [764–766]), BD-1054(Russian Academy of Medical Science), BGC-20-0406 (RS-0406; SEN-1269 [767]; Senexis underlicense from BTG under license from Sankyo), BGC-20-0466 (BTG), BGC-20-1178 (Senexis under licensefrom BTG under license from Sankyo), BGC-20-1259(BTG under license from Sankyo), BMS-181168(BMY-21502; Bristol-Myers Squibb [768–770]),BTG-4247 (BTG), BW-394-U (GSK), CG-301338(CrystalGenomics), CL-287663 (KE-748; LederleLaboratories, now Pfizer), of CLL-502 (CLL Pharma),CM-2433 (Cenomed, a subsidiary of Abraxis Bio-Science in collaboration with Medical College ofGeorgia), CX-417, CX-423 and CX-438 (CortexPharmaceuticals), DW-514 and DW-0811 (DaewonPharm), DWJ-209 (Daewoong Pharmaceutical Co.),of the coumarin derivative ensaculin (KA-672;Dr. Willmar Schwabe [771–777]), ETX-6765 (e-Therapeutics), F-94517 (Pierre Fabre), gedocarnil(Bayer Schering Pharma), GM-1416 (GliaMed Inc.),GT-4035 and GT-4054 (Gliatech), HL-026 (HanAllBiopharma), HMR-2420 (Hoechst Marion Roussel,now sanofi), HSB-13 (EncephRx, a spin-out of theSouthern Methodist University and the University ofTexas [1719]), HTC-867 (Wyeth, now Pfizer), IQ200and IQ-201 (Immune Network under license from

Page 28: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

28 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

the University of British Columbia), JNJ-39393406(Janssen Pharmaceutica), LX-104 (Laxdale, AmarinCorp.), of the MBARC program (EnVivo Phar-maceuticals), MK-0249 (Merck & Co), MK-5757(Merck & Co), MG-19649 (Molecular Geriatrics,now Applied Neuro Solutions), MP-100 (AddictionTherapeutix), MR-708 (Medea Research), NDD-094(Novartis [778]), NeoEA-1001 (neoCodex), nooglutil(Russian Academy of Medical Sciences [779]), NSA-789 (Wyeth, now Pfizer), OF-5858 and OF-6145 (AllRussian Research Institute of Pharmaceutical Chem-istry), Oligotropin (HF-0420; Loyola University ofChicago), Org-31433 (Organon Biosciences, nowMSD OSS BV), PF-05297909 (Pfizer), Prisotinol(CGS-5649B; Ciba-Geigy, Novartis [780–783]), pro-caine hydrochloride (Samaritan Pharmaceuticals),PRX-4001 and PRX-4006 (Proximagen), R-641 andRo-40-1641 (both Roche), RU-47067 and RU-52583(Roussel-Uclaf, now sanofi), sabeluzole (JanssenPharmaceutica NV, Johnson & Johnson [784–789]),SCH-54388 (Schering-Plough, now Merck; a metabo-lite of felbamate), sibopirdine (EXP921; Du PontMerck [790]), silicon-containing pyridylsubstitutedisoxazoles (Latvian Institute of Organic Synthesis),SLV-351 (Solvay Pharmaceuticals, now AbbottLaboratories), SN-104 (Sention), SNK-882 (SanwaKagaku Kenkyusho), SPPI-339 (SPI-339, NEO-339;Spectrum Pharmaceuticals, formerly NeoThera-peutics), SRA-997 (Novartis), ST-587 (BoehringerIngelheim), T-9021 and T-9022 (QRxPharma), TAK-065 (Takeda), tenilsetam (Hoechst Marion Roussel,now sanofi [791, 792]) trifusal (Grupo Uriach),V-0191 (Pierre Fabre) and of Z-4105 (Zambon) wasterminated.

PEPTIDES

Cerebrolysin (Ebewe Pharmaceuticals, Vienna,Austria, launched, available in 1 ml, 5 ml, and 10 mlampoules and in vials of 30 ml and 50 ml for intramus-cular or intravenous injection or intravenous infusion)is “derived through a biotechnological procedure fromhighly purified porcine brain proteins and is com-prised of free amino acids and biologically active,short chain peptides” [793]. Cerebrolysin acted asa presynaptic GABAB receptor agonist [794]. Thenootropic effects of cerebrolysin were investigated indepth [795–799]. Cerebrolysin protected neurons fromischemia-induced loss of microtubule-associated pro-tein 2 [800]. Effects of cerebrolysin on A� depositionin transgenic mice were studied extensively [801–807].

The pharmacology of neurotrophic treatment withcerebrolysin was reviewed [808]. Clinical data werepublished (in chronological order) [793, 809–823].First clinical results of a combination treatment of cere-brolysin and donepezil were reported [824]. Data ofcerebrolysin in AD were compiled [825]. The safetyprofile of cerebrolysin in dementia and stroke trials wasdescribed [826].

Cortexin (Geropharm, St. Peterburg, Russia)is a launched polypeptide with neuroprotective,nootropic and antioxidant properties, for the intra-muscular treatment of central nervous system injurycaused by cerebrovascular accidents, encephalitis,encephalopathies, epilepsy and trauma (ThomsonReuters Pharma, update of October 05, 2012). Thestructure was not communicated.

Davunetide (intranasal, AL-108, NAP, Allon Ther-apeutics, Vancouver) is an 8-amino acid peptide(NAPVSIPQ) derived from the activity-dependentneuroprotective protein. A Phase III clinical trial wasinitiated for the treatment of patients with progres-sive supranuclear palsy in December 2010. A PhaseII clinical trial in AD patients started in January 2007(Thomson Reuters Pharma, update of July 17, 2012).Allon Therapeutics is also developing injectable intra-venous and subcutaneous formulations of davunetide.

There are numerous publications on the extensivepreclinical characterization of davunetide describ-ing protection of the brain against ischemic injuryin rats [827], inhibition of the aggregation of A�[828], decrease of anxiety-like behavior in aging mice[829], reduction of the severity of traumatic headinjury [830], reduction of accumulation of A� andof tau hyperphosphorylation [831–833], stimulationof microtubule assembly [834, 835], enhancementof cognitive behavior in transgenic mice [836]),and improvement in motor function and reductionof �-synuclein inclusions in mice overexpressing�-synuclein [837]. Reviews on davunetide werepublished [838–847]. The effects of davunetide on cog-nition and functional capacity in schizophrenia weredescribed [848] (Thomson Reuters Pharma, update ofSeptember 21, 2012).

AM-111 (XG-102, D-JNKI-1; Auris Medical andXigen, a spin off from the Centre Hospitalier Uni-versitaire Vaudois and the University of Lausanne) isan injectable protease-resistant peptidic derivative ofthe JNK-inhibiting protein IB-1 for the i.v. treatmentof acute sensorineural hearing loss, stroke, and AD[849–852]. A Phase IIb trial for acute sensorineuralhearing loss was initiated in January 2009 (n = 210) andresults are expected in autumn of 2012. Auris Medical

Page 29: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 29

is also investigating a topical gel formulation of AM-111 (Thomson Reuters Pharma, update of August 31,2012).

Etanercept (ENBREL, NK-001; Neurokine Phar-maceuticals, Vancouver) is a recombinant fusionprotein of 934 amino acids (mol. weight: 150 kDa)composed of a dimer of the extracellular portion ofhuman TNFR-2 fused to the Fc fragment of humanIgG1 [853, 854]. A rapid cognitive improvementfollowing perispinal etanercept administration to 15probable-AD patients treated once weekly for 6 monthswas reported [855–857]. See also the commentary[858]. In December 2010, the drug was in Phase IIdevelopment for the potential treatment of neurocogni-tive impairment following coronary artery bypass graftsurgery (Thomson Reuters Pharma, update of July 9,2012).

FGL (ENKAM Pharmaceuticals, Copenhagen,Denmark) is a peptide neural cell adhesion moleculemimetic for the potential treatment of AD and stroke.In December 2011, clinical studies including threePhase I studies, one proof-of-concept Phase IIa studyin AD, and a pilot study in patients recovering fromstroke was planned to begin in 2012 (Thomson ReutersPharma, update of April 23, 2012). Enkam was previ-ously developing FGLL, a peptide neural cell adhesionmolecule mimetic, for the potential intranasal treat-ment of AD and stroke. In May 2005, the drug wasshown to be safe and well tolerated in a Phase I studyfor AD. By December 2011, the drug was no longerbeing developed due to the lack of flexibility for theroute of administration.

Glypromate (Gly-Pro-Glu) is naturally cleavedfrom the N-terminal sequence of IGF-I and displayedneuroprotective actions in vitro and in vivo [859–861].It protected against A�-induced somatostatin depletionin rat cortex [862, 863].

NNZ-2566 (Neuren Pharmaceuticals, Auckland,NZ) (Fig. 11) is an analogue of glypromate withan additional �-methyl-group on the proline moiety,which resulted in an improved half-life and betteroral bioavailability. In March 2012, a randomized,double-blind, placebo-controlled Phase I study wasinitiated in healthy volunteers (n = 32) in Australia.Potential indications are mild traumatic brain injury,Rett syndrome, PD, and AD [864– 867]. Excellent syn-thetic organic chemistry was described [868–871]. InNovember 2012 the IND was filed for a Phase II trial.At that time an application was submitted to the TexasChildren’s Hospital IRB and enrollment was expectedto begin pending approval by the FDA and the TexasChildren’s Hospital IRB. (Thomson Reuters Pharma,

update of November 23, 2012). (Thomson ReutersPharma, update of August 31, 2012). Neuren is alsodeveloping a formulation for intravenous infusion.

There are many peptides in preclinical evaluation forthe potential treatment of AD (in alphabetical order):

AL-408 (Allon Therapeutics, Vancouver) is theorally active D-amino acid derivative of davune-tide (AL-108) and an active element of the PARP-1activating activity-dependent neuroprotective protein(Thomson Reuters Pharma, update of November 9,2011).

Alzimag (IMAGENIUM, Paris, France) is a peptidefor the potential treatment of AD (Thomson ReutersPharma, update of December 22, 2011). The structureof the compound were not communicated.

C3bot peptides (Charite Medical School Berlin andHannover Medical School) are short neuron-specificneuritogenic peptides derived from the C3 exoenzymeof Clostridium botulinum, which transiently activatedRhoA for the potential treatment of neurodegenera-tive disorders including AD, PD, Huntington’s chorea,spinal cord and traumatic brain injury [872–875](Thomson Reuters Pharma, update of June 7, 2012).

COG-112, COG-133, and COG-1410 (CognosciInc., Research Triangle Park, NC) are apoE-mimeticpeptides that showed consistent reduction of both A�deposition and of tau hyperphosphorylation in three tautransgenic mouse models and in two A�PP transgenicmouse models [876, 877] (Thomson Reuters Pharma,update of June 5, 2012 for COG-112 and of September11, 2012 for COG-133 and COG-1410).

G-79 (BN-201; Bionure, Barcelona) is a peptide forthe potential treatment of multiple sclerosis, ALS, AD,PD, and glaucoma (Thomson Reuters Pharma, updateof July 4, 2012).

KIBRA pathway modulators (Amnestix Inc., awholly owned subsidiary of SYGNIS Pharma; Heidel-berg, Germany) offer a new genetic link to cognitionthat may benefit patients suffering from AD. For theassociation of KIBRA and late onset AD, see [878];for enhancement of cognition in anxiety disorders[879] (Thomson Reuters Pharma, update of August14, 2012).

Leptin reduced the accumulation of A� and phos-phorylated tau in rabbit organotypic slices [880–882].The company Neurotez (Bridgewater, NJ) is inves-tigating leptin as an A� synthesis inhibitor for thepotential treatment of AD. The company planned tofile an IND in 2012. Effects of leptin on memoryprocessing were described [883–885]. Leptin inducedproliferation of neuronal progenitor cells [886] (Thom-son Reuters Pharma, update of August 24, 2012).

Page 30: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

30 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

MT-007 (MT-007-LRPIV; recombinant LRP frag-ments; Socratech LLC, Rochester NY) is produced bystable transfected baby hamster kidney cells express-ing LRP-IV. The preparation improved cerebral bloodflow, learning and memory in a mouse model of AD(Thomson Reuters Pharma, update of April 23, 2012).

Netrin-1 (BioMarin Pharmaceuticals, Novato, CA,under license from the Buck Institute) interacted withA�PP and regulated A� production [887, 888]. Netrin-1 increased soluble A�PP� and decreased A�1-40and A�1-42 levels in a J20 mouse model of AD.An intranasal delivery is planned (Thomson ReutersPharma, update of July 20, 2012).

NNZ-4921 and NNZ-4945 (NRP-2945, an 11-mer;CuroNZ under license from Neuren Pharmaceuticals,Auckland, NZ) are neuronal regeneration peptides forthe potential treatment of multiple sclerosis and motorneuron disease, respectively [889] (Thomson ReutersPharma, updates of June 18, 2012 and September 7,2012, respectively).

NRG-101 (Mind-NRG, Geneva, Switzerland underlicense from ProteoSys, Mainz, Germany) is aninjectable neuregulin peptide for the potential treat-ment of PD, AD, and schizophrenia (Thomson ReutersPharma, update of July 6, 2012).

NT-1 and NT-2 (Neurotez, Bridgewater, NJ) aresmall peptides, which block the interaction betweenmutant presenilin and cytoplasmic linker protein 170(CLIP-170) that is linked to increased A� levels (BothThomson Reuters Pharma, update of June 6, 2012).

NX-210 (Neuronax, Saint Beauzire, France) is thelead compound of a series of peptides for the potentialtreatment of spinal cord injury and other neurologicaldisorders, such as stroke, AD, PD, and traumatic braininjury (Thomson Reuters Pharma, update of June 29,2012). The structure was not communicated.

Pepticlere (DP-68 and DP-74; ProteoTech, Kirk-land, WA) is the name of small molecule nasal sprays,6- to 9-mer peptides that inhibit A� fibril forma-tion (Thomson Reuters Pharma, update of August 22,2012). The structures were not disclosed.

PP-0301 (Hybio Pharmaceutical, Guangdong,China) is a polypeptide for the potential treatment AD(Thomson Reuters Pharma, update of September 20,2011).

RAP-310 (Rapid Pharmaceuticals, Zug, Switzer-land) is a small stabilized receptor active peptidetargeting the CCR5 receptor for the potential treatmentof AD (Thomson Reuters Pharma, update of October1, 2012). The structure was not communicated.

RG-01, RG-09, and RG-018 (ReGen Therapeutics,London) are neuroprotectant peptides from colostrinin

(vide infra) for the potential treatment of neurode-generative diseases including AD (Thomson ReutersPharma, update of June 27, 2012). The structures werenot communicated.

SX-AZD1 (Serometrix, Pittsford, NY) is a peptidemimetic interacting with APOE4 (Thomson ReutersPharma, update of July 29, 2011). The structure wasnot communicated.

XD4 is a heptapeptide isolated from a Ph.D.-C7library through phage display that rescued memorydeficits in AD transgenic mice by significantly inhibit-ing A�42-induced cytotoxicity, increasing microglialphagocytosis of A�, and decreasing A�-induced gen-eration of ROS and nitric oxide [890].

Colostrinin (ReGen Therapeutics, London) is apolypeptide complex isolated from ovine colostrumcontaining a high proportion of proline (25%) andhydrophobic amino acids (40%). It is composed ofpeptides of molecular mass up to 3,000 Da. It reducedaggregation of A� [891, 892] and alleviated A�-induced toxicity [893]. It facilitated learning andmemory in rats [894] and chicks [895]. Effects on geneexpression were described [896]. Clinical results werepresented (in chronological order) [897–901]. Reviewson colostrinin were published [902, 903].

The development of colostrinin for the treatmentof AD patients was discontinued. The compound waslaunched as a nutraceutical in Australia in July 2007(Thomson Reuters Pharma, update of May 17, 2011).

The development of ANA-1 and ANA-5 (AlzhymePty; phage peptides, which bind to A� to inhibit its gen-eration of hydrogen peroxide [904]), adrenomedullinpeptides (National Institutes of Health), AS-602704(Merck Serono under license from Axonyx), C-AVP-(4-9) (Yakult Central Institute for MicrobiologicalResearch), ebiratide (Hoe-427; Hoechst, now sanofi;an ACTH (6-9) derivative [905–907]), gilatide(a nonapeptide of Axonyx under license fromThomas Jefferson University), metallothionein (Neu-rosciences Victoria and the University of Tasmania[908, 909]) and of noopept (GVS-111, SGS-111;DVD-111; Saegis Pharmaceuticals under license fromthe Russian Academy of Sciences; a nootropicdipeptide analogue of piracetam [910–920]) wasterminated. The development of NC-1900 (Nip-pon Chemiphar, the active fragment analog ofarginine vasopressin [921–925]), PR-21C (Phar-maxon; a polysialylated form of the neural celladhesion molecule) and of RGX-100 and RGX-200 (RemeGenix Inc., Cortica Neuroscience; A�production-inhibiting BRI2-derived peptides for theintranasal administration to AD patients [926–933])

Page 31: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 31

was discontinued. The development of SEM-606 (Uni-versity of Manchester) and of TKP-1001 (EUSAPharma, formerly Tasliker under license from TheOpen University) was terminated.

DRUGS PREVENTING AMYLOID-�AGGREGATION

Plasma A� levels can be linked directly to spe-cific cognitive changes that constitute the conversionfrom MCI to AD [934–939]. The relationship betweenatrophy and A� deposition in AD was investigatedthoroughly [940–942]. A review of the literature cor-relating Alzheimer disease neuropathologic changeswith cognitive status was provided [947]. The molecu-lar and neurophysiological mechanisms of amyloid-�and cognition in Alzheimer’s disease were outlined[1720]. A rapid decline of memory in healthy olderadults with high amyloid-� load was described [1721],which was more important than ApoE genotype[1722]. Cognitive decline in adults with high amyloid-� load was evaluated [1723]. A� assemblies mediatedrapid disruption of synaptic plasticity and memory[943]. The topic “A� toxicity in Alzheimer’s disease”was reviewed [944, 945]. A critical review on the amy-loid cascade hypothesis was published [946]. AlthoughA� plaques may play a key role in AD pathogenesis,the severity of cognitive impairment correlates bestwith the burden of neurofibrillary tangles [947]. Thekey interactions of apolipoprotein E and A� pathologywere reviewed [948].

The inhibition and reversion of A� misfolding andaggregation is an approach, which has been followedup by many research groups during years. Excellentreviews dealing with this subject were published (inchronological order) 1996: [949], 1998: [950, 951],1999: [952–956], 2001: [957], 2002: [958–960], 2005:[961, 962], 2006: [963], 2007: [964, 965], 2009: [495,966–968], 2010: [969–971], 2011: [972, 973], 2012:[974–976].

Tafamidis (PF-06291826, Fx-1006, Vyndaquel,Pfizer following its acquisition of FoldRx Pharma-ceuticals under license from the Scripps ResearchInstitute) (Fig. 12) is small-molecule transthyretin sta-bilizer for the oral treatment of transthyretin familialamyloid polyneuropathy [977–980]. The drug wasapproved by EMA in November 2011 was launchedin Europe in March 2012 [1724]. In June 2012, theFDA issued a complete response letter and requestedthe completion of a second efficacy study. For the iden-tification of A� binding sites on transthyretin see [981];

for transthyretin amyloidosis see [982, 983] (ThomsonReuters Pharma, update of September 25, 2012).

Eprodisate (1,3-propanedisulfonate disodium salt;NC-503, Fibrillex, Kiacta; Celtic Therapeutics, St.Thomas VI under license from Bellus Health) (Fig. 12)is an orally active sulfated glycosaminoglycan mimeticdesigned to inhibit the formation and deposition ofamyloid fibrils. A Phase III study was initiated inDecember 2010 for the treatment of renal disease inpatients with AA amyloidosis [984–987] (ThomsonReuters Pharma, update of September 5, 2012).

For ARC-029 (Archer Pharmaceuticals, RoskampInstitute, Sarasota, FL; Phase III clinical trials),see Chapter 7, Drugs interacting with Ion Channels( /= Receptors).

For Davunetide (intranasal of via i.v. or s.c admin-istration; Allon Therapeutics, Vancouver, Phase IIIclinical trials), see Chapter 15, Peptides.

For APH-0703 (Aphios Corp., Woburn MA), ananoparticle formulation of APPH-9601 in Phase IIclinical trials since May 2010 (Thomson ReutersPharma, update of July 5, 2012), see Part 2, Chapter2.35, Drugs interacting with Protein Kinase C.

Doxycycline hyclate (Fondazione IRCCS Policlin-ico San Matteo, Pavia, Italy) is in a Phase II study(NCT01171859) in patients with transthyretin amy-loidosis (n = 40) since July 2010. At oral doses of200 mg for three months, doxycycline produced asignificantly lower decline in the ADAScog scores in101 mild-to-moderate AD patients [988] (ThomsonReuters Pharma, update of April 27, 2012).

ELND-005 (AZD-103; scyllo-inositol; scyllo-cyclohexanehexol, Elan, Dublin, Ireland, and EllipsisNeurotherapeutics, formerly Transition Therapeutics,Toronto, Canada) (Fig. 12) is an orally availableinhibitor of A� peptide aggregation. Results of a PhaseII dose-ranging, randomized, placebo-controlled studyin 351 patients with mild-to-moderate AD treated for78 weeks with either 250, 1,000, or 2,000 mg/day werereported. Patients with mild AD receiving 250 mg ofELND-005 had higher scores on the Neuropsycho-logical Test Battery compared with placebo, whichwere significant. In contrast ADCS-ADL scores werenot significant. The two higher doses were discon-tinued [989]. In August 2012, a Phase II studywas initiated in bipolar disorder patients in the US.The study is expected to be completed in August2014 (Thomson Reuters Pharma, update of September14, 2012).

The endogenous brain inositols stabilized smallaggregates of A�, which are non-toxic to NGF-differentiated PC-12 cells and primary human neuronal

Page 32: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

32 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 12. Drugs interacting with amyloid-�.

Page 33: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 33

cultures [990]. In particular scyllo-inositol, whengiven orally to transgenic mice, reversed AD phe-notype, improved impaired cognition, and alteredcerebral A� pathology [991, 992]. Scyllo-inositoldose-dependently rescued long-term potentiation inmouse hippocampus from the inhibitory effects of sol-uble oligomers of cell-derived human A� [993, 994].Elevated scyllo-inositol concentrations were found inpatients with AD [995].

Syntheses of scyllo-inositol derivatives have beenreported [996]. The synthesis of the PET ligand [18F]-1-deoxy-1-fluoro-scyllo-inositol was described [997].A comparison of three amyloid assembly inhibitors,scyllo inositol, epigallocatechin gallate and the molec-ular tweezer CLR01 (vide infra) was published[998].

SOM-0226 (SOM Biotech SL, Barcelona) is a drugfor the potential treatment of transthyretin amyloido-sis in Phase II clinical development (Thomson ReutersPharma, update of May 30, 2012). The structure wasnot communicated.

For AAD-2004 (GNT Pharma, Suwon, SouthKorea) in Phase I clinical trials (Thomson ReutersPharma, update of May 4, 2012). See Part 2, Chap-ter 2.34. Drugs interacting with Prostaglandin D & ESynthases, Part 2, Fig. 22.

Beta amyloid modulator (Medipost, Seoul) is inPhase I clinical trials in South Korea since Septem-ber 2011 for the potential treatment of AD (ThomsonReuters Pharma, update of May 7, 2012). The structurewas not communicated.

BLU-8499 (formerly NRM-8499; Bellus Health,formerly Neurochem, Quebec) is a prodrug oftramiprosate (vide infra). It was evaluated in a single-center, randomized, double-blind, placebo-controlledPhase I study in 84 young and elderly healthy subjectsto assess safety, tolerability and pharmacokinetics,which started in March 2010. The data were reportedin January 2011. It showed improved gastrointesti-nal tolerability and lower inter-individual variabilityof drug exposure compared to comparable doses oftramiposate (Thomson Reuters Pharma, update ofSeptember 5, 2012).

DWP-09031 (presumed to be DWJ-301; DaewongPharmaceutical Co. in collaboration with Medifron,both South Korea) inhibited the production and aggre-gation of A�. By January 2012, the Korean FDAhad approved an IND for a Phase I trial. Therandomized, double-blind, placebo-controlled study(NCT01522586) is planned in healthy male volun-teers (n = 64) to assess the safety, pharmacokinetics andpharmacodynamics of DWP-09031 (Thomson Reuters

Pharma, update of July 27, 2012). The structure wasnot disclosed.

For Exebryl-1 (ProteoTech, Kirkland, WA andChina licensee Tasly Pharmaceuticals; in Phase I clin-ical trials), see Chapter 13, Natural Products.

For NP-61 (NP-0361; Noscira, previously Neu-ropharma, Madrid; Phase I clinical trials; ThomsonReuters Pharma, update of May 10, 2012), see Part2, Chapter 2.1.1.2., Dual acetylcholinesterase and A�inhibitors.

Systebryl (ProteoTech, Kirkland, WA) is the leadof a series of small molecules including PTI-19 andPTI-51 for the potential treatment of systemic AA amy-loidosis. In December 2011, Systebryl was in Phase Istudies (Thomson Reuters Pharma, update of Decem-ber 21, 2011). Structures were not communicated.

There are many A� aggregation inhibitors in pre-clinical evaluation (in alphabetical order):

Alzheimer’s disease therapeutics (BioChromixPharma AB, Solna, Sweden) are compounds, whichachieved a significant reduction in plaque load andneurotoxic A� aggregates. (Thomson Reuters Pharma,update of August 24, 2012). The structures were notcommunicated.

A� oligomer cellular prion protein bindinginhibitors (AstraZeneca under a sublicense from Axe-rion Therapeutics under license from Yale University)are interacting with the cellular prion protein, thehigh affinity receptor for A� oligomers [999–1003](Thomson Reuters Pharma, update of August 17,2012). No structures were disclosed.

A� protein inhibitors (Neuropore Therapies, SanDiego CA) are peptidomimetic compounds that inter-fere with A� aggregates (Thomson Reuters Pharma,update of July 16, 2012). Structures were not commu-nicated.

A�/tau protein aggregation inhibitors (CNRS,FIST SA, Paris, France) are investigated for the poten-tial treatment of AD (Thomson Reuters Pharma, updateof August 24, 2012). Structures were not communi-cated.

Amyloid-derived diffusible ligands are investi-gated by Merz Frankfurt, Germany under license fromAcumen Pharmaceuticals (Thomson Reuters Pharma,update of June 12, 2012). The structures of the com-pounds were not communicated.

ARN-4261 (New York University and Aria Neu-rosciences, Hamden CT) (Fig. 12) and ARN-2966(Fig. 12) are inhibitors of A� aggregation. ARN-2966reduced A� deposition and memory deficit in trans-genic mice. Following treatment a significant 25%reduction in A� plaque load was noted compared with

Page 34: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

34 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

mice that received a vehicle control. The data demon-strated that ARN-2966 was biostable and well toleratedin transgenic mice with blood-brain barrier penetra-tion after both oral and intravenous dosing (ThomsonReuters Pharma, update of August 28, 2012).

AVCRI-104P4 (University of Barcelona) (Fig. 12)inhibited A� aggregation including acetylcholines-terase- and self-induced-A� aggregation, �-secretase,acetylcholinesterase, and butyrylcholinesterase (Tho-mson Reuters Pharma, update of September 28, 2012).

AVN-457, AVN-458 and AVN-492 (Avineuro Phar-maceuticals, San Francisco) are nootropic agents forthe potential treatment of cognitive disorders (Thom-son Reuters Pharma, update of October 22, 2012). Thestructures were not disclosed.

AZP-2006 (AlzProtect, Loos, France in collabora-tion with INSERM and the University of Lille II) is anA�PP modulator of undisclosed structure (ThomsonReuters Pharma, update of July 4, 2012).

Beta-amyloid aggregation inhibitors (MedisynTechnologies, Minnetonka, MN / Mount Sinai Schoolof Medicine) demonstrated significant A�-loweringand anti-aggregation activity in vitro. Two compoundsalso reduced the amount of A� in mouse brain in vivo(Thomson Reuters Pharma, update of June 21, 2012).Structures were not communicated.

Beta-amyloid/alpha-synuclein/tau aggregationinhibitors (Snowdon Inc., Vancouver) are expectedto enter Phase I trials in 2012 (Thomson ReutersPharma, update of August 19, 2011). The structuresof the compounds were not communicated.

Beta amyloid beta sheet formation inhibitors (ACImmune, Lausanne, Switzerland) are small molecules,which inhibited the aggregation of A� to oligomericand fibrillar species for the potential treatment of ADand glaucoma [1004]. Two 3-aminopyrazole moietiescarrying additional aryl substituents were connectedvia a linker. The concept of 3-aminopyrazoles with adonor-acceptor-donor hydrogen bond pattern compli-mentary to that of the �-sheet of A�42 was first investi-gated by Schrader and colleagues [1005–1013] (Thom-son Reuters Pharma, update of September 24, 2012).

Beta-amyloid inhibitor (Icogenex, Seattle, WA) isa small molecule therapeutic that normalizes the pro-duction of A� (Thomson Reuters Pharma, update ofJune 26, 2012). The structure was not disclosed.

Beta-amyloid inhibitor (Star Scientific, Glen AllenVI through its subsidiary Rock Creek Pharmaceuticalsin collaboration with the Roskamp Institute) reducedA� when applied to cells (Thomson Reuters Pharma,update of January 3, 2012). The structure was notdisclosed.

Beta-amyloid modulators (Crossbeta Biosciences,Utrecht, The Netherlands) are small molecules, whichtarget A� oligomers and misfolded proteins (ThomsonReuters Pharma, update of August 24, 2012). Struc-tures were not communicated.

Beta-amyloid precursor protein modulators(Alzcor Pharmaceuticals, Arlington, MA) are inves-tigated for the potential treatment of AD (ThomsonReuters Pharma, update of July 27, 2011). Structureswere not disclosed.

BMS-869780 (Bristol-Myers Squibb) (Fig. 12)reduced A�42 levels in transgenic mice at oral dosesand displayed an IC50 value at A�42 in the lownM range. It was not hepatotoxic (Thomson ReutersPharma, update of August 6, 2012).

BTA-EG4 (Johns Hopkins University and George-town University) is a benzothiazole aniline derivativeand amyloid-� synthesis inhibitor for the potentialtreatment of Alzheimer’s disease (Thomson ReutersPharma, update of October 30, 2012). The structurewas not communicated.

C36 (Medisyn Technologies, Minnetonka, MNin collaboration with the Mount Sinai School ofMedicine) is an A�40 and A�42 lowering smallmolecule for the potential treatment of AD (ThomsonReuters Pharma, update of December 6, 2011). Thestructure was not disclosed.

Caprospinol (SP-233; Samaritan Pharmaceuticals,Las Vegas, NV under license from Georgetown Uni-versity) (Fig. 12), a spirostenol drug, blocked theoligomerization of A�42 exerting a direct effecton mitochondria [1014–1017] (Thomson ReutersPharma, update of March 18, 2011).

Carvedilol is a launched unselective �1, �1 and �2blocker, which inhibited A� fibril formation [1018].

CLR01 (University of Duisburg-Essen, RensselaerPolytechnic Institute, NY and UCLA) (Fig. 12) is amolecular tweezer, which binds to lysine residues withmicromolar affinity and interferes with a combinationof hydrophobic and electrostatic interactions that areimportant in the self-assembly of most amyloidogenicproteins including A�, tau, and �-synuclein [1007,1018–1024].

CLR-097(Clera Inc., Toronto) inhibited the processof A� plaque formation (Thomson Reuters Pharma,update of December 5, 2011). The structure was notdisclosed.

Cotinine, a natural metabolite of nicotine, may be apotential new therapeutic agent against AD [1025]. Itreduced amyloid-� aggregation and improved memoryin Alzheimer’s disease mice [1725].

Daunomycin inhibited A� fibril formation [1018].

Page 35: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 35

DBT-1339 (Medifron, Seoul and licensee Dae-woong, South Korea) (Fig. 12) is the lead compoundfrom A� protein deposition inhibitors (ThomsonReuters Pharma, update of June 1, 2012).

Enoxaparin (Enox, a low molecular weight hep-arin) lowered brain A� load in a mouse model of AD[1026]. It also improved cognition in APPSWE/PS1dE9mice [1027, 1028].

GAG/carbohydrate compounds (ProteoTech,Kirkland, WA) are glycosaminoglycan modulatorsfor the potential treatment of AD (Thomson ReutersPharma, update of June 7, 2012). Structures were notdisclosed.

Galantamine inhibited A� aggregation and cyto-toxicity [1029].

haw-AD-14 (Hawthorn, Madison, Mississippiunder license from CoPlex) is an A� synthesis and tauphosphorylation inhibitor (Thomson Reuters Pharma,update of September 26, 2011). The structure was notcommunicated.

HO-4160 (University of California at Davis)(Fig. 12) is a spin-labeled fluorene compound thatspecifically disrupted A� oligomers [1030].

Imipramine in part through inhibition of TNF-�prevented cognitive decline and A� accumulation in amouse model of AD [1031].

IPS-04001, IPS-04001 and IPS-04003 (InnoPhar-maScreen, South Korea) is a specific inhibitorof A� peptide and VEGF-165 interaction. ByJune 2012, in vivo efficacy studies had shown asignificantly enhanced effect on the memory ofNSE-PS2/N141Ltransgenic mice (Thomson ReutersPharma, update of July 16, 2012). The structure werenot communicated.

KMS-88 series (Hanmi Pharmaceutical, SouthKorea, the Korea Institute of Science and Technologyand Seoul National University) (Fig. 12) is a series ofaminostyryl-benzofuran derivatives inhibiting A� fib-ril formation [1032, 1033] (Thomson Reuters Pharma,update of September 12, 2012).

Minocycline is a second generation tetracyclinethat can effectively cross the blood-brain barrier.It improved cognitive impairment in AD models[1034]. It provided protection against A�25-35 inducedalterations of the somatostatin signaling pathway[1035, 1036]. It reduced microglial activation [1037,1038]) and A� derived neuroinflammation [1039]. Itrecovered MTT-formazan exocytosis impaired by A�[1040]. It reduced the development of abnormal tauspecies in models of AD [1041–1043]. Minocyclineprotected PC12 cells against NMDA-induced injuryvia inhibiting 5-lipoxygenase activation [1044, 1045].

Minocycline improved negative symptoms in patientswith early schizophrenia [1046]. See also Part 2, Chap-ter 2.25. Drugs interacting with 5-Lipoxygenase.

NPT-4003 (Neuropore Therapies, San Diego CA) isthe lead of heterocyclic compounds that interfere withamyloid-� aggregates. Detailed data were presentedat the ICAD Vancouver in July 2012 [1726]. (Thom-son Reuters Pharma, update of July 16, 2012). Thestructure was not communicated.

Rifampicin inhibited A� aggregation and neu-rotoxicity [1047–1049]. In a clinical trial in 101mild-to-moderate AD patients, statistically significantimprovements in the ADAScog scores after treatmentwith 300 mg rifampicin for three months were found[988]. Rifampicin and caffeine caused an upregula-tion of LRP1 [1050]. The brain efflux index of A�in rifampicin and caffeine treated mice was signifi-cantly higher (82% and 80%, respectively) than thebrain efflux index of control mice (62%). It appearsthat a yet to be identified transporter/receptor plays asignificant role in A� clearance, which is upregulatedby rifampicin and caffeine.

Rolitetracycline is effective as inhibitor of A� fibrilformation [1018].

SD-1002 (Synaptic Dynamics, Farmington CT) is alysosomal modulator that reduced protein depositionof amyloid-�1-42 oligomers for the potential treatmentof Alzheimer’s disease (Thomson Reuters Pharma,updates of October 22, 2012). The structure was notcommunicated.

SEN-1500 (Senexis, Cambridge, UK) (Fig. 12) andfollow-up compound SEN-1576 (structure not dis-closed) are small molecule A� aggregation inhibitors[1727] (Thomson Reuters Pharma, updates of March23 and August 2, 2012, respectively).

Small molecule A� modulators (Asceneuron, aspun-out of Merck Serono, Geneva, Switzerland) aredrugs for the potential treatment of AD (ThomsonReuters Pharma, update of October 3, 2012). Structureswere not disclosed.

SP-08 (Samaritan Pharmaceuticals, Las Vegas, NVunder license from Georgetown University) (Fig. 12)is an A� antagonist with neuroprotectant properties(Thomson Reuters Pharma, update of March 18, 2011).

TAK-070 (University of Tokyo under license fromTakeda) is a �-secretase and A� aggregation inhibitor(Thomson Reuters Pharma, update of July 10, 2012).The structure was not communicated.

Tetracycline(s) showed anti-amyloidogenic activ-ity in vitro [1051] and protected Caenorhabditiselegans from A�-induced toxicity by targetingoligomers [1052].

Page 36: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

36 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

For VP-20629 (Indole-3-propionic acid; OX1; IN-OX1; OXIGON, ViroPharma, Exton PA, under licensefrom Intellect Neurosciences under license from NewYork University and Mindset BioPharmaceuticals;Thomson Reuters Pharma, update of August 10, 2012),see Chapter 11. Antioxidants.

AGT-160 (ArmaGen Technologies, Santa Mon-ica, CA) is a recombinant IgG fusion proteinformed by the fusion of a single chain Fv anti-body against A� plaque formation to the company’shuman insulin receptor-targeting monoclonal anti-body Trojan horse for transport across the blood-brainbarrier for the potential detection and treatment ofAD (Thomson Reuters Pharma, update of January23, 2012). See also Part 1. Chapter 1.16. Insulinreceptors.

EDN-OL1 (Edunn Biotechnology, St. Louis, Mis-souri under license from St. Louis University) is anantisense oligonucleotide targeting A� production,which improved memory and learning in three differ-ent AD mouse models. By February 2012, EDN-OL1had demonstrated activity in patients with Down syn-drome (Thomson Reuters Pharma, update of August24, 2012).

NPT-001 (NeuroPhage Pharmaceuticals, Cam-bridge, MA) is a filamentous M13 bacteriophage thatdisrupted plaque aggregation through binding of A�.Direct binding of M13 bacteriophage to fibrils ofaggregated A� was observed with high affinity of4 nM. In an AD mouse model, A� plaques werereduced by 70% after a single intracranial adminis-tration (Thomson Reuters Pharma, update of July 16,2012).

Spheron-based therapeutics (Nymox Pharmaceu-tical Corp., Quebec) are compounds capable ofblocking the transformation of human spherons intoplaques [1053, 1054] (Thomson Reuters Pharma,update of February 22, 2012).

A�-aggregation inhibitor research programs wereongoing at Elan [1055], at GrenPharma [1056], at GSK[1057, 1058], at Johnson & Johnson [1059], at Pfizer[1060], at Pharmacia-Farmitalia Carlo Erba [1061] andat Scios [1062].

Excellent papers on A� aggregation inhibitors fromuniversities were presented (in chronological order)1995: [1063], 1996: [1064], 1997: [1065, 1066], 2001:[1067], 2002: [563, 1068], 2004: [1069, 1070], 2005:[1071–1073], 2006: [1074–1082], 2007: [1083–1094],2008: [745, 1043, 1095, 1096, 1097–1105] 2009:[1106–1113], 2010: [391, 551, 980, 1114–1117], 2011:[549, 643, 1118–1122] and 2012: [1033, 1123–1129,1728].

Excellent papers on theoretical calculations onA�-aggregation inhibitors were published (in chrono-logical order) 2006: [1130, 1131], 2008: [1066,1132–1134], 2009: [1135–1137], 2011: [1138, 1139][1140, 1141], 2012: [1142–1144].

The Phase III development of tramiprosate(3-amino-1-propanesulfonic acid, homo-taurine, NC-531, Alzhemed, Cerebril; Bellus Health, formerNeurochem), an orally available glycosaminogly-can mimetic, was terminated in 2007 [1145–1151].The compound was launched as a nutraceutical formemory protection [1152]. Also the development ofALS-499 and ALS-633 (Advanced Life Sciences,in collaboration with Argonne National Labora-tory), amyloid beta aggregation inhibitor (Zyentia),amyloid oligomer specific antagonists (Treventis),amyloid protein deposition inhibitor (ProteoTech),AN-531 (Athena Neurosciences, now Elan Phar-maceuticals), AS-602704 (Ac-FPFFD-NH2, MerckSerono [1153]), beta amyloid aggregation inhibitors(Cortex, Elan, Hybridon, Oceanix, Queens Universityat Kingston and University of Wisconsin-Madison),beta-amyloid modulators (Inflame Therapeutics,Neuro-Hitech and Q-RNA), beta-amyloid precur-sor protein modulators (SIBIA Neurosciences incollaboration with BMS), beta amyloid synthesisinhibitors (BTG), CLR-01 (Clear Therapeutics, aspin-off from UCLA), CT-500 (Creabilis Therapeu-tics), ID-1135 and ID-1567 (Bellus Health), lysosomalmodulators (Synaptic dynamics), MPI-442690 andMPI-442691 (Myriad Genetics in collaboration withthe Mayo Clinic), NC-503 and NC-531 (Neu-rochem; Bellus Health), NHT-0112 (Neuro-Hitech),NOX-A42 (Noxxon Pharma), NVP-AAM147 andNVP-AAM155 (Novartis), phenserine-based amy-loid inhibitors (Message Pharmaceuticals), PPI-368,PPI-558 and PPI-1019 (Apan; all Praecis Phar-maceuticals [953, 957]), PTI-777 (ProteoTech),Reumacon (Meda under license from Conpharm),Ro-47-1816-001, Ro-65-7652-000, Ro-65-8564-001and Ro-8815-001 (Roche), RS-1178 (BGC-20-1178;Senexis under license from BTG under licensefrom Sankyo [1154]), RS-0406 (BGC-20-0406;Senexis under license from BTG under license fromSankyo [1155–1158]) and RS-0466 (Sankyo), RX-AD(Rimonyx Pharmaceuticals), SAN-61 and SAN-161(Sanomune, a subsidiary of DiaMedica), Semapimod(CNI-1493; Cytokine PharmaSciences, CPSI, for-merly Cytokine Networks), a cytokine inhibitor, whichinhibited A� production, plaque formation and cogni-tive deterioration in an animal model of AD [39, 40,1729]), SEN-304, SEN-606, SEN-1186, SEN-1203,

Page 37: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 37

and SEN-1329 (Senexis), SIB-1848 (SIBIA Neu-rosciences, now Merck and Bristol-Myers Squibb),SKF-746532 (SmithKline Beecham, now GSK),synthetic anti-beta sheet peptides (Mayo Foun-dation), THUR-24 (Thuris Corp.), transthyretin(Research Foundation of the State University of NewYork), and of VIP-SSM (University of Illinois) wasterminated.

Ligands interacting with amyloid-β

Since the discovery of the 11C-PiB-PET ligand,tremendous progress has been made in the devel-opment of new PET ligands. Considerable evidencesuggests that A� deposition precedes decline in cogni-tion [1159, 1160]. The development of PET amyloid-�imaging agents was described [1730].

11C-PiB (University Pittsburgh) (Fig. 13) had aKD = 1.4 nM for AD frontal cortex and KD = 4.7 nMfor A� fibrils and did not bind to neurofibrillary tan-gles [1161]. The log P value is 1.2 [1162]. Patientswith AD typically showed a marked 11C-PiB reten-tion (with a significant 1.5 to 2-fold increase) incortical areas known to contain large amounts ofA� plaques [1163–1165]. 11C-PiB positivity in MCIindicated already advanced A� pathology [1166].Significant correlations between 11C-PiB retentionand CSF biomarkers were found in MCI patients[1167]. Clinical severity of AD measured by theClinical Dementia Rating scale sum of boxes corre-lated with 11C-PiB uptake in PET [1168]. 11C-PiBimaging of human immunodeficiency virus-associatedneurocognitive disorder was described [1169]. For adirect comparison of 11C-PiB, 18F-Florbetapir, 18F-Florbetaben, and 18F-Flutemetamol, see [1170] fora comparison between Pittsburgh Compound B andFlorbetapir see [1731]. Longitudinal imaging of ADpathology with 11C-PiB, 18F-FDDNP, and 18F-FDGwas described [1171–1173]. For the prediction of cog-nitive decline via PET of brain A� and tau, see [1174].The clinical perspective of imaging of cerebral A�plaques was discussed [1175, 1205]. The correspon-dence between in vivo 11C-PiB-PET and postmortemassessment of plaques was discussed [1176]. Imag-ing brain amyloid in nondemented adults with Downsyndrome using Pittsburgh Compound B was reported[1732].

Fred Van Leuven and coworkers carried out a sys-tematic evaluation of the isomeric compounds withthe OH groups attached in 4, 5, and 7 positions. The5-hydroxy analogue had the most favorable in vivopharmacokinetics in brain [1177, 1178].

18F-Florbetapir (18F-AV-45; Amyvid; AvidRadiopharmaceuticals, Philadelphia, PA, a subsidiaryof Eli Lilly, under license from the University of Penn-sylvania) (Fig. 13) is a launched 18F PET imaging agenttargeting A�. It had a Ki of 2.87 nM, did not bind totau, and can easily be labeled with 18F by an automatedsynthesis [1179–1187]. By March 2010, more than700 patients were enrolled in a Phase III trial presumedto be the 18F-AV-45-A07 study. Several clinical papersappeared describing imaging with 18F-Florbetapir[1188–1195]. Correlations between 18F-Florpetapirand FDG images were reported [1196]. 18F-Florpetapiris useful to quantify brain amyloid load [1197]. Fora direct comparison of 11C-PiB, 18F-Florbetapir, 18F-Florbetaben, and 18F-Flutemetamol, see [1170]; for aclinical comparison of 18F-Florbetapir and 18F-FDGPET in patients with AD and controls, see [1198]. Aprospective cohort study was described [1199].

A longitudinal assessment of 18-month of cognitivedecline in mild cognitive impairment and Alzheimer’sdisease patients using florbetapir was communicated[1733]. “From Alois to Amyvid” see [1734]. PETimaging was also carried out in a murine model ofamyloid-� plaque deposition [1735].

FDA approved 18F-Florpetapir as a PET imag-ing agent to estimate A� neuritic plaque density inpatients with cognitive impairment on April 6, 2012.Lilly launched the imaging agent in June 2012. Thecompound will be manufactured and distributed byCardinal Health [1200] (Thomson Reuters Pharma,update of September 17, 2012).

18F-Florbetaben (18F-BAY 94-9172; 18F-AV-1/ZK; Piramal Healthcare, Mumbai, India from anasset acquisition from Bayer under license from AvidPharmaceuticals and the University of Pennsylvania)(Fig. 13) is the phenyl analogue of florbetapir. It hasa Ki of 2.22 nM [1181]. In November 2009, a PhaseIII clinical trial in AD patients (n = 292) began toassess safety and efficacy of florbetaben to detector exclude cerebral A�. Data from clinical studieswere reported [1201–1208]. An in vitro characteriza-tion was disclosed [1736]. The one-step radiosynthesiswas described [1209], as was the radiation dosimetry[1210]. In the NCT01138111 trial, 18F-FlorbetabenPET imaging was studied in MCI patients. For adirect comparison of 11C-PiB, 18F-Florbetapir, 18F-Florbetaben, and 18F-Flutemetamol, see [1170]; fora comparison of 11C-PiB and 18F-Florbetaben, see[1211]; for the impact of 18F-Florbetaben PET imag-ing on confidence in early diagnosis of AD, see [1212](Thomson Reuters Pharma, update of September 6,2012).

Page 38: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

38 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 13. Radioligands for PET scans of amyloid-�.

18F-Flutemetamol (GE-067; GE Healthcare, Chal-font, UK and Universities of Pittsburgh and Uppsala)(Fig. 13) has a Ki of 0.74 nM [1181]. Results of aPhase I study were reported [1213], as were resultsof a Phase II study [1214, 1215]. In December 2009,a Phase III trial sponsored by GE Healthcare was ini-tiated in the US. By April 2010, Phase III trials werealso underway in Europe. In April 2012, preliminarydata from the two Phase III studies in terminally illpatients and young healthy subjects were reported.Data showed that the primary endpoint was met.Patients (who underwent brain autopsy) showed con-cordance between 18F-Flutemetamol PET images andAD-associated A� brain pathology and healthy sub-jects showed concordance with the known lack of brainA�. The association between in vivo 18F-FlutemetamolPET imaging and in vivo cerebral cortical histopathol-ogy was described [1216], as was a combination ofbiomarkers PET 18F-Flutemetamol and MRI [1217].For a direct comparison of 11C-PiB, 18F-Florbetapir,18F-Florbetaben, and 18F-Flutemetamol, see [1170].Binary classification of 18F-Flutemetamol PET usingmachine learning was disclosed [1218]. The pharma-cokinetics of 18F-Flutemetamol in wild-type rodents

were reported [1219] (Thomson Reuters Pharma,update of October 3, 2012).

18F-AZD-4694 (Navidea Biopharmaceuticals,Dublin, Ohio, formerly Neoprobe Corporation underlicense from AstraZeneca) is an i.v. PET ligand for thepotential imaging of A� depositions in AD in PhaseII development [1220]. In December 2011, a Phase IIIprogram was planned to start in early 2013 (ThomsonReuters Pharma, update of September 21, 2012). Thestructure was not communicated. For syntheses ofadditional 18F-ligands of AstraZeneca see [1737].

BAY-1006578 (Bayer) is in Phase I clinical trials inFinland and Sweden since June 2010 (n = 36). By Octo-ber 2011, the trial was completed (Thomson ReutersPharma, update of April 30, 2012). The structure wasnot communicated. A potential follow up compound isBAY-1008472 [1221].

123I-MNI-168 (Institute for NeurodegenerativeDisorders, IND, New Haven, CT) is a SPECT lig-and to detect A� deposition in patients with AD. APhase I clinical trial was initiated in the US in January2009 (n = 34). In February 2011, the trial was termi-nated (Thomson Reuters Pharma, update of February10, 2012). The structure was not communicated.

Page 39: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 39

18F-MNI-558 (Molecular NeuroImaging in col-laboration with the Institute for Neuro-degenerativeDisorders, IND, New Haven, CT) is a PET agent thatbinds A�. The compound was in a Phase 0 trial in ADpatients and in volunteers (n = 10) from October 2010to July 2011 (Thomson Reuters Pharma, update ofApril 30, 2012). The structure was not communicated.

11C-BF-227 (Fig. 14) was used for amyloid PETimaging [1222–1225], for PET imaging of �-synucleindeposition [1226, 1227], and for in vivo detection ofprion amyloid plaques [1228].

123I-DRM-106 (Fujifilm Pharma) (Fig. 14) isa radioiodinated imidazopyridine derivative for thepotential use as SPECT imaging agent for A� for thediagnosis of AD (Thomson Reuters Pharma, update ofSeptember 21, 2011).

18F-Hexethal (Pfizer, under license from the Uni-versity of Aberdeen), a barbiturate derivative, is an A�imaging agent (Thomson Reuters Pharma, update ofSeptember 24, 2012).

11C-SB-13 (Fig. 14) was evaluated first in post-mortem brain tissues [1229] and in AD patients [1230].

18F-Florbetapir dimer (Avid Radiopharmaceuti-cals, Philadelphia, PA, Eli Lilly) is a PET imagingagent for the potential diagnosis of cerebral amyloidangiopathy [1231] (Thomson Reuters Pharma, updateof May 8, 2012).

Dicyanovinylnaphtalenes were described for neu-roimaging of A� [1232]. Also 18F-labeled benzoxa-zoles [1233] and 18F-labeled 2-pyridinylbenzoxazolesand 2-pyridinylbenzothiazoles were presented for PETimaging of A� plaques [1234].

The development of 11C-AZD-2184 and 11C-AZD-2995 (both AstraZeneca [1235–1237, 1738])and 18F-FDDNP (Siemens Medical Solutions Molec-ular Imaging and UCLA) (Fig. 14) was terminated.18F-FDDNP was the first PET tracer used in vivo fordetection of cerebral A� plaques in 2002 [1238]. 18F-FDDNP binds to A� and neurofibrillary tangles in AD,to prion plaques in Creutzfeldt-Jakob disease, to A�deposits in cerebral amyloid angiopathy, and to Lewybodies in PD and DLB [1161]. It was used for mea-suring amyloid-� and tau levels in adults with Down’ssyndrome [1739]. The ligand is still used for predic-tion of cognitive decline [1174, 1740]. Longitudinalimaging of AD pathology using 11C-PiB, 18F-FDDNP,and 18F-FDG PET was described [1239]. 18F-FDDNPPET allowed a prediction of cognitive decline basedon hemispheric cortical surface maps [1240].

123I-IMPY (Avid Radiopharmaceuticals, Philadel-phia, PA, under license from University of Pennsylva-nia, Fig. 14) is a SPECT ligand for imaging studies

[1241, 1242]. Its Ki is 15 nM [1243]. The safety andbiodistribution was evaluated [1244]. The signal-to-noise ratio for plaque labeling is not as robust as thatof 11C-PiB. 11C-PiB showed a S/N ratio of about 2.5,while 123I-IMPY displayed a ratio of 1.8–2.0, between30 and 50 min after an i.v. injection [1181]. Its devel-opment was terminated.

The development of amyloid binding PETligands (Aventis), fluoropegylated indolyl-phenyl-acetylenes (Avid Radio-pharmaceuticals), 11C-6-Me-BTA (University of Pittsburgh), and of 18F-SMIBR-W372 (Siemens Medical Solutions Molecular Imag-ing) was terminated.

Inhibitor of serum amyloid P component binding

The normal plasma protein serum amyloid P com-ponent (SAP) binds to fibrils in all types of A� depositsand contributes to the pathogenesis of amyloidosis[1245–1247].

Ro-63-8695 (CPHPC; Pentraxin Therapeutics, Lon-don under license from Roche) (Fig. 14) is acompetitive inhibitor of serum amyloid P componentbinding to amyloid fibrils (IC50 = 0.9 �M). First resultsfrom in vivo mouse and human studies have been com-municated [1245, 1248–1250]. In addition, PentraxinTherapeutics and GSK are investigating a combinationof CPHPC with a humanized antibody for the potentialtreatment of amyloidosis (Thomson Reuters Pharma,update of March 22, 2012).

Vaccines against amyloid-β

AN-1792 (AIP-001; Elan and American HomeProducts, later Wyeth, now Pfizer) was the first vac-cine against AD in clinical trials. The antigen wassynthetic A�42, which was administered with QS-21from the Stimulon family of saponins purified from thebark of Quillaja saponaria as adjuvant [1251, 1252].The multicenter double-blind Phase IIa study in theUS and Europe involving 375 patients with mild-to-moderate AD started in the third quarter of 2001.Treatment of patients with AN-1792 plus adjuvantand the adjuvant alone as placebo was in a ratio of4 : 1. 18 patients developed meningoencephalitis prob-ably due to an inappropriate T cell activation and/orthe proinflammatory Th1 type of adjuvant. The studywas discontinued in March 2002 [1253]. Neverthe-less, many important data could be collected. From129 patients immunized with the drug, 25 were clas-sified as antibody responders after 4.5 years. Thesepatients showed significantly slower decline on the

Page 40: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

40 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 14. PET and SPECT ligands.

disability assessment for dementia and a significantdifference on a dependence scale compared to placebo-treated patients. Antibody responders also showed lessof a decline in a memory test. The scientific harvestof this single clinical trial is listed in chronologicalorder [1254–1286] (Thomson Reuters Pharma, updateof August 24, 2012).

Affitope AD-02 (Affiris, Vienna AT and licenseeGlaxoSmithKline Biologicals) is a six amino acid pep-tide vaccine targeting the N-terminus of A� only, whenit is free. The adjuvant is aluminum hydroxide. Theantibody response is focused exclusively on A� anddid not show crossreactivity to A�PP [1287, 1288]. AEuropean Phase II clinical trial in 420 patients in Aus-tria, Germany, France, the Czech Republic, Slovakia,and Croatia started in April 2010 (Thomson ReutersPharma, update of August 1, 2012).

CAD-106 (Cytos Biotechnology, Zurich and Novar-tis) is an A�1-6 peptide linked to a Q� virus-likeparticle for the s.c. treatment of patients with AD.One Phase IIa trial was initiated in July 2008 (n = 27),and a second Phase IIa trial was started in October2008 (n = 30). The safety, tolerability, and antibodyresponse of active A� immunotherapy with CAD-106 in patients with AD was published [1289]. Forcomments, see [1290, 1291]. It was found that 80%

of the patients involved in the trial developed theirown protective antibodies against A� without suffer-ing any side-effects over the three years of the study.The researchers believe that the CAD106 vaccine is atolerable treatment for patients with mild-to-moderateAD. In March 2010, a randomized, placebo-controlled,multicenter, Phase II trial began in patients (n = 120)in the US, Canada, and Europe with mild AD [1276,1292, 1293]. Preclinical results in transgenic mice werepresented [1294] (Thomson Reuters Pharma, update ofJuly 26, 2012).

Vanutide cridificar (ACC-001, PF-05236806;Janssen Alzheimer Immunotherapy, Dublin, Ire-land acquiring Elan’s Alzheimer’s ImmunotherapyProgram and Pfizer) is a peptide fragment of A� conju-gated to the mutated diphtheria toxin protein CRM197.The adjuvant is QS-21. In May 2007, a randomized,multicenter, double-blind, placebo-controlled, parallelassignment, safety/tolerability/immunogenicity PhaseII trial in patients with mild-to-moderate AD (n = 56)began [1295]. In July 2009, Pfizer began a multi-center, randomized, unblinded, QS-21-adjuvanted,long-term, Phase II trial of vanutide cridificar (3, 10,and 30 microg, im) in patients (n = 160) in the US.At that time the estimated study completion date wasSeptember 2014. In August 2009, an additional study

Page 41: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 41

was initiated in Japanese subjects (n = 32) [1276](Thomson Reuters Pharma, update of August 13,2012).

ACI-24 (AC Immune, Lausanne, Switzerland) is anA�1-15 peptide to which on both ends two lysineswere attached, which are tetrapalmitoylated on the�-nitrogens. The antigen is embedded in a lipo-some membrane. Adjuvant is a mixture of the lipidsDMPC, DMPG, cholesterol, and MPLA in a ratioof 9 : 1 : 7 : 0.06, respectively [1296, 1297]. The drugrestored memory defects of transgenic AD mice.The IgF subclasses of the antibodies generated fromthe vaccine were mostly IgG2b indicating a non-inflammatory Th2 isotype. CD and NMR revealedpredominantly �-sheet conformation. The drug iscurrently evaluated in a Phase I/II clinical trial inDenmark, Finland, and Sweden (Thomson ReutersPharma, update of June 19, 2012).

Affitope-AD-03 (Affiris, Vienna, Austria andlicensee GlaxoSmithKline Biologicals) entered aPhase I clinical trial in October 2010. In Novem-ber 2011, the study was completed (Thomson ReutersPharma, update of April 4, 2012).

UB-311 (United Biomedical, Hauppauge, NY) isan intramuscularly administered vaccine targeting N-terminal amino acids 1-14 of A� in Phase I clinicaltrials in Taiwan in patients with mild-to-moderate AD(n = 18) [1298] (Thomson Reuters Pharma, update ofJune 25, 2012).

V-950 (Merck, Whitehouse Station, NJ) is anN-terminal A� peptide conjugated to an aluminum-containing adjuvant with or without ISCOMATRIX(an adjuvant containing saponin, cholesterol, andphospholipids) [1299]. Mild-to-moderate AD patients(n = 124) received six injections/year of increasingdoses of either placebo or V-950 in the presence orabsence of varying concentrations of ISCOMATRIXin Phase I clinical trials in the US and Sweden overa four year period [1276] (Thomson Reuters Pharma,update of February 24, 2012).

There are numerous vaccine preparations in preclin-ical evaluation (in alphabetical order):

ABvac40 and ABvac42 (Araclon Biotech,Zaragoza, Spain) are active therapeutic antibodyvaccines awaiting approval for clinical trials [1741](Thomson Reuters Pharma, update of June 25, 2012).

ADepVac (Ichor Medical Systems, San Diego, CAin collaboration with the University of California atIrvine and the Institute for Molecular Medicine) is aDNA vaccine using the TriGrid electroporation tech-nology [1300] (Thomson Reuters Pharma, update ofJuly 13, 2012).

ALZ-101 (Alzinova, Goteborg, Sweden, a spin-offof MIVAC Development) is a specific oligomer-directed therapeutic vaccine (Thomson ReutersPharma, update of July 18, 2012).

ALZ-301 (Alzinova, Goteborg, Sweden, a spin-offof MIVAC Development) is an A�40 oligomer tar-geted replacement therapy (Thomson Reuters Pharma,update of July 18, 2012).

Alzheimer’s disease vaccine (VLP Biotech, SanDiego CA) is a vaccine that activates antibodies againstA� protein (Thomson Reuters Pharma, update of May3, 2012).

Amyloid-� 3-10 DNA vaccination (China Medi-cal University, Shenyang) suggested a potential newtreatment for AD [1301–1305].

Active immunization with Ankyrin G, a neuronalcytoskeletal protein, with Freund’s adjuvant completeof arcA� mice reduced A� pathology [1306].

BAN-2203 (BioArctic Neuroscience, Stockholm,Sweden) is an immunotherapeutic vaccine targetingA� protofibrils (Thomson Reuters Pharma, update ofJanuary 7, 2011).

BBS-1 BACE inhibitor mAb vaccine (Nasvax,Ness Ziona, IL under license from Ramot at Tel AvivUniversity) is based on a lead mAb candidate blocking�-site-1, which inhibited the ability of BACE to cleaveA�PP (Thomson Reuters Pharma, update of July 30,2012).

C12 (Pharma Bio, Moscow, Russia) is a programof therapeutic vaccines comprised of synthetic peptidefragments of the �7 nicotinic acetylcholine receptorused to generate antibodies that block A� binding toneurons (Thomson Reuters Pharma, update of March29, 2012).

EB-101 (Atlas Pharmaceuticals, Sunnyvale, CA)is a synthetic human A�42/sphingosine-1-phosphate/liposome vaccine (Thomson Reuters Pharma, updateof July 27, 2012).

A T cell-based vaccination with Glatimer acetateof AD double-transgenic (A�PP/PS1) mice resultedin decreased plaque formation and induction ofneurogenesis. Dendritic-like (CD11c) cells producedinsulin-like growth factor 1 [1307].

MER-5101 (Mercia Pharma, Scarsdale, NY) is avaccine comprised of an A� peptide conjugate coupledto an immunogenic carrier protein and the com-pany’s Th2-biased adjuvant MAS-1 (Thomson ReutersPharma, update of January 27, 2012).

Mimovax (MV-01; Affiris, Vienna AT) is anAFFITOPE-based vaccine targeting truncated andmodified forms of A� (Thomson Reuters Pharma,update of April 2, 2012).

Page 42: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

42 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

NU-700 (Nuron Biotech, Exton, PA under licensefrom Vitruvian BioMedical under license from theUniversity of Texas Southwestern Medical Center) isan adjuvant free, gene-base (DNA) A�42 vaccine forthe potential treatment or prevention of AD (ThomsonReuters Pharma, update of May 22, 2012).

Recombinant adenovirus vector vaccine (Vaxin,Birmingham, AL in collaboration with AnC Bio,South Korea) elicited an immune response against A�using Crucell’s PER.C6 technology for the potentialintranasal treatment of AD (Thomson Reuters Pharma,update of May 2, 2012).

RV-03 (Intellect Neurosciences, New York, NY) isa peptide vaccine developed using the RECALL-VAXtechnology targeting both A� and a truncated delta tauprotein (Thomson Reuters Pharma, update of August21, 2012).

SeV-amyloid beta RNA vaccine (DNAVEC,Tsukuba, Japan) is an intranasal vaccine comprisinga Sendai virus vector encoding the A� gene [1308](Thomson Reuters Pharma, update of September 21,2011).

Excellent papers on A� vaccines from universitieswere presented (in chronological order) 2000: [1309,1310]; 2001: [1311]; 2004: A�1-42 gene vaccination[1312]; 2005: a synthetic universal Th cell pan HLADR epitope, pan HLA DR-binding peptide (PADRE),in which the PADRE-A�1-15 sequence lacks the T cellepitope of A� [1313]; 2006: an A� derivative in alumadjuvant [1314], an intranasal dendrimeric A�1-15 vac-cine [1315–1319]; 2007: an oral vaccination usinga recombinant adeno-associated viral vector carry-ing A� cDNA (AAV/A�) [1320], a transcutaneousadministration of aggregated A�1-42 plus the adjuvantcholera toxin [1321], a mannan-A�28 conjugate [1322,1323]; 2008: a DNA epitope vaccine that expresses epi-topes of A�42 [1324]; 2009: K6 A�1-30-NH2 in alumadjuvant [1325], an A�1-33-MAP peptide to markedlyreduce intracellular A� deposits [1326], K6A�1-30 forimmunizations of old primates [1327], A�1-42 vac-cinations also reducing mouse tau pathology [1328];2010: an oral vaccination with GFP-A� [1329], immu-nization of aged beagle dogs with aggregated A�1-42formulated in an alum adjuvant [1330], active immu-nization with A�1-42 emulsified in CFA containingMycobacterium tuberulosis extract [1331], loweringof A� plaque burden by the SDPM1 peptide, a 20amino acid peptide bound by cysteines that bindstetramer forms of A�1-40 and A�1-42 amyloids [1332];2011: two novel anti-A� vaccines consisting of viruslike particles [1333], the identification of the short-est A�-peptide generating specific antibodies [1334];

2012: preventive immunization of aged primates[1335].

Excellent reviews on A� vaccines have been pro-vided (in chronological order) 2001: [1336], [1337],2002: [1257, 1338–1341], 2003: [1342]), 2004:[1343]), 2005: [1344–1346], 2006: [1347–1349],[1350], 2007: [1351–1355], 2008: [1356–1359],2009: [1282, 1360–1363], 2010: [1364–1370], 2011:[1371], [1372–1374], and 2012: [23].

A clinical review of active and passive immunother-apeutic approaches in AD targeting A� was presented[1375, 1376].

The development of A� retroparticles (Universityof Heidelberg and Novartis [1377]), Abloid (Virion-ics), ACC-002 (Elan), AFFITOPE AD-01 (Affiris),Alzheimer’s vaccine (Lundbeck under license fromPharmexa), Alzheimer’s vaccine (Mindset BioPhar-maceuticals), Alzheimer’s disease vaccines (Univer-sity of South Florida and University of New Mexico),anti-amyloid-� vaccines (Wyeth, now Pfizer andJanssen Alzheimer Immunotherapy), DNA vaccine(University of Texas, Southwestern Medical Cen-ter), immunotherapy vaccine (Prana Biotechnology),KNX-Vaccine3A (Kinexis under license from the Uni-versity of California at Irvine), PEVIPRO (PevionBiotech [1378]), PX-106 (Lundbeck/Pharmexa), andRV-01 (Intellect Neurosciences) was terminated.

Antibodies against amyloid-β

Bapineuzumab (AAB-001; Janssen AlzheimerImmunotherapy, Dublin, Ireland following the acqui-sition of Elan’s Alzheimer’s Immunotherapy Programand Pfizer) is a humanized monoclonal antibody raisedagainst eight amino acids in the N-terminus of A� forthe potential i.v. and s.c. treatment of AD. A Phase IIIclinical study started in December 2007. The Phase IIIprogram included two trials in ApoE4-positive patients(n = 800 in the US and EU) and two in ApoE4-negativepatients (n = 1,250 in the US and EU) for 18 monthseach and three extension studies for both ApoE4 andnon-ApoE4 arms with three doses, 0.5, 1, and 2 mg/kg.The highest dose had to be abandoned due to vasogenicedema safety concerns. One extension study started inJuly 2009 (n = 1,350), two additional extension studiesin December 2009 in the EU and Australia (n = 800 and1,000, respectively). Clinical results from the PhaseI and Phase II studies were reported [1276, 1361,1379–1382] as was a 11C-PiB PET assessment [1383]and the effects on CSF biomarkers [1384]. Reviews onbapineuzumab were published (in chronological order)[23, 1385–1392]. For the scientific basis see the Nature

Page 43: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 43

Medicine paper [1393]. The pharmacokinetic profile of125I-labeled 3D6, the mouse parent antibody of bap-ineuzumab, in transgenic mice was reported [1742].

In July 2012, Pfizer reported that the ’302 study inApoE4 carriers did not meet its primary endpoints.Pfizer, Johnson & Johnson, and Elan announced thatdevelopment of i.v. bapineuzumab was terminated(Thomson Reuters Pharma, update of September 18,2012).

Solanezumab (LY-20622430; Lilly) is a mid-domain humanized monoclonal antibody selective forsoluble A�. Three Phase III studies were initiatedin patients with mild-to-moderate AD (n = 1,000) inMay 2009, patients received 400 mg of solanezumabor placebo i.v. every 4 weeks for 80 weeks.First results were disclosed (in chronological order)[1276, 1394–1398]. Safety and biomarker effects ofsolanezumab in patients with AD was communicated[1399]. Solanezumab failed to meet the cognitive andfunctional primary endpoints of two Phase III AD tri-als. But it showed a significant reduction in cognitivedecline in patients with mild AD. The open-labelEXPEDITION-EXT extension trial, which is fullyenrolled, will continue as planned (Thomson ReutersPharma, update of September 7, 2012).

Gantenerumab (R-1450; Roche under license fromMorphoSys) is a human anti-A� monoclonal anti-body. A Phase II clinical trial in Canada (n = 360)was initiated in January 2011. Prodromal AD patients(NCT01224106) are to receive 105 or 225 mg admin-istered by s.c. injection every 4 weeks for 104 weeks toevaluate effects on cognition and functioning, safety,and pharmacokinetics. In June 2012, the trial wasexpanded to a Phase II/III trial. The trial size will beincreased from 360 to 770 participants. The mecha-nism of A� removal was described [1400, 1401]. Anexpert opinion was provided [1402] (Thomson ReutersPharma, update of September 5, 2012).

Crenezumab (MABT-5102A; RG-7412; Genen-tech under license from AC Immune, Lausanne,Switzerland) is an anti-A� humanized mono-clonal antibody as a conformation-specific, passiveimmunotherapy for the potential i.v or s.c. treatment ofAD. A Phase II clinical trial (n = 372; NCT01397578)was initiated in April 2011. The first Alzheimer’s Pre-vention Initiative will study the effects of crenezumabin 300 people from a large family in Columbia witha rare genetic mutation that typically triggers ADsymptoms around age of 45. It is a collaborationbetween the National Institutes of Health (NIH), Ban-ner’s Alzheimer’s Institute, and Genentech over fiveyears starting in 2013. Data and findings will be shared

publicly after the study completion. The scientific basisfor the selection of crenezumab was published recently[1403] (Thomson Reuters Pharma, update of Septem-ber 5, 2012).

GSK-933776A (GSK) is a monoclonal antibodyfor the i.v. treatment of AD and age-related maculardegeneration. A Phase II trial (n = 162) of age-relatedmacular degeneration started in the US in August2011 (Thomson Reuters Pharma, update of August 24,2012).

The topic intravenous immunoglobulins as a treat-ment for AD, the rationale and current evidence wasdescribed in reviews [1404, 1405, 1743].

Immune globulin intravenous human (IVIg;launched as Gammagard in the US and as Kiovigin Europe; Baxter International, Deerfield, IL) isa highly purified solvent/detergent-treated, sterile,freeze-dried preparation derived from human plasmafor the treatment of primary immunodeficiency dis-ease. This pool of human immunoglobulins obtainedfrom healthy donors contains naturally occurringanti-A� antibodies. In a Phase II trial in 24 patients,the eight-person placebo group worsened, whereasthe 16 treated patients improved moderately on bothcognitive and quality-of-life measures over the first6 months [1406–1408]. A Phase III clinical trial inAD patients enrolling 390 patients was initiated inSeptember 2008. Trial completion is expected by theend of 2012 (NCT00818662, NCT00299988) [1276].The first report of long-term (three-year) stabilizationof AD symptoms with IVIG (Gammagard, Baxter),including no decline on measures of cognition, mem-ory, daily functioning, and mood, was reported inJuly 2012 at the Alzheimer’s Association InternationalConference in Vancouver. Sales in 2011 amounted to1,541 million USD. For preclinical data, see [1409,1410] (Thomson Reuters Pharma, update of October3, 2012).

Octagam (Octapharma, Lachen, Switzerland) isa 10% liquid intravenous immunoglobulin launchedfor the treatment of primary immunodeficiency. ByDecember 2008, a double-blind, randomized, PhaseII clinical trial (NCT 00812565) in 58 patients withAD had been initiated in the US. Clinical data werepresented at the AAIC in Paris in July 2011 (ThomsonReuters Pharma, update of April 3, 2012).

Flebogamma DIF 10% (or 5%) (Grifols SA,Barcelona) is a 10% (or 5%) double inactivated andfiltered immunoglobulin therapy for the treatment ofprimary immunodeficiency. Data were presented on asingle-center, open-label, pilot study on the use of Fle-bogamma DIF 0.5 g/kg i.v., q2w for 6 months in AD

Page 44: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

44 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

patients (n = 4) (Thomson Reuters Pharma, update ofMarch 23, 2012).

BAN-2401 (Eisai under license from BioarcticNeuroscience, Stockholm, Sweden) is a humanizedmonoclonal antibody that targets the large solubleamyloid product (protofibril A�). A Phase I trial in 80patients with mild-to-moderate AD began in Septem-ber 2010. Eisai plans to co-administer BAN-2401 withdonepezil and memantine (Thomson Reuters Pharma,update of July 19, 2012).

NI-101 (BIIB-037, BART; Biogen Idec, Weston,MA under license from Neuroimmune Therapeutics,Zurich Switzerland) is a recombinant chimeric humanIgG1 mAb targeted against A�. A Phase I clinical trial(n = 40) in patients with mild-to-moderate AD startedin the US in July 2011 (Thomson Reuters Pharma,update of September 24, 2012).

PF-05236812 (AAB-003; Janssen Alzheimer Imm-unotherapy and Pfizer) is a humanized monoclonalantibody targeted against A�. In October 2010, a ran-domized, double-blind, placebo-controlled, adaptive,Phase I trial was initiated in patients with mild-to-moderate AD (n = 80) in the US [1411] (ThomsonReuters Pharma, update of August 13, 2012).

RN6G (Rinat Neuroscience Corp., New York, nowPfizer) is an anti-A� antibody for the potential i.v.treatment of age-related macular degeneration in PhaseI trial (n = 45) since April 2009 in the US. In April2012, a randomized, double-blind, placebo-controlled,Phase II study was planned in patients with geographicatrophy secondary to age-related macular degeneration(expected n = 276) to assess the safety and efficacy ofRN6G [1412] (Thomson Reuters Pharma, update ofSeptember 11, 2012).

SAR-228810 (sanofi) is an anti-protofibrillar A�monoclonal antibody. A randomized, double-blind,placebo-controlled, Phase I trial in patients with mild-to-moderate AD (n = 48) was initiated in Sweden inJanuary 2012. The study is scheduled to complete inDecember 2013 (Thomson Reuters Pharma, update ofJuly 27, 2012).

There are currently many antibodies for the poten-tial treatment of AD and closely related diseases inpreclinical evaluation (in alphabetical order):

4E10 (Prana Biotechnology, Parkville, Australia) isa monoclonal antibody targeting a proprietary patho-logical A� target epitope (Thomson Reuters Pharma,update of November 2, 2011).

6F6 (GSK) is an anti-A� monoclonal antibody forthe potential treatment of age-related macular degen-eration (Thomson Reuters Pharma, update of June 9,2011).

9D5 (University of Gottingen, MBM Science-Bridge) targets pyro-Glu-A� peptide oligomers for thepotential diagnosis and treatment of AD [1413] (Thom-son Reuters Pharma, update of August 7, 2011).

A-887755 (Abbott Laboratories) is an A�,oligomer-selective, mouse monoclonal antibodygenerated using a homogenous, synthetic A�20-42oligomer peptide (Thomson Reuters Pharma, updateof February 27, 2012).

A-992401 (Abbott Laboratories) is a mAb target-ing the receptor for advanced glycation-end-products(Thomson Reuters Pharma, update of December 1,2010).

Ab40-4-42 (Ilsung Pharmaceutical, Seoul) is abiological therapeutic, which acted by inhibitingA� aggregation and cytotoxicity (Thomson ReutersPharma, update of May 14, 2012).

ACU-193 (Acumen Pharmaceuticals, LivermoreCA) is a monoclonal antibody against amyloid-deriveddiffusible ligands (Thomson Reuters Pharma, updateof August 16, 2012).

AD-0802 (Bioarctic Neuroscience, Stockholm,Sweden) is a humanized mAb with affinity for bindingto A� protofibrils. It is a potential follow-up compoundto BAN-2401 (vide supra) (Thomson Reuters Pharma,update of January 7, 2011).

AGT-160 (ArmaGen Technologies, Santa Monica,CA) is a recombinant IgG fusion protein formed by thefusion of a single chain Fv antibody against A� plaqueformation to the company’s human insulin receptor-targeting monoclonal antibody Trojan horse for thetransport across the blood-brain barrier. A 1 mg/kg i.v.dose of AGT-160 was administered twice a week for12 weeks to transgenic AD mice resulting in a 40%decrease in the brain concentration of A� (ThomsonReuters Pharma, update of January 23, 2012). See alsoPart 1. Chapter 1.16. Drugs interacting with the insulinreceptor.

ALZ-201 (Alzinova, Goteborg, Sweden, a spin-off of MIVAC Development) is an oligomer-specificA� monoclonal antibody (Thomson Reuters Pharma,update of July 20, 2012).

Amyloid precursor protein C-terminalfragment-targeted monoclonal antibodies (EcolePolytechnique Federale de Lausanne, EPFL) isa monoclonal antibody targeted to the 99 aminoacid C-terminal fragment of A�PP (A�PP-C99)(Thomson Reuters Pharma, update of March 31,2011).

Anti-amyloid beta antibodies (Kyowa HakkoKirin under license from Immunas Pharma, OncoTher-apy Science Inc.) are antibodies targeting A�

Page 45: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 45

oligomers (Thomson Reuters Pharma, update ofAugust 6, 2012).

Brain-targeted BACE1 antibody (Genentech,Roche Holding) is a bi-specific antibody againstBACE1 and the transferrin receptor to allow tran-scytosis across the blood-brain barrier [1414, 1415](Thomson Reuters Pharma, update of May 14, 2012).This technique was pioneered by Prof. William M.Pardridge of UCLA [1416–1422].

CPHPC+antibody (Pentraxin, London and GSK) isa combination of CPHPC, which lowers blood levels ofserum amyloid P component (see Chapter 16.2) and ahumanized antibody for the treatment of amyloidosis[1249] (Thomson Reuters Pharma, update of March22, 2012).

DLX-212 (Delenex Therapeutics, Zurich, Switzer-land) is an anti-A� antibody fragment (ThomsonReuters Pharma, update of March 29, 2012).

Fully human monoclonal antibodies (Intrexon,Blacksburg VI after its acquisition of Immunologix)are targeting A� plaques for the potential treatmentof AD (Thomson Reuters Pharma, update of June 13,2012).

IN-N01 (Intellect Neurosciences under license fromImmuno-Biological Laboratories, IBL, New York,NY) is an antibody drug conjugate consisting of aA�-specific humanized mAb employing its ANTI-SENILIN technology, for the potential treatment ofAD, glaucoma, age-related macular degeneration andtraumatic brain injury. In September 2012, Intellectplanned to conduct studies in combination with taumAbs TOC-1and TauC3 (Thomson Reuters Pharma,update of September 20, 2012).

IN-N01-OX2 (Intellect Neurosciences, New York,NY) is a humanized IgG4 monoclonal antibody conju-gated to melatonin developed using its CONJUMAB-A technology for the potential treatment of age-relatedmacular degeneration and Alzheimer’s disease (Thom-son Reuters Pharma, update of October 2, 2012).

Lpathomab (LT-3015; Lpath Inc., San Diego,CA) is a systemic formulation of a humanized anti-lysophosphatidic acid monoclonal antibody for thepotential treatment of fibrosis, ocular inflammation,and CNS disorders including AD [1423] (ThomsonReuters Pharma, update of August 28, 2012).

MDT-2007 (Medronic, Minneapolis, MN) is ahumanized anti-A� monoclonal antibody, the human-ized version of the mouse mAb 6E10 (ThomsonReuters Pharma, update of January 6, 2012).

Nanobody therapeutics (Ablynx, Gent Belgiumand licensee Boehringer Ingelheim) are naturally-occurring single chain antibodies of Camelidae. In

April 2012, a CTA was submitted to European regula-tory authorities for a Phase I study (Thomson ReutersPharma, update of August 23, 2012).

NEOD-001 (Onclave Therapeutics, Dublin, Irelandand Neotope Biosciences, South San Francisco, CAand Elan) is a proprietary monoclonal antibody forthe potential treatment of AL amyloidosis. In February2012, the drug was awarded Orphan status by the FDAfor AA amyloidosis and AL amyloidosis (ThomsonReuters Pharma, update of June 27, 2012).

STC-103 (STC Biologics, Cambridge, MA) is anovel Fc fusion protein therapeutic for the potentialtreatment of AD (Thomson Reuters Pharma, update ofAugust 10, 2012).

Excellent papers on A� antibodies from universitiesand basic research from companies were presented (inchronological order) 1996: [1424], 1997: [1425]; 2000:[1393]; 2005: [1426, 1427], 2006: [1428, 1429], 2007:[1430–1441], 2008: [1442–1447], 2009: [1448–1454],2010: [1366, 1455–1460], 2011: [1461–1465], 2012:[1466, 1467, 1744].

Excellent reviews were provided (in chronologi-cal order) 2006: [1350], 2007: [1468–1470], 2008:[1356, 1358, 1471, 1472], 2009: [1360, 1361,1473–1475], 2010: [1369]; 2011: [23, 1371, 1388,1389, 1476–1478], 2012: [1479]. The challengeof adverse effects of immunotherapy for AD wasaddressed [1480].

A clinical review of active and passive immunother-apeutic approaches in AD targeting A� was presented[1375, 1376].

The development of 11A1 (a monoclonal anti-body against the toxic conformer of A�42; TokyoMetropolitan Institute of Gerontology, Kyoto Uni-versity and Immuno-Biological Laboratories), A-11(A� deposition-inhibiting antibody; Kinexis underlicense from the University of California), AAB-002 (Janssen Alzheimer Immunotherapy and Pfizer),ABP-102 (Abiogen Pharma; a catalytic monoclonalantibody, an abzyme), ACU-5A5, ACU-0101979and huC091 (Acumen Pharmaceuticals), anti-Abetamonoclonal antibodies (Mapp Biopharmaceutical incollaboration with Johns Hopkins University), theanti-amyloid beta antibody therapy targeting spe-cific forms of the A� parenchymal plaque (sanofi underlicense from Rockefeller University), anticalin (Pieris,an antibody against residues 16-26 of A�), AZD-3102 (AstraZeneca), bapineuzumab (AAB-001;Janssen Alzheimer Immuno-therapy and Pfizer), betaamyloid deposition-inhibiting antibodies (amyloi-dosis, Kinexis), beta amyloid-targeting antibodies(Mindset BioPharmaceuticals), encapsulated scFv-

Page 46: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

46 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

beta 1-expressing cells targeting the EFRH tetrapep-tide region of A� (scFv-beta 1; Novartis in collabo-ration with the Institute for Research in Biomedicineand the Ecole Polytechnique Federale de Lau-sanne), ESBA-212 (ESBA Tech, Nestle, a humanizedsingle-chain antibody directed against A�), KNX-Monoclonal204 and KNX-Monoclonal205 (Kinexisunder license from the University of California atIrvine), m266 (an antibody specific for the centraldomain of A�; Lilly [1481, 1482]), and ponezumab(PF-4360365; RN-1219; RI-1219; Rinat Neuroscienceand Pfizer) [1276, 1483–1485] a humanized mono-clonal antibody specific for the C-terminus of A�40,was terminated.

DRUGS INTERACTING WITH TAU

A hallmark of the AD brain is the presenceof inclusions within neurons that are comprised offibrils formed from hyperphosphorylated microtubule-stabilizing protein tau (paired helical filaments andneurofibrillary tangles [1486–1489]. The formationof misfolded multimeric tau species is believed tocontribute to the progressive neuron loss and cogni-tive impairments of AD [1490]. Tau suppression ina neurodegenerative mouse model improved memoryfunction [1491]. The precise composition of the neu-rotoxic species Tau-P* is not defined yet [1486]. Thelevels of early multimeric tau-aggregates that precededthe neurofibrillary tangles were found to correlate withmemory deficits [1492]. The sites of phosphoryla-tion and the involved kinases were compiled [1493,1494]. The tau phosphorylation pathway genes weredescribed [1495]. Although A� plaques may play akey role in AD pathogenesis the severity of cognitiveimpairment correlated best with the burden of neu-rofibrillary tangles described in an exhaustive reviewof the literature [1496, 956]. A� and tau influence eachother in the mediation of toxicity. A� formation inA�PP transgenic mice caused hyperphosphorylationof tau, whereas there was no overt A� plaque pathol-ogy in tau transgenic mice. Immunization against A�in triple transgenic mice resulted in reduced levelsof hyperphosphorylated tau. Tau reduction preventedA� induced defects in the axonal transport of mito-chondria. Combination of these data led to the twostep tau axis hypothesis of AD [1497–1499]. First,postsynaptic toxicity of A� is tau-dependent, becausetau interacts with FYN [1500], which is localized tothe dendritic compartment, where it phosphorylatesthe NMDA receptor subunit 2B, mediates their inter-

action with the postsynaptic density protein 95, aninteraction required for A� toxicity and second, A�triggers progressively increased phosphorylation of taucompromising the binding of tau to microtubules lead-ing to increasing dendritic tau levels [1501]. For avery instructive figure depicting these interactions, see[1502]. The synaptic accumulation of hyperphospho-rylated tau oligomers is associated with dysfunction ofthe ubiquitin-proteasome system [1503].

A cascade of biomarkers was proposed: A� precedestau-mediated neuronal injury; A�- and tau-mediatedabnormalities precede neuroimaging biomarkers suchas changes in hippocampal or ventricular volumetrymeasured by MRI; and all of these precede cog-nitive change marked by progressive deteriorationin episodic memory and other cognitive domainssuch as executive functions abilities [1504–1506].In a longitudinal study in 172 participants (AD:n = 41; MCI: n = 85; and normal controls: n = 46),the temporal relations among the four classes ofbiomarkers were examined. Results indicated that CSFA� effects on cognition change were substantiallyattenuated by CSF tau and measures of brain structureand function and CSF tau effects on cognitive changewere attenuated by neuroimaging variables. Contraryto hypotheses, CSF A� and CSF tau were observed tohave independent effects on neuroimaging and CSF tauhad a direct effect on baseline cognition independentof brain structure and function [1507].

Small molecules preventing tau aggregation

TRx-0014 (methylthioninium chloride, methyleneblue, Rember; TauRx Therapeutics, Singapore, a spin-out from the University of Aberdeen) (Fig. 15) is atau protein aggregation inhibitor. In August 2004, aplacebo-controlled, dose-ranging Phase II trial wasinitiated in subjects (n = 275) with AD associateddementia in the UK. The doses were 30, 60, or 100 mgtid. In July 2007, a Phase II open-label continuationstudy of two doses of 30 or 60 mg tid gelatin cap-sules started for 52 weeks. The drug reduced cognitivedecline by 81% over 1 year and no significant lossof cognitive function was seen over 19 months [1508,1509]. In November 2010, the EC granted the drugOrphan status for frontotemporal dementia, progres-sive non-fluent aphasia and progressive supranuclearpalsy. In February 2012, an open-label trial in patientswith frontotemporal dementia was ongoing. For thepreclinical characterization, see [1510–1516]. Onlywhen levels of soluble tau protein were concomitantly

Page 47: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 47

reduced by a very high concentration of methylene bluecognitive improvement was observed in a mouse modelof human tauopathy [1517]. Methylene blue also inhib-ited the aggregation of A� [1518, 1519] (ThomsonReuters Pharma, update of September 10, 2012).

LMT-X (TauRx Therapeutics, Singapore) is apotential follow-up compound and prodrug of Rember.In September 2012, a Phase III study for frontotem-poral dementia began in 180 subjects for 12 months.In October 2012 two global phase III studies wereinitiated for AD. The first study will involve 833patients with mild to moderate Alzheimer’s diseaseover 12 months, while the second study will include500 patients with mild Alzheimer’s disease over 18months. The clinical trials will be conducted in paralleland on a global basis including sites in Australia, Bel-gium, Canada, Finland, Germany, Italy, Russia, Spain,Netherlands, Singapore, Malaysia, Taiwan, US and UK(Thomson Reuters Pharma, update of November 21,2012). The structure was not communicated.

PBT-2 (Prana Biotechnology, Parkville, Australia)(see Fig. 9) is an oral zinc ionophore metal-proteinattenuating compound that reduced levels of solu-ble A� and tau hyperphosphorylation in a Phase IIastudy in AD patients. (Thomson Reuters Pharma,update of October 2, 2012) See also Chapter 12. MetalChelators.

Tideglusib (NP-12, NP-031112; Nypta; Noscira,previously known as Neuropharma, Madrid) (Fig. 15)is a GSK-3� inhibitor in Phase II clinical trials for AD.Results from the 309-patient study were expected byOctober 2012. First positive results of a clinical pilotstudy were reported [1520] (Thomson Reuters Pharma,update of July 23, 2012). See also Part 2, Chapter 2.16.Drugs interacting with GSK-3�.

BMS-241027 (Bristol-Myers Squibb) is a smallmolecule microtubule stabilizer aimed at preventingthe production of abnormal tau protein. A randomized,double-blind, placebo-controlled Phase I study was ini-tiated in patients with mild AD (expected n = 100) inthe US and in Europe in March 2012 (Thomson ReutersPharma, update of August 6, 2012). The structure wasnot communicated.

PP2A stimulator (Se-015; Ve-015; Velacor Ther-apeutics, Victoria, Australia) is a selenium derivativeacting as protein phosphatase 2A (PP2A) stimulatorresulting in dephosphorylation of Akt kinase and tau.By February 2011, a Phase I trial had been completedand the company received the approval to initiate aPhase IIa trial in AD patients in Australia (ThomsonReuters Pharma, update of February 10, 2011). Thestructure has not been communicated.

There are many compounds inhibiting tau aggre-gation in preclinical evaluation (in alphabeticalorder):

A�/tau protein aggregation inhibitors (CNRS,FIST SA, Paris, France) are investigated for the poten-tial treatment of AD (Thomson Reuters Pharma, updateof August 24, 2012). The structures of the compoundswere not communicated.

Astemizole and Lansoprazole have been found toselectively interact with tau polymers [1521].

Berberine attenuated tau hyperphosphorylation inHEK293 cells [1745].

Bezafibrate (Bezalip, Bezatol; BoehringerMannheim, now Roche, co-marketing with KisseiPharmaceuticals, launched 1977) (Fig. 15), a drugfor the treatment of hyperlipidemia, is a pan-PPARagonist, which improved behavioral deficits and taupathology in P301S mice exerting a preventive effect[1522].

BLV-0703 (BLV-200703; Bioalvo, Lisbon, Por-tugal) (Fig. 15) is a tau aggregation inhibitor inpreclinical development (Thomson Reuters Pharma,update of May 9, 2011).

Epothilone D improved microtubule density, axonalintegrity and cognition in a transgenic mouse model oftauopathy [1523].

Fulvic acid inhibited aggregation and promoted thedisassembly of tau fibrils associated with AD [639].

Insulin intranasal ameliorated tau hyperphospho-rylation in a rat model of type 2 diabetes [1524].

L-3-n-butylphthalide reduced tau phosphorylationand improved cognitive deficits in A�PP/PS1-Alzheimer’s transgenic mice [1746].

NBB BSc3504 (TU Darmstadt and Max-Planck-Unit for Structural Molecular Biology Hamburg)(Fig. 15) is a N ′-benzylidene-benzohydrazideshowing remarkable tau aggregation inhibition(IC50 > 0.97 �M) and paired helical filamentdepolymerization (DC50 > 1.14 �M), but pooraffinity towards A�1-42 fibrils (IC50 > 33 �M) [1525].

NC-11813 (Eli Lilly and Nymirum, Ann Arbor, MI)is an inhibitor of tau exon 10 splicing, which stabilizedthe tau splice regulatory element (Thomson ReutersPharma, update of August 10, 2012). The structure wasnot communicated.

NP-111001 derivatives (Noscira, Madrid) act as tauphosphorylation inhibitors (Thomson Reuters Pharma,update of January 12, 2012). The structures were notcommunicated.

NPT-002 (NeuroPhage Pharmaceuticals, Cam-bridge, MA) targets A� and tau aggregates for thepotential treatment of AD (Thomson Reuters Pharma,

Page 48: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

48 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 15. Molecules preventing tau aggregation.

Page 49: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 49

update of July 16, 2012). The structure was not com-municated.

Protein phosphatase methylesterase 1 (PME1)inhibitors (Signum Biosciences and GSK) are smallmolecules blocking the enzyme which demethylatesPP2A. Normally PP2A is almost all methylated, but inAlzheimer’s disease PP2A methylation is reduced inhalf. The structural mechanism of demethylation andinactivation of protein phosphatase 2a was described[1747–1749] (Thomson Reuters Pharma, updateof November 15, 2012). The structures were notcommunicated.

Protein phosphatase 2A stimulators (Cognosci,Research Triangle Park, NC) inhibit the formation ofapoE/inhibitor 2 of PP2A (I2PP2A) complex. The to-pic protein phosphatases and AD was reviewed [1526,1527] (Thomson Reuters Pharma, update of February24, 2012). The structures were not communicated.

ReS3-T, ReS8-T, ReS10-T and ReS19-T(reMYND, a spin-off from the University of Leuven,Belgium and licensee Roche) are four series of taudetoxifying compounds targeting tau-mediated cyto-toxicity (Thomson Reuters Pharma, update of Septem-ber 14, 2012). The structures were not communicated.

The ReS9-S7 and ReS12-S programs (reMYND,a spin-off from the University of Leuven, Belgiumand licensee Roche) investigate the attenuation ofthe cytotoxic effects of �-synuclein aggregation indopaminergic neurons for the potential treatment ofPD (Thomson Reuters Pharma, update of September14, 2012). The structures were not communicated.

SIG-1012 (Cognion) and SIG-1106 (Signum Bio-sciences, Monmouth Junction NJ in collaboration withPrinceton Univ.) are phosphoprotein 2A modulatorsand presumed to also include the PP2A methylesterase1 inhibitor eicosanoyl-5-hydroxytryptamide (EHT), allextracted from coffee and acting as tau aggregationinhibitors. (Thomson Reuters Pharma, update of Octo-ber 05, 2012). The structures were not communicated.

Small molecule Tau protein modulators(Asceneuron, a spun-out of Merck Serono, Geneva,Switzerland) are drugs for the potential treatment ofAD (Thomson Reuters Pharma, update of October 3,2012). Structures were not disclosed.

Small molecule tau modulators (Yuma Thera-peutics, Brookline, MA) target neurofibrillary tanglesresulting from abnormal forms of the protein tau(Thomson Reuters Pharma, update of March 29, 2011).

Sodium selenate mitigated tau pathology and func-tional deficits in AD models [1528].

Tau aggregation inhibitors (Catholic University ofLeuven, Belgium) are currently in the preclinical Phase

(Thomson Reuters Pharma, update of July 30, 2012).Structures were not communicated.

Tau aggregation inhibitors (ProteoTech, Kirk-land, WA) have a potential for the treatment of AD(Thomson Reuters Pharma, update of July 30, 2012).Structures were not communicated.

Tau oligomer inhibitors (Oligomerix, New York,NY) are presumably curcumin derivatives (ThomsonReuters Pharma, update of April 24, 2012).

Tau phosphorylation inhibitors (ProQinase,Freiburg im Breisgau, Germany) (Fig. 15) reducedin vivo GSK-3� selective phosphorylation ofSer396/Ser404 and Ser262 in the brains of tripletransgenic mice after i.p. administration. It appearsthat the development was terminated (ThomsonReuters Pharma update of October 11, 2012).

Therapeutic program (B & C Biopharm andEquispharm, both South Korea) is targeting taukinase (Thomson Reuters Pharma update of May 18,2012).

Thiamet-G (Simon Fraser University) is a potentinhibitor of human O-GlcNAcase (Ki = 21 nM)(Fig. 15) and efficiently reduced phosphorylation oftau at Thr231, Ser396, and Ser422 in both rat cor-tex and hippocampus [1529–1533]. For commentaries,see [1534, 1535]. See also Part 2, Chapter 2.27. Drugsinteracting with O-GlcNAcase.

THQ-4S and THQ-55 (Fig. 15) interact specificallywith oligomeric forms of tau inhibiting their assemblyinto AD filaments [1536, 1537].

TRx-0237 (TauRx Therapeutics, Singapore) is thelead compound of second-generation tau aggregationinhibitors. Safety and efficacy studies in AD and fron-totemporal dementia patients are planned for 2012(Thomson Reuters Pharma, update of September 25,2012). The structure was not communicated.

Tubastatin A (Fig. 15) is a potent and selectiveHDAC6 inhibitor [1750]. Chronic tubastatin treatmentfor two months decreased total tau levels in rtg4510mice [1751].

Natural products as a source of tau-targeting drugswere described [1538].

Excellent papers on small molecule inhibitors oftau aggregation were published by researchers atuniversities (in chronological order) 2004: [1539],2005: [1540] (benzothiazoles such as N-744, Fig. 15),[1541, 1542] 2006: [1543]; 2007: [1544–1550], 2009:[1551–1555], 2010: [1556, 1557], 2011: [1558, 1559],2012: [1560, 1561].

Excellent reviews on tau-focused drug discoveryfor AD were presented (in chronological order)2000: [1562]; 2002: [1563], 2006: [1564], 2007:

Page 50: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

50 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1565], 2008: [1566–1569], 2009: [1570, 1571], 2010:[1572–1574], 2011: [1575, 1576] [1577, 1578], 2012:[1579–1581]. Interestingly, synergistic interactionsbetween repeats in tau protein and A� were described[1582].

The development of the tau aggregation inhibitorsaminothienopyridazines (University of Pennsyl-vania), AZD-1080 (AstraZeneca), BPU-410 andBPU-430 (Champions Biotechnology under licensefrom Johns Hopkins University), LDN-33960 (astriatal-enriched protein tyrosine phosphatase (STEP)inhibitor, Yale University), NHT-0112 (Neuro-Hitech), protein phosphatase methylesterase 1(PME1) inhibitors (Scripps Research Institute andMassachusetts Institute of Technology), SDGI-T200801 and SDGII-T200801 (both Bioalvo),SRN-003-556 (SIRENADE Pharmaceuticals) and ofTRx-0037 (TauRx) were terminated.

Ligands interacting with tau

T-777, T-807, and T-808 (Siemens Medical Solu-tion Molecular Imaging) (Fig. 15) are tau-bindingmolecules as PET tracers for the diagnosis of AD.They are benz[4,5]imidazo[1,2-a]pyrimidines. 18F-T808 displayed a high level of binding affinity(Kd = 22 nM) and good selectivity for tau aggregatesover A� plaques [1583] (Thomson Reuters Pharma,update of September 19, 2011).

Novel in vivo tau imaging agents 18F-THK-523(University of Melbourne) [1584] and 11C-THK-951(Tohoku University) (both Fig. 15) were published[1585] after preliminary results of 2005 [1752] (Thom-son Reuters Pharma, update of September 219, 2012).

The University of Melbourne is investigating18F labeled arylquinoline derivatives, such as18F-THK-5105, 18F-THK-5116, 18F-THK-5117, and18F-THK-5125 for the potential use in imaging taudeposition in neurofibrillary tangles for AD (Thom-son Reuters Pharma, update of June 13, 2012). Thestructures were not communicated.

Researchers of Amersham (GE Healthcare) pub-lished a patent describing ligands with an exceptionalselectivity to tau: compound 18 (Fig. 15): Kd = 0.9 nMfor tau and Kd = 30 �M for A�1-40 [1586].

A 125I-3-oxindole derivative stained neurofib-rillary tangles in AD brain sections [1587].Also bis(arylvinyl)pyrazines, -pyrimidines, and-pyridazines were described as imaging agents for taufibrils and A� plaques [1588] as were 2-styrylindoliumbased fluorescent probes [1753].

Research toward tau imaging was reviewed [1553,1589, 1590] stating that ligand polarizability con-tributes to tau fibril binding affinity.

Vaccines against tau

Recombinant misfolded truncated tau pro-tein vaccine (Axon Neurosciences, Vienna, Austria)attenuated pathology in vivo in a transgenic rat modelof tauopathy (Thomson Reuters Pharma, update ofFebruary 17, 2011).

RV-03 (Intellect Neurosciences, New York, NY)is a peptide vaccine developed using the RECALL-VAX technology targeting both the A� and a truncateddelta tau protein (Thomson Reuters Pharma, update ofAugust 21, 2012).

Phosphorylated tau peptides were used to immunizetransgenic mice to produce robust anti-neurofibrillarytangle effects [1591].

The development of the chimeric peptide vaccineRV-02 (Intellect Neurosciences) was terminated.

Antibodies against tau and α-synuclein

Humanized tau monoclonal antibodies (ACImmune, Lausanne, Switzerland) had high specificaffinity binding to pTau. The compound was out-licensed to Genentech (Thomson Reuters Pharma,update of June 19, 2012).

Lilly presented data on passive immunization withanti-tau antibodies in two transgenic mouse models[1754].

NI-105 (Biogen Idec, Weston, MA through theacquisition of Panima, previously a subsidiary ofNeurimmune Therapeutics, Zurich Switzerland) is ahuman recombinant monoclonal antibody targeted totau protein created using Neurimmune’s reverse trans-lational medicine platform (Thomson Reuters Pharma,update of May 4, 2012).

PD-0805 (Bioarctic Neuroscience, Stockholm,Sweden) is a monoclonal antibody targeting �-synuclein for the potential treatment of PD and DLB(Thomson Reuters Pharma, update of September 2,2011).

Pfizer presented the first X-ray structure of an avianantibody to its cognate phosphopeptide pT231/pS235at 1.9 A resolution [1755].

T01-OX2 (Intellect Neurosciences, New York, NY)is an antibody drug conjugate that consists of a mAbtargeting oligomeric forms of tau protein conjugatedto melatonin (Thomson Reuters Pharma, update ofAugust 17, 2012).

Page 51: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 51

TauC3 monoclonal antibody (Intellect Neu-rosciences, New York, NY under license fromNorthwestern University) is targeting the neoepitopetauC3 (Thomson Reuters Pharma, update of September20, 2012).

Tau protein modulator (iPierian, South SanFrancisco, CA) is a monoclonal antibody for thepotential treatment of AD, frontotemporal dementiaand progressive supranuclear palsy (Thomson ReutersPharma, update of May 22, 2012).

Tau-targeted antibody therapy (Neotope Bio-sciences, South San Francisco, CA and Elan) is amonoclonal antibody targeted to tau (Thomson ReutersPharma, update of August 13, 2012).

TOC-1 (Intellect Neurosciences, New York underlicense from Northwestern University) is a tau-oligomer-targeting monoclonal antibody [1592, 1593](Thomson Reuters Pharma, update of September 20,2012).

The detection of naturally occurring anti-tau anti-bodies was described [1594].

Already in 1985 monoclonal antibodies to AD neu-rofibrillary tangles were described [1595]. Excellentpapers on passive immunization targeting pathologicalphospho-tau protein were published (in chronologi-cal order) 2005: [1596], 2007: [1597], 2008: [1598,1599], 2009: [1600, 1601], 2010: [1602, 1603], 2011:[1604–1607], 2012: [1756].

The development of TTRY78F (University ofPorto), an anti-transthyretin monoclonal antibody, wasterminated.

STEM CELLS

Substantial progress has been achieved in researchof stem cells for the potential treatment of ADcommunicated in chronological order: 2000: [1608],2001: [1609–1611], 2006: [1612–1614], 2007: [1615,1616], 2008: [1617, 1618], 2009: [1619–1622], 2010:[1623–1630], 2011: [1631–1635], 2012: [1636–1642,1757].

Human neural stem cells overexpressing cholineacetyltransferase restored cognitive function of kainic-acid-induced learning and memory deficits in rats[1643, 1644]. Neural stem cells reduced hippocam-pal tau and reelin accumulation [1645]. Intracerebraltransplantation of bone marrow-derived mesenchymalstem cells reduced A� deposition and rescued mem-ory deficits in AD mice by modulation of immuneresponses [1626]. Neural stem cells improved cog-nition via BDNF in a transgenic model of AD

[1646–1648]. Human neural stem cells geneticallymodified to express human NGF gene restoredcognition in mice [1649]. Human neural stem cellsgenetically modified to overexpress BDNF promotefunctional recovery and neuroprotection in a mousestroke model [1650]. Neural stem cells improved learn-ing and memory in rats with AD [1651] and improvedneuronal survival in cultured postmortem brain tissuefrom aged and AD patients [1652].

Induced pluripotent stem cells may be used to modelpatient-specific AD in vitro [1653, 1654]. For a com-mentary, see [1655]; to model pathology in Downsyndrome [1656] and in HD [1657]. They may cre-ate new opportunities for disease modeling and drugdiscovery [1758].

GDNF/BDNF-producing glial and brain-derivedstem cells (NurOwn; BrainStorm Cell Therapeutics,New York, NY) is investigated as a potential treatmentof PD, sciatica, and multiple sclerosis [1658, 1659].A Phase I/II clinical trial in ALS patients started inJune 2011 in Israel. In January 2012 positive data werereported (Thomson Reuters Pharma, update of July 24,2012).

Human neural stem cell therapy (HuCNS-SC;StemCells, Newark, CA, formerly CytoTherapeuticsunder license from NeuroSperes) is a proprietary trans-plantable human neural stem cell therapy for thepotential treatment of spinal cord injury, age-relatedmacular degeneration, and AD. Human neural stemcells induced functional myelination in mice [1759]and in the human brain [1760]. A Phase II trial forspinal cord injury was initiated in Switzerland in March2011. The neuroprotective effects in retinal degenera-tion were described [1660] (Thomson Reuters Pharma,update of September 28, 2012).

Mesenchymal bone marrow-derived stem celltherapy (Stemedica Cell Technologies, San Diego,CA) is being developed for the intravenous treatmentof ischemic stroke in Phase I/II since February 2011in the US (n = 35). The study was expected to be com-pleted in February 2013 (Thomson Reuters Pharma,update of April 11, 2012).

NSI-189 (Neuralstem, Rockville, MD, in collabora-tion with Japanese licensee Summit Pharmaceuticals)(Fig. 16) is an orally bioavailable small-molecule neu-rogenesis stimulator in Phase I, which was completedin October 2011. The FDA approved the Phase Ib trialin December 2011 (three cohorts of eight patients each). In June 2012, the first patients were dosed (ThomsonReuters Pharma, update of September 28, 2012).

NSI-566RSC (Neuralstem, Rockville, MD) arehuman neural stem cells including spinal cord stem

Page 52: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

52 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Fig. 16. A neurogenesis stimulator and two SV2A ligands.

cells for the potential injectable treatment of ALS,stroke, HD, and AD. Long-distance growth and con-nectivity of neural stem cells after severe spinal cordinjury were investigated [1661]. A Phase I clinicaltrial for ALS started in January 2010 in the US. Theresults of a Phase I trial in 12 ALS patients receivinglumbar intraspinal injection of neural stem cells weredescribed [1662] (Thomson Reuters Pharma, update ofSeptember 28, 2012).

Neurostem-AD (Medipost, South Korea) is anumbilical cord blood-derived mesenchymal stem celltherapy, which regenerates nerve cells. A Phase Itrial has been initiated in February 2011 in patients(expected n = 9) with dementia of AD-type (ThomsonReuters Pharma, update of September 25, 2012).

There are several stem cell preparations in preclini-cal evaluation (in alphabetical order):

Adult mesenchymal precursor stem cell therapy(Mesoblast, Melbourne, Australia and Cephalon, awholly-owned subsidiary of Teva) is evaluated for thepotential treatment of AD and PD and stroke (ThomsonReuters Pharma, update of September 6, 2012).

Allogenic umbilical cord stem cell therapy(U-CORD; CellPraxis Bioengenharia, Bela Vista,Brazil) is investigated for the potential treatmentof Alzheimer’s disease. (Thomson Reuters Pharma,update of November 07, 2012).

Brain-derived stem cell therapy (Celprogen, SanPedro, CA, in collaboration with the Indiana UniversitySchool of Medicine) can be differentiated into neurons.In vivo preclinical studies demonstrated improvementin short and long term memory in AD experimentalmodels (Thomson Reuters Pharma, update of July 14,2011).

CPG23NEUR (Celprogen, San Pedro, CA) derivesfrom cord blood stem cells, which are trans-differentiating into neuronal cells (Thomson ReutersPharma, update of January 30, 2012).

Glial progenitor cell therapy (Q Therapeutics, SaltLake City, UT) has the potential to replace myelinon damaged neurons for the treatment of transversemyelinitis, spinal cord injury, multiple sclerosis, PD,and AD (Thomson Reuters Pharma, update of January20, 2012).

Human neural progenitor cells (Cedars-SinaiMedical Center, Los Angeles, CA), which secretegrowth factors including glial cell-derived neurotro-phic factor, may be useful for the treatment of ALS(Thomson Reuters Pharma, update of July 31, 2012).

Human umbilical cord blood cells (Saneron CCELTherapeutics, Tampa FL) are cerebroprotective agentswhich showed cognition enhancing and amyloid-�reducing activities for the potential treatment of strokeand Alzheimer’s disease (Thomson Reuters Pharma,update of October 26, 2012).

NBI-18 (NeoCytex Biopharma, Covington, KY),a heterocyclic pyrimidine derivative, is a stem cellstimulator for the potential treatment of neurodegener-ative diseases including ALS, AD, and PD (ThomsonReuters Pharma, update of January 19, 2012). Thestructure was not communicated.

Neural stem cell therapy (Stemedica Cell Tech-nologies, San Diego, CA) is an allogeneic therapyfor the potential treatment of AD (Thomson ReutersPharma, update of February 22, 2012). Anotherstem cell therapy is evaluated for spinal cord injury(Thomson Reuters Pharma, update of February 28,2012).

NeurotrophinCell (Living Cell Technologies,Auckland, NZ) is an alginate-microencapsulatedporcine choroid plexus cell product for the potentialtreatment of neurodegenerative diseases including HD,PD, and dementia [1663, 1664] (Thomson ReutersPharma, update of May 16, 2012).

NGN-9079 (neural stem cell therapy; NeuroGener-ation, Los Angeles, CA) is evaluated for the potential

Page 53: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 53

treatment of AD. Transplantation of neuronal cellsinto the brain was shown to improve and delay thesymptoms of AD in animal models (Thomson ReutersPharma, update of March 2, 2012).

ReN-004 (ReNeuron, Guildford, UK) is a neuralstem cell therapy for PD and AD (Thomson ReutersPharma, update of September 5, 2012).

ReN-005 (ReNeuron, Guildford, UK) is a neuralstem cell therapy for HD (Thomson Reuters Pharma,update of December 24, 2010).

Stem cell therapeutics (Samaritan Pharmaceuti-cals, Las Vegas, NV) are non-embryonic neuronal stemcell differentiation therapeutics including the naturallyoccurring compounds SP-sc4 and SP-sc7 to inducestem cell differentiation for the potential treatment ofAD (Thomson Reuters Pharma, update of March 16,2011).

Allopregnanolone (University of Southern Califor-nia, Los Angeles, CA) is a potent and highly efficaciousproliferative agent in vitro and in vivo of both rodentand human neural stem cells [1665–1667].

Granulocyte colony-stimulating factor is knownto mobilize hematopoietic stem cells from the bonemarrow into the peripheral blood. Subcutaneousadministration of granulocyte colony-stimulating fac-tor into two different A�-induced AD mouse modelssubstantially rescued their cognitive/memory func-tions [1668].

Valproic acid can induce neurogenesis of neuralprogenitor/stem cells both in vitro and in vivo via mul-tiple signaling pathways [1669].

The development of GRNOPC-1 (Geron), oligo-dendrocyte precursor cells differentiated from a humanembryonic stem cell line that produce neurotrophic fac-tors and remyelinate axons for the potential injectabletreatment of PD, stroke, AD, multiple sclerosis, andspinal cord injury in Phase I since October 2010was terminated. Also the development of the humanembryonic stem cell therapy (GRNIC-1, Geron),the cell-based therapy MDA-200C (Medeia Thera-peutics), NEBO-176 (Neuro Bioscience and RLI),propentofylline (Endogenous Stem Cells Activators(ESAI) under license from sanofi) and of stem celltherapies for glaucoma, macular degeneration, PD,traumatic brain injury, and vascular dementia (Stemed-ica) was terminated.

MISCELLANEOUS

Autophagy inducer JRP-900 (Prous Institute forBiomedical Research, Barcelona) is investigated for

the potential treatment of neurodegenerative diseaseincluding AD, ALS, HD, and PD (Thomson ReutersPharma, update of July 23, 2012). The structurewas not communicated. For reviews on autophagy inAlzheimer’s disease and tauopathies see [1761–1764].

Cellular homeostasis modulator CNS-102 (Coy-ote Pharmaceuticals) acts on protein misfolding forthe potential treatment of AD, multiple sclerosis, andALS (Thomson Reuters Pharma, update of October 2,2012). The structure was not communicated.

Glycan inhibitors (Stelic Institute, Tokyo) areinvestigated for the potential treatment of Parkinson’sand Alzheimer’s disease. An exhaustive review wasprovided [1765]. (Thomson Reuters Pharma, update ofOctober 16, 2012). Structures were not communicated.

Macrophage migration inhibitory factor (MIF)inhibitor INV-88 (Innovimmune Biotherapeutics,New York) is investigated for the potential oral treat-ment of inflammatory diseases, central nervous systemdiseases including Alzheimer’s disease and multiplesclerosis and age-related macular degeneration (Thom-son Reuters Pharma, update of October 18, 2012). Thestructure was not communicated.

MicroRNA (miRNA) mimetics (University ofGottingen, MBM Science) are evaluated for the poten-tial diagnosis and treatment of memory impairment.By June 2012, proof of concept was achieved in anAD mouse model and in human AD patient samples.The relationship of miRNAs in the brain and A� gener-ation was discussed [1670] (Thomson Reuters Pharma,update of August 7, 2012).

Proteasome-gating modulators are investigatedby Proteostasis Therapeutics, Cambridge, MA underlicense from Harvard University for the potential treat-ment of neurodegenerative diseases including AD andPD (Thomson Reuters Pharma, update of August 15,2012).

Synaptic vesicle glycoprotein 2A (SV2A) ligandlevetiracetam (L-059, Keppra; UCB Pharma, Brus-sels, Belgium and Japanese licensee Otsuka) (Fig. 16)is an antiepileptic drug launched in the US and EUin 2000 and in Japan in 2010 [1671–1676]. It bindsto the same binding site as botulinum neurotoxin Aand tetanus neurotoxin [1677, 1678]. It is still unclearhow levetiracetam modulates SV2A’s function(s). Itinhibited presynaptic Ca2+ channels through an intra-cellular pathway [1679, 1680]. It may promoteglutamate uptake by increasing glutamate transporterexpression [1681]. There are hints that levetiracetammay interact with GABAA receptors [1682–1684].Levetiracetam had a positive impact on emotionallearning and memory in naıve mice [1685] and

Page 54: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

54 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

reversed cognitive deficits in hA�PP transgenic mice[1686]. Levetiracetam improved cognitive function indrug-naıve epilepsy patients [1687] and in childrenaged 4 to 16 years [1688]. The sales of levetiracetamreported by UCB for 2011 were USD 1,345 million andby Otsuka, USD 60.5. An injectable formulation waslaunched in the US in August 2006, an extended releaseformulation in September 2008 (Thomson ReutersPharma, update of October 1, 2012).

Low-dose therapy Levetiracetam (AgeneBio,Carmel, IN under license from John Hopkins Uni-versity) is investigated for the potential treatmentof amnestic MCI (aMCI) in Phase II clinical tri-als since December 2009 in 144 aMCI patients inthe US. Reduction of hippocampal hyperactivity inaMCI patients by using a low dose of levetiracetamimproved cognition [1766]. It appears that the devel-opment was terminated (Thomson Reuters Pharma,update of September 7, 2011).

Brivaracetam (UCB-34714; Rikelta; UCB Pharma,Brussels, Belgium) (Fig. 16), the n-propyl analogueof levetiracetam, is an orally active ligand of synapticvesicle protein 2A (SV2A) currently in Phase III clini-cal trials for the treatment of epilepsy since November2008 (n = 600). The study is expected to complete inJune 2015 [1689, 1690]. Its binding characteristics asa high affinity SV2A ligand in rat, mouse and humanbrain were reported [1691] as was its effect on theinhibition of Na+ currents [1692]. Its neurocognitiveeffects were similar to the ones of levetiracetam [1693,1694] (Thomson Reuters Pharma, update of August 8,2012).

CONCLUSION

Two products were launched in 2012: tafamidis(Vyndaquel, Pfizer) for the treatment of transthyretinfamilial amyloid polyneuropathy in Europe in Marchand 18F-florbetapir (Amyvid; Avid Radiopharma-ceuticals, Lilly) as a PET imaging agent to estimateA� neuritic plaque density in patients with cognitiveimpairment in the US in June.

Phase III Alzheimer’s disease trials of a diseasemodifying drug, the monoclonal antibody against A�solanezumab (Lilly), are close to completion. Firstresults showed that solanezumab failed to meet the cog-nitive and functional primary endpoints. But it showeda significant reduction in cognitive decline in patientswith mild AD. For a commentary, see [1695]. Theopen-label EXPEDITION-EXT extension trial, whichis fully enrolled, is continuing as planned.

The development of the second Phase III mono-clonal antibody bapineuzumab (Janssen AlzheimerImmunotherapy and Pfizer) was terminated.

The Phase III clinical trials of other A� PETimaging agents, 18F-florbetaben (Piramal) and 18F-flutemetamol (GE Healthcare), may be successfullycompleted by next year. The completion of PhaseIII clinical trials of the VMAT2 PET ligand 18F-florbenazine (Avid Radiopharmaceuticals, Lilly) isplanned for September 2014.

Phase II clinical trials of other disease modi-fying drugs, of monoclonal antibodies against A�gantenerumab, crenezumab, GSK-93377A, intra-venous immune globulin, and octagam, of vaccinesagainst A� AD-02, CAD-106, and vanutide cridi-ficar and the PET ligand 18F-AZD-4694 (Navidea)will require several years. This applies also for thePhase II clinical trials of small molecules preventingA� aggregation or inhibiting formation of transthyretinamyloid fibrils, such as doxycycline hyclate, ELND-005, and SOM-0226 or preventing tau aggregation,such as PBT-2, tideglusib, and TRx-0014.

Concerning drugs for palliative treatment of AD,two Phase III compounds are currently frontrunners,DP-b99 (a calcium and zinc chelator) and SK-PC-B70M (a natural product) followed by 22 Phase IIcompounds, RV-208 (a gene expression stimulator),ARC-029 and ZSET-1446/ST-101 (calcium channelblockers), CERE-110, GM-607, PYM-50058, andT-817MA (trophic factor stimulators), circadin, KD-501, PPL, PTX-200, resveratrol, RPh-201 (naturalproducts), LSL-001, N-251, PF-03049423, TRM-189, VI-1121 (nootropics), davunetide, AM-111,etanercept, and FGL (peptides).

DISCLOSURE STATEMENT

Authors’ disclosures available online (http://www.j-alz.com/disclosures/view.php?id=1543).

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, DeRubeis RJ,Dubois B, Geyer MA, Goodwin GM, Gorwood P, Jay TM,Joels M, Mansuy IM, Meyer-Lindenberg A, Murphy D,Rolls E, Saletu B, Spedding M, SweeneyJ, WhittingtonM, Young LJ (2012) Cognitive dysfunction in psychiatric

Page 55: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 55

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. JAlzheimers Dis 32, 793-887.

[6] Citron M (2004) Strategies for disease modification inAlzheimer’s disease. Nat Rev Neurosci 5, 677-685.

[7] Pangalos MN, Jacobsen SJ, Reinhart PH (2005) Diseasemodifying strategies for the treatment of Alzheimer’s dis-ease targeted at modulating levels of the beta-amyloidpeptide. Biochem Soc Trans 33, 553-558.

[8] Hull M, Berger M, Heneka M (2006) Disease-modifyingtherapies in Alzheimer’s disease: How far have we come?Drugs 66, 2075-2093.

[9] Vellas B, Andrieu S, Sampaio C, Wilcock G (2007)Disease-modifying trials in Alzheimer’s disease: A Euro-pean task force consensus. Lancet Neurol 6, 56-62.

[10] Sadowski M, Wisniewski T (2007) Disease modifyingapproaches for Alzheimer’s pathology. Curr Pharm Des13, 1943-1954.

[11] Salloway S, MintzerJ, Weiner MF, Cummings JL (2008)Disease-modifying therapies in Alzheimer’s disease.Alzheimers Dement 4, 65-79.

[12] 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.

[13] Vellas B, Coley N, Andrieu S (2008) Disease modifyingtrials in Alzheimer’s disease: Perspectives for the future. JAlzheimers Dis 15, 289-301.

[14] Duara R, Barker W, Loewenstein D, Bain L (2009) Thebasis for disease-modifying treatments for Alzheimer’sdisease: The Sixth Annual Mild Cognitive ImpairmentSymposium. Alzheimers Dement 5, 66-74.

[15] 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.

[16] Citron M (2010) Alzheimer’s disease: Strategies for dis-ease modification. Nat Rev Drug Discov 9, 387-398.

[17] 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.

[18] Dash SK (2011) Future targeted disease modifying drugsfor Alzheimer’s disease. Recent Pat CNS Drug Discov 6,65-76.

[19] Galimberti D, Scarpini E (2010) Treatment of Alzheimer’sdisease: Symptomatic and disease-modifying approaches.Curr Aging Sci 3, 46-56.

[20] Galimberti D, Scarpini E (2011) Disease-modifying treat-ments for Alzheimer’s disease. Ther Adv Neurol Disord 4,203-216.

[21] Galimberti D, Scarpini E (2011) Alzheimer’s disease:From pathogenesis to disease-modifying approaches. CNSNeurol Disord Drug Targets 10, 163-174.

[22] Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F(2012) New pharmacological strategies for treatment ofAlzheimer’s disease: Focus on disease modifying drugs.Br J Clin Pharmacol 73, 504-517.

[23] Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logros-cino G, Santamato A, Greco A, Seripa D, Pilotto A(2012) Immunotherapy for Alzheimer’s disease: Fromanti-beta-amyloid to tau-based immunization strategies.Immunotherapy 4, 213-238.

[24] Cummings JL, Doody R, Clark C (2007) Disease-modifying therapies for Alzheimer disease: Challenges toearly intervention. Neurology 69, 1622-1634.

[25] Cummings JL (2008) Optimizing phase II of drug devel-opment for disease-modifying compounds. AlzheimersDement 4, S15-S20.

[26] Cummings JL (2009) Defining and labeling disease-modifying treatments for Alzheimer’s disease. AlzheimersDement 5, 406-418.

[27] Jiang T, Yu JT, Tan L (2012) Novel disease-modifyingtherapies for Alzheimer’s disease. J Alzheimers Dis 31,475-492.

[28] Lee KS, Chung JH, Choi TK, Suh SY, Oh BH, HongCH (2009) Peripheral cytokines and chemokines inAlzheimer’s disease. Dement Geriatr Cogn Disord 28,281-287.

[29] Gitter BD, Cox LM, Rydel RE, May PC (1995) Amyloidbeta peptide potentiates cytokine secretion by interleukin-1 beta-activated human astrocytoma cells. Proc Natl AcadSci U S A 92, 10738-10741.

[30] Harries LW, Bradley-Smith RM, Llewellyn DJ, PillingLC, Fellows A, Henley W, Hernandez D, GuralnikJM, Bandinelli S, Singleton A, Ferrucci L, Melzer D(2012) Leukocyte CCR2 expression is associated withmini-mental state examination score in older adults. Reju-venation Res 15, 395-404.

[31] Ralay Ranaivo H, Craft JM, Hu W, Guo L, Wing LK, VanEldik LJ, Watterson DM (2006) Glia as a therapeutic tar-get: Selective suppression of human amyloid-beta-inducedupregulation of brain proinflammatory cytokine produc-tion attenuates neurodegeneration. J Neurosci 26, 662-670.

[32] Bachstetter AD, Norris CM, Sompol P, Wilcock DM,Goulding D, Neltner JH, St CD, Watterson DM, VanEldik LJ (2012) Early stage drug treatment that normalizesproinflammatory cytokine production attenuates synapticdysfunction in a mouse model that exhibits age-dependentprogression of Alzheimer’s disease-related pathology. JNeurosci 32, 10201-10210.

[33] Hu W, Ralay RH, Roy SM, Behanna HA, Wing LK,Munoz L, Guo L, Van Eldik LJ, Watterson DM (2007)Development of a novel therapeutic suppressor of brainproinflammatory cytokine up-regulation that attenuatessynaptic dysfunction and behavioral deficits. Bioorg MedChem Lett 17, 414-418.

[34] Karpus WJ, Reynolds N, Behanna HA, Van Eldik LJ, Wat-terson DM (2008) Inhibition of experimental autoimmuneencephalomyelitis by a novel small molecular weightproinflammatory cytokine suppressing drug. J Neuroim-munol 203, 73-78.

[35] Lloyd E, Somera-Molina K, Van Eldik LJ, Watterson DM,Wainwright MS (2008) Suppression of acute proinflamma-tory cytokine and chemokine upregulation by post-injuryadministration of a novel small molecule improveslong-term neurologic outcome in a mouse model of trau-matic brain injury. J Neuroinflammation 5, 28.

[36] Somera-Molina KC, Robin B, Somera CA, Anderson C,Stine C, Koh S, Behanna HA, Van Eldik LJ, Watter-son DM, Wainwright MS (2007) Glial activation linksearly-life seizures and long-term neurologic dysfunction:Evidence using a small molecule inhibitor of proin-

Page 56: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

56 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

flammatory cytokine upregulation. Epilepsia 48, 1785-1800.

[37] O’Hare E, Scopes DI, Treherne JM, Monaghan J, PalmerPM, Amijee H, Kim EM (2011) Novel anti-inflammatorycompound SEN1176 alleviates behavioral deficits inducedfollowing bilateral intrahippocampal injection of aggre-gated amyloid-beta. J Alzheimers Dis 25, 219-229.

[38] Shi JQ, Wang BR, Jiang WW, Chen J, Zhu YW, ZhongLL, Zhang YD, Xu J (2011) Cognitive improvement withintrathecal administration of infliximab in a woman withAlzheimer’s disease. J Am Geriatr Soc 59, 1142-1144.

[39] Bacher M, Dodel R, Aljabari B, Keyvani K, MarambaudP, Kayed R, Glabe C, Goertz N, Hoppmann A, SachserN, Klotsche J, Schnell S, Lewejohann L, Al-Abed Y(2008) CNI-1493 inhibits Abeta production, plaque for-mation, and cognitive deterioration in an animal model ofAlzheimer’s disease. J Exp Med 205, 1593-1599.

[40] Bach JP, Mengel D, Wahle T, Kautz A, Balzer-GeldsetzerM, Al-Abed Y, Dodel R, Bacher M (2011) The role of CNI-1493 in the function of primary microglia with respect toamyloid-beta. J Alzheimers Dis 26, 69-80.

[41] Nilsson P, Iwata N, Muramatsu S, Tjernberg LO, Win-blad B, Saido TC (2010) Gene therapy in Alzheimer’sdisease–potential for disease modification. J Cell Mol Med14, 741-757.

[42] Goldberg TE, Weinberger DR (2004) Genes and the pars-ing of cognitive processes. Trends Cogn Sci 8, 325-335.

[43] Zetterstrom TS, Pei Q, Madhav TR, Coppell AL, LewisL, Grahame-Smith DG (1999) Manipulations of brain5-HT levels affect gene expression for BDNF in rat brain.Neuropharmacology 38, 1063-1073.

[44] Coppell AL, Pei Q, Zetterstrom TS (2003) Bi-phasicchange in BDNF gene expression following antidepressantdrug treatment. Neuropharmacology 44, 903-910.

[45] Bourhis E, Maheux J, Rouillard C, Levesque D (2008)Extracellular signal-regulated kinases (ERK) and proteinkinase C (PKC) activities are involved in the modulationof Nur77 and Nor-1 expression by dopaminergic drugs. JNeurochem 106, 875-888.

[46] Uusi-Oukari M, Korpi ER (2010) Regulation of GABA(A)receptor subunit expression by pharmacological agents.Pharmacol Rev 62, 97-135.

[47] Southwell AL, Patterson PH (2011) Gene therapy in mousemodels of huntington disease. Neuroscientist 17, 153-162.

[48] Morishita R, Aoki M, Hashiya N, Makino H, Yamasaki K,Azuma J, Sawa Y, Matsuda H, Kaneda Y, Ogihara T (2004)Safety evaluation of clinical gene therapy using hepatocytegrowth factor to treat peripheral arterial disease. Hyperten-sion 44, 203-209.

[49] Makino H, Aoki M, Hashiya N, Yamasaki K, Azuma J,Sawa Y, Kaneda Y, Ogihara T, Morishita R (2012) Long-term follow-up evaluation of results from clinical trialusing hepatocyte growth factor gene to treat severe periph-eral arterial disease. Arterioscler Thromb Vasc Biol 32,2503-2509.

[50] Anonymous (2011) Rvx 208. Drugs R D 11, 207-213.[51] Nicholls SJ, Gordon A, Johannson J, Ballantyne CM,

Barter PJ, Brewer HB, Kastelein JJ, Wong NC, BorgmanMR, Nissen SE (2012) ApoA-I induction as a potentialcardioprotective strategy: Rationale for the SUSTAIN andASSURE studies. Cardiovasc Drugs Ther 26, 181-187.

[52] Bailey D, Jahagirdar R, Gordon A, Hafiane A, Campbell S,Chatur S, Wagner GS, Hansen HC, Chiacchia FS, Johans-son J, Krimbou L, Wong NC, Genest J (2010) RVX-208:

A small molecule that increases apolipoprotein A-I andhigh-density lipoprotein cholesterol in vitro and in vivo. JAm Coll Cardiol 55, 2580-2589.

[53] McNeill E (2010) RVX-208, a stimulator of apolipopro-tein AI gene expression for the treatment of cardiovasculardiseases. Curr Opin Investig Drugs 11, 357-364.

[54] Davidson MH (2011) Apolipoprotein A-I therapy promise,challenges, and disappointment. J Am Coll Cardiol 57,1120-1121.

[55] Nicholls SJ, Gordon A, Johansson J, Wolski K, Ballan-tyne CM, Kastelein JJ, Taylor A, Borgman M, NissenSE (2011) Efficacy and safety of a novel oral inducer ofapolipoprotein a-I synthesis in statin-treated patients withstable coronary artery disease a randomized controlledtrial. J Am Coll Cardiol 57, 1111-1119.

[56] Shah PK (2011) Atherosclerosis: Targeting endogenousapo A-I –a new approach for raising HDL. Nat Rev Cardiol8, 187-188.

[57] 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.

[58] Yang SY, He XY, Miller D (2007) HSD17B10: A geneinvolved in cognitive function through metabolism ofisoleucine and neuroactive steroids. Mol Genet Metab 92,36-42.

[59] Hafez DM, Huang JY, Richardson JC, Masliah E, PetersonDA, Marr RA (2012) F-spondin gene transfer improvesmemory performance and reduces amyloid-beta levels inmice. Neuroscience 223, 465-472.

[60] Chiba T, Yamada M, Hashimoto Y, Sato M, Sasabe J,Kita Y, Terashita K, Aiso S, Nishimoto I, MatsuokaM (2005) Development of a femtomolar-acting humaninderivative named colivelin by attaching activity-dependentneurotrophic factor to its N terminus: Characterizationof colivelin-mediated neuroprotection against Alzheimer’sdisease-relevant insults in vitro and in vivo. J Neurosci 25,10252-10261.

[61] Chiba T, Nishimoto I, Aiso S, Matsuoka M (2007)Neuroprotection against neurodegenerative diseases:Development of a novel hybrid neuroprotective peptideColivelin. Mol Neurobiol 35, 55-84.

[62] Matsuoka M, Hashimoto Y, Aiso S, Nishimoto I (2006)Humanin and colivelin: Neuronal-death-suppressing pep-tides for Alzheimer’s disease and amyotrophic lateralsclerosis. CNS Drug Rev 12, 113-122.

[63] Yamada M, Chiba T, Sasabe J, Terashita K, Aiso S, Mat-suoka M (2008) Nasal Colivelin treatment amelioratesmemory impairment related to Alzheimer’s disease. Neu-ropsychopharmacology 33, 2020-2032.

[64] Arakawa T, Niikura T, Arisaka F, Kita Y (2008) Activity-dependent neurotrophic factor, ADNF, determines thestructure characteristics of Colivelin, a fusion protein ofADNF9 and Humanin analog. J Pept Sci 14, 631-636.

[65] Hashimoto Y, Ito Y, Niikura T, Shao Z, Hata M, OyamaF, Nishimoto I (2001) Mechanisms of neuroprotectionby a novel rescue factor humanin from Swedish mutantamyloid precursor protein. Biochem Biophys Res Commun283, 460-468.

[66] Maximov V, Martynenko A, Hunsmann G, Tarantul V(2002) Mitochondrial 16S rRNA gene encodes a functionalpeptide, a potential drug for Alzheimer’s disease and targetfor cancer therapy. Med Hypotheses 59, 670-673.

Page 57: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 57

[67] Tortosa E, Santa-Maria I, Moreno F, Lim F, Perez M, AvilaJ (2009) Binding of Hsp90 to tau promotes a conforma-tional change and aggregation of tau protein. J AlzheimersDis 17, 319-325.

[68] Salminen A, Ojala J, Kaarniranta K, Hiltunen M, Soini-nen H (2011) Hsp90 regulates tau pathology throughco-chaperone complexes in Alzheimer’s disease. ProgNeurobiol 93, 99-110.

[69] Vilenchik M, Solit D, Basso A, Huezo H, Lucas B, He H,Rosen N, Spampinato C, Modrich P, Chiosis G (2004) Tar-geting wide-range oncogenic transformation via PU24FCl,a specific inhibitor of tumor Hsp90. Chem Biol 11, 787-797.

[70] He H, Zatorska D, Kim J, Aguirre J, Llauger L, She Y, WuN, Immormino RM, Gewirth DT, Chiosis G (2006) Identi-fication of potent water soluble purine-scaffold inhibitorsof the heat shock protein 90. J Med Chem 49, 381-390.

[71] Caldas-Lopes E, Cerchietti L, Ahn JH, Clement CC, Rob-les AI, Rodina A, Moulick K, Taldone T, Gozman A, GuoY, Wu N, de SE, White J, Gross SS, Ma Y, VarticovskiL, Melnick A, Chiosis G (2009) Hsp90 inhibitor PU-H71,a multimodal inhibitor of malignancy, induces completeresponses in triple-negative breast cancer models. ProcNatl Acad Sci U S A 106, 8368-8373.

[72] Luo W, Dou F, Rodina A, Chip S, Kim J, Zhao Q, MoulickK, Aguirre J, Wu N, Greengard P, Chiosis G (2007) Rolesof heat-shock protein 90 in maintaining and facilitatingthe neurodegenerative phenotype in tauopathies. Proc NatlAcad Sci U S A 104, 9511-9516.

[73] Cole P, Vasiliou S, Mormeno D, Rosa E (2009) Medicinalchemistry selections from the 237th American ChemicalSociety National Meeting & Exposition. Drugs Future 34,509-520.

[74] Taldone T, Chiosis G (2009) Purine-scaffold Hsp90inhibitors. Curr Top Med Chem 9, 1436-1446.

[75] Taldone T, Zatorska D, Patel PD, Zong H, Rodina A, AhnJH, Moulick K, Guzman ML, Chiosis G (2011) Design,synthesis, and evaluation of small molecule Hsp90 probes.Bioorg Med Chem 19, 2603-2614.

[76] Biamonte MA, Shi J, Hong K, Hurst DC, Zhang L, FanJ, Busch DJ, Karjian PL, Maldonado AA, Sensintaffar JL,Yang YC, Kamal A, Lough RE, Lundgren K, BurrowsFJ, Timony GA, Boehm MF, Kasibhatla SR (2006) Orallyactive purine-based inhibitors of the heat shock protein 90.J Med Chem 49, 817-828.

[77] Dickey CA, Dunmore J, Lu B, Wang JW, Lee WC, KamalA, Burrows F, Eckman C, Hutton M, Petrucelli L (2006)HSP induction mediates selective clearance of tau phos-phorylated at proline-directed Ser/Thr sites but not KXGS(MARK) sites. FASEB J 20, 753-755.

[78] Dickey CA, Kamal A, Lundgren K, Klosak N, Bailey RM,Dunmore J, Ash P, Shoraka S, Zlatkovic J, Eckman CB,Patterson C, Dickson DW, Nahman NS Jr, Hutton M, Bur-rows F, Petrucelli L (2007) The high-affinity HSP90-CHIPcomplex recognizes and selectively degrades phosphory-lated tau client proteins. J Clin Invest 117, 648-658.

[79] Oddo S, Caccamo A, Tseng B, Cheng D, Vasilevko V,Cribbs DH, LaFerla FM (2008) Blocking Abeta42 accu-mulation delays the onset and progression of tau pathologyvia the C terminus of heat shock protein70-interacting pro-tein: A mechanistic link between Abeta and tau pathology.J Neurosci 28, 12163-12175.

[80] Patterson KR, Ward SM, Combs B, Voss K, Kanaan NM,Morfini G, Brady ST, Gamblin TC, Binder LI (2011) Heatshock protein 70 prevents both tau aggregation and the

inhibitory effects of preexisting tau aggregates on fastaxonal transport. Biochemistry 50, 10300-10310.

[81] Wilhelmus MM, de Waal RM, Verbeek MM (2007) Heatshock proteins and amateur chaperones in amyloid-Betaaccumulation and clearance in Alzheimer’s disease. MolNeurobiol 35, 203-216.

[82] Evans CG, Chang L, Gestwicki JE (2010) Heat shock pro-tein 70 (hsp70) as an emerging drug target. J Med Chem53, 4585-4602.

[83] Yarbrough GG (1983) Minireview. Thyrotropin releasinghormone and CNS cholinergic neurons. Life Sci 33, 111-118.

[84] Mellow AM, Sunderland T, Cohen RM, Lawlor BA, HillJL, Newhouse PA, Cohen MR, Murphy DL (1989) Acuteeffects of high-dose thyrotropin releasing hormone infu-sions in Alzheimer’s disease. Psychopharmacology (Berl)98, 403-407.

[85] Molchan SE, Mellow AM, Lawlor BA, Weingartner HJ,Cohen RM, Cohen MR, Sunderland T (1990) TRH attenu-ates scopolamine-induced memory impairment in humans.Psychopharmacology (Berl) 100, 84-89.

[86] Khan A, Mirolo MH, Claypoole K, Bhang J, Cox G, HoritaA, Tucker G (1994) Effects of low-dose TRH on cognitivedeficits in the ECT postictal state. Am J Psychiatry 151,1694-1696.

[87] Urayama A, Yamada S, Hirano K, Deguchi Y,Kimura R (2001) Brain receptor binding characteris-tics and pharmacokinetic-pharmacodynamic analysis ofthyrotropin-releasing hormone analogues. Life Sci 70, 647-657.

[88] Urayama A, Yamada S, Kimura R, Zhang J, Watanabe Y(2002) Neuroprotective effect and brain receptor bindingof taltirelin, a novel thyrotropin-releasing hormone (TRH)analogue, in transient forebrain ischemia of C57BL/6Jmice. Life Sci 72, 601-607.

[89] Tanabe M, Tokuda Y, Takasu K, Ono K, Honda M,Ono H (2007) The synthetic TRH analogue taltirelinexerts modality-specific antinociceptive effects via distinctdescending monoaminergic systems. Br J Pharmacol 150,403-414.

[90] Grigorova M, Sherwin BB, Tulandi T (2006) Effectsof treatment with leuprolide acetate depot on workingmemory and executive functions in young premenopausalwomen. Psychoneuroendocrinology 31, 935-947.

[91] Bowen RL, Verdile G, Liu T, Parlow AF, Perry G,Smith MA, Martins RN, Atwood CS (2004) Luteiniz-ing hormone, a reproductive regulator that modulates theprocessing of amyloid-beta precursor protein and amyloid-beta deposition. J Biol Chem 279, 20539-20545.

[92] Casadesus G, Webber KM, Atwood CS, Pappolla MA,Perry G, Bowen RL, Smith MA (2006) Luteinizing hor-mone modulates cognition and amyloid-beta deposition inAlzheimer APP transgenic mice. Biochim Biophys Acta1762, 447-452.

[93] Casadesus G, Zhu X, Atwood CS, Webber KM, Perry G,Bowen RL, Smith MA (2004) Beyond estrogen: Targetinggonadotropin hormones in the treatment of Alzheimer’sdisease. Curr Drug Targets CNS Neurol Disord 3, 281-285.

[94] Casadesus G, Atwood CS, Zhu X, Hartzler AW, WebberKM, Perry G, Bowen RL, Smith MA (2005) Evidence forthe role of gonadotropin hormones in the development ofAlzheimer disease. Cell Mol Life Sci 62, 293-298.

[95] Meethal SV, Smith MA, Bowen RL, Atwood CS (2005)The gonadotropin connection in Alzheimer’s disease.Endocrine 26, 317-326.

Page 58: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

58 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[96] Wilson AC, Meethal SV, Bowen RL, Atwood CS (2007)Leuprolide acetate: A drug of diverse clinical applications.Expert Opin Investig Drugs 16, 1851-1863.

[97] Baker LD, Barsness SM, Borson S, Merriam GR, Fried-man SD, Craft S, Vitiello MV (2012) Effects of growthhormone-releasing hormone on cognitive function inadults with mild cognitive impairment and healthy olderadults: Results of a controlled trial. Arch Neurol 69, 1420-1429.

[98] Mori M, Iriuchijima T, Yamada M, Murakami M,Kobayashi S (1991) A novel TRH analog, YM14673, stim-ulates intracellular signaling systems in the brain morepotently than predicted by its pituitary actions. Res Com-mun Chem Pathol Pharmacol 71, 17-26.

[99] Yamamoto M, Ozawa Y, Takeuchi H (1993) Effects of YM-14673, a new thyrotropin-releasing hormone analogue, onimpaired learning of passive avoidance in mice. Arch IntPharmacodyn Ther 321, 5-13.

[100] Matsushita M, Yonemori F, Hamada A, Toide K, Iwata K(1995) Effect of JTP-2942, a novel thyrotropin-releasinghormone analogue, on pentobarbital-induced anesthesia inrats. Eur J Pharmacol 276, 177-182.

[101] Katsumata T, Katayama Y, Yonemori H, Muramatsu H,Otori T, Nishiyama Y, Yamada H, Nakamura H, TerashiA (2001) Delayed administration of JTP-2942, a novelthyrotropin-releasing hormone analogue, improves cere-bral blood flow and metabolism in rat postischaemic brain.Clin Exp Pharmacol Physiol 28, 48-54.

[102] Katsumata T, Katayama Y, Ootori T, Muramatsu H,Nishiyama Y, Nakamura H, Seta T, Terashi A (2001)Effect of long-term administration of JTP-2942, a novelthyrotropin-releasing hormone analogue, on neurologicaloutcome, local cerebral blood flow and glucose utiliza-tion in a rat focal cerebral ischemia. Brain Res 901,62-70.

[103] Itoh Y, Ogasawara T, Mushiroi T, Yamazaki A, Ukai Y,Kimura K (1994) Effect of NS-3, a thyrotropin-releasinghormone analog, on in vivo acetylcholine release in ratbrain: Regional differences and its sites of action. J Phar-macol Exp Ther 271, 884-890.

[104] Bristow LJ, Bennett GW (1989) Effect of chronic intra-accumbens administration of the TRH analogue CG3509on histamine-induced behaviour in the rat. Br J Pharmacol97, 745-752.

[105] Fone KC, Johnson JV, Bennett GW, Marsden CA (1989)Involvement of 5-HT2 receptors in the behaviours pro-duced by intrathecal administration of selected 5-HTagonists and the TRH analogue (CG 3509) to rats. Br JPharmacol 96, 599-608.

[106] Fone KC, Johnson JV, Marsden CA, Bennett GW(1989) Comparative behavioural and biochemical effectsof repeated intrathecal administration of thyrotrophin-releasing hormone (TRH) or two analogues of TRH inadult rats. Neuropharmacology 28, 867-875.

[107] Parnetti L, Ambrosoli L, Abate G, Azzini C, BalestreriR, Bartorelli L, Bordin A, Crepaldi G, Cristianini G,Cucinotta D, et al. (1995) Posatirelin for the treat-ment of late-onset Alzheimer’s disease: A double-blindmulticentre study vs citicoline and ascorbic acid. ActaNeurol Scand 92, 135-140.

[108] Parnetti L, Ambrosoli L, Agliati G, Caratozzolo P, FossatiL, Frattola L, Martucci N, Murri L, Nappi G, Puca FM, PoliA, Girardello R, Senin U (1996) Posatirelin in the treat-ment of vascular dementia: A double-blind multicentrestudy vs placebo. Acta Neurol Scand 93, 456-463.

[109] Gasbarrini G, Stefanini G, Addolorato G, Foschi F, Ricci C,Bertolotti P, Voltolini G, Bonavita E, Bertoncelli R, RenziG, Bianchini G, Bonaiuto S, Giannandrea E, Cavassini G,Mazzini V, Chioma V, Marzara G, D’Addetta G, TotaroG, Dalmonte E, Tassini D, Giungi F, De NC, Di FG,Tessitore A, Guadagnino M, Tessitore E, Spina P, LuppiM, Bignamini A, Peracino L, Fiorentino M, Beun-GarbeD, Poli A, Ambrosoli L, Girardello R (1998) Posatirelinfor the treatment of degenerative and vascular dementia:Results of explanatory and pragmatic efficacy analyses.Arch Gerontol Geriatr 26, 33-47.

[110] Suzuki T, Fujimoto K, Oohata H, Kawashima K (1989)Effects of TRH and DN-1417 on high potassium-evokedacetylcholine release from rat basal forebrain slices deter-mined directly by radioimmunoassay. Gen Pharmacol 20,239-242.

[111] Ballard TM, Hunter AJ, Bennett GW (1996) Effect ofa thyrotrophin-releasing hormone analogue, RX77368,on AMPA-induced septal-hippocampal lesioned rats inan operant delayed non-matching to position test. Psy-chopharmacology (Berl) 127, 265-275.

[112] Itkin A, Dupres V, Dufrene YF, Bechinger B, RuysschaertJM, Raussens V (2011) Calcium ions promote formation ofamyloid beta-peptide (1-40) oligomers causally implicatedin neuronal toxicity of Alzheimer’s disease. PLoS One 6,e18250.

[113] Kawahara M, Ohtsuka I, Yokoyama S, Kato-NegishiM, Sadakane Y (2011) Membrane incorporation, chan-nel formation, and disruption of calcium homeostasis byAlzheimer’s beta-amyloid protein. Int J Alzheimers Dis2011, 304583.

[114] Mezler M, Barghorn S, Schoemaker H, Gross G, Nimm-rich V (2012) A beta-amyloid oligomer directly modulatesP/Q-type calcium currents in Xenopus oocytes. Br J Phar-macol 165, 1572-1583.

[115] Chakroborty S, Kim J, Schneider C, Jacobson C, Molgo J,Stutzmann GE (2012) Early presynaptic and postsynapticcalcium signaling abnormalities mask underlying synapticdepression in presymptomatic Alzheimer’s disease mice.J Neurosci 32, 8341-8353.

[116] Woods NK, Padmanabhan J (2012) Neuronal calcium sig-naling and Alzheimer’s disease. Adv Exp Med Biol 740,1193-1217.

[117] Lim YA, Giese M, Shepherd C, Halliday G, KobayashiM, Takamatsu K, Staufenbiel M, Eckert A, Gotz J (2012)Role of hippocalcin in mediating Abeta toxicity. BiochimBiophys Acta 1822, 1247-1257.

[118] Chakroborty S, Stutzmann GE (2011) Early calciumdysregulation in Alzheimer’s disease: Setting the stagefor synaptic dysfunction. Sci China Life Sci 54,752-762.

[119] Corona C, Pensalfini A, Frazzini V, Sensi SL (2011) Newtherapeutic targets in Alzheimer’s disease: Brain deregu-lation of calcium and zinc. Cell Death Dis 2, e176.

[120] Fedrizzi L, Carafoli E (2011) Ca2+ dysfunction in neu-rodegenerative disorders: Alzheimer’s disease. Biofactors37, 189-196.

[121] Yagami T, Kohma H, Yamamoto Y (2012) L-type voltage-dependent calcium channels as therapeutic targets forneurodegenerative diseases. Curr Med Chem 19, 4816-4827.

[122] Bachmeier C, Beaulieu-Abdelahad D, Mullan M, ParisD (2011) Selective dihydropyiridine compounds facili-tate the clearance of beta-amyloid across the blood-brainbarrier. Eur J Pharmacol 659, 124-129.

Page 59: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 59

[123] Paris D, Bachmeier C, Patel N, Quadros A, Volmar CH,Laporte V, Ganey J, Beaulieu-Abdelahad D, it-Ghezala G,Crawford F, Mullan MJ (2011) Selective antihypertensivedihydropyridines lower Abeta accumulation by targetingboth the production and the clearance of Abeta across theblood-brain barrier. Mol Med 17, 149-162.

[124] Iwasaki K, Mishima K, Egashira N, Al-Khatib IH,Ishibashi D, Irie K, Kobayashi H, Egawa T, FujiwaraM (2003) Effect of nilvadipine on the cerebral ischemia-induced impairment of spatial memory and hippocampalapoptosis in rats. J Pharmacol Sci 93, 188-196.

[125] Iwasaki K, Egashira N, Takagaki Y, Yoshimitsu Y, Hatip-Al-Khatib I, Mishima K, Fujiwara M (2007) Nilvadipineprevents the impairment of spatial memory induced bycerebral ischemia combined with beta-amyloid in rats. BiolPharm Bull 30, 698-701.

[126] Paris D, Quadros A, Humphrey J, Patel N, CrescentiniR, Crawford F, Mullan M (2004) Nilvadipine antago-nizes both Abeta vasoactivity in isolated arteries, and thereduced cerebral blood flow in APPsw transgenic mice.Brain Res 999, 53-61.

[127] Kennelly SP, Abdullah L, Paris D, Parish J, MathuraV, Mullan M, Crawford F, Lawlor BA, Kenny RA(2011) Demonstration of safety in Alzheimer’s patientsfor intervention with an anti-hypertensive drug Nilvadip-ine: Results from a 6-week open label study. Int J GeriatrPsychiatry 26, 1038-1045.

[128] Kennelly S, Abdullah L, Kenny RA, Mathura V, LuisCA, Mouzon B, Crawford F, Mullan M, Lawlor B (2012)Apolipoprotein E genotype-specific short-term cognitivebenefits of treatment with the antihypertensive nilvadipinein Alzheimer’s patients–an open-label trial. Int J GeriatrPsychiatry 27, 415-422.

[129] Hanyu H, Hirao K, Shimizu S, Iwamoto T, Koizumi K,Abe K (2007) Favourable effects of nilvadipine on cogni-tive function and regional cerebral blood flow on SPECTin hypertensive patients with mild cognitive impairment.Nucl Med Commun 28, 281-287.

[130] Hanyu H, Hirao K, Shimizu S, Sato T, Kiuchi A, IwamotoT (2007) Nilvadipine prevents cognitive decline of patientswith mild cognitive impairment. Int J Geriatr Psychiatry22, 1264-1266.

[131] Matsuda H, Araki N, Kuji I, Ohkubo T, Imabayashi E, Shi-mazu K (2008) Effect of nilvadipine on regional cerebralblood flow in a patient with early Alzheimer disease. ClinNucl Med 33, 34-35.

[132] Moriguchi S, Shioda N, Yamamoto Y, Tagashira H,Fukunaga K (2012) The T-type voltage-gated calciumchannel as a molecular target of the novel cognitiveenhancer ST101: Enhancement of long-term potentiationand CaMKII autophosphorylation in rat cortical slices. JNeurochem 121, 44-53.

[133] Yamaguchi Y, Miyashita H, Tsunekawa H, MouriA, Kim HC, Saito K, Matsuno T, Kawashima S,Nabeshima T (2006) Effects of a novel cogni-tive enhancer, spiro[imidazo-[1,2-a]pyridine-3,2-indan]-2(3H)-one (ZSET1446), on learning impairments inducedby amyloid-beta1-40 in the rat. J Pharmacol Exp Ther 317,1079-1087.

[134] Shioda N, Yamamoto Y, Han F, Moriguchi S, YamaguchiY, Hino M, Fukunaga K (2010) A novel cogni-tive enhancer, ZSET1446/ST101, promotes hippocampalneurogenesis and ameliorates depressive behavior in olfac-tory bulbectomized mice. J Pharmacol Exp Ther 333,43-50.

[135] Anekonda TS, Quinn JF, Harris C, Frahler K, WadsworthTL, Woltjer RL (2011) L-type voltage-gated calcium chan-nel blockade with isradipine as a therapeutic strategy forAlzheimer’s disease. Neurobiol Dis 41, 62-70.

[136] Anekonda TS, Quinn JF (2011) Calcium channel block-ing as a therapeutic strategy for Alzheimer’s disease: Thecase for isradipine. Biochim Biophys Acta 1812, 1584-1590.

[137] Copenhaver PF, Anekonda TS, Musashe D, Robinson KM,Ramaker JM, Swanson TL, Wadsworth TL, KretzschmarD, Woltjer RL, Quinn JF (2011) A translational continuumof model systems for evaluating treatment strategies inAlzheimer’s disease: Isradipine as a candidate drug. DisModel Mech 4, 634-648.

[138] Saletu B, Darragh A, Salmon P, Coen R (1989) EEG brainmapping in evaluating the time-course of the central actionof DUP 996–a new acetylcholine releasing drug. Br J ClinPharmacol 28, 1-16.

[139] van Dyck CH, Lin CH, Robinson R, Cellar J, Smith EO,Nelson JC, Arnsten AF, Hoffer PB (1997) The acetyl-choline releaser linopirdine increases parietal regionalcerebral blood flow in Alzheimer’s disease. Psychophar-macology (Berl) 132, 217-226.

[140] Borjesson A, Karlsson T, Adolfsson R, Ronnlund M,Nilsson L (1999) Linopirdine (DUP 996): Cholinergictreatment of older adults using successive and non-successive tests. Neuropsychobiology 40, 78-85.

[141] Rose GM, Ong VS, Woodruff-Pak DS (2007) Efficacy ofMEM 1003, a novel calcium channel blocker, in delayand trace eyeblink conditioning in older rabbits. NeurobiolAging 28, 766-773.

[142] Denolle T, Sassano P, Allain H, tue-Ferrer D, Breton S,Cimarosti I, Ouatara B, Merienne M, Gandon JM (2002)Effects of nicardipine and clonidine on cognitive func-tions and electroencephalography in hypertensive patients.Fundam Clin Pharmacol 16, 527-535.

[143] Schwartz BL, Fay-McCarthy M, Kendrick K, Rosse RB,Deutsch SI (1997) Effects of nifedipine, a calcium channelantagonist, on cognitive function in schizophrenic patientswith tardive dyskinesia. Clin Neuropharmacol 20, 364-370.

[144] Ban TA, Morey L, Aguglia E, Azzarelli O, Balsano F,Marigliano V, Caglieris N, Sterlicchio M, Capurso A,Tomasi NA, (1990) Nimodipine in the treatment of old agedementias. Prog Neuropsychopharmacol Biol Psychiatry14, 525-551.

[145] Izquierdo I (1990) Nimodipine and the recovery of mem-ory. Trends Pharmacol Sci 11, 309-310.

[146] Tedeschi D (1991) Calcium regulation in brain aging bynimodipine: A multicenterctrial in Italy. Curr Ther Res 50,553-563.

[147] de Jonge MC, Traber J (1993) Nimodipine: Cognition,aging, and degeneration. Clin Neuropharmacol 16(Suppl1), S25-S30.

[148] Fischhof PK (1993) Divergent neuroprotective effects ofnimodipine in PDD and MID provide indirect evidence ofdisturbances in Ca2+ homeostasis in dementia. MethodsFind Exp Clin Pharmacol 15, 549-555.

[149] Parnetti L, Senin U, Carosi M, Baasch H (1993) Mentaldeterioration in old age: Results of two multicenter, clinicaltrials with nimodipine. The Nimodipine Study Group. ClinTher 15, 394-406.

[150] Bernhardt T, Kuebler J, Erzigkeit H (1995) Impairmentof cerebral function in old age: Nimodipine in generalpractice. Eur J Clin Res 7, 205-215.

Page 60: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

60 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[151] Fritze J, Walden J (1995) Clinical findings with nimodip-ine in dementia: Test of the calcium hypothesis. J NeuralTransm Suppl 46, 439-453.

[152] Sze KH, Sim TC, Wong E, Cheng S, Woo J (1998) Effectof nimodipine on memory after cerebral infarction. ActaNeurol Scand 97, 386-392.

[153] Forette F, Seux ML, Staessen JA, Thijs L, BirkenhagerWH, Babarskiene MR, Babeanu S, Bossini A, Gil-Extremera B, Girerd X, Laks T, Lilov E, Moisseyev V,TuomilehtoJ, Vanhanen H, Webster J, Yodfat Y, FagardR (1998) Prevention of dementia in randomised double-blind placebo-controlled Systolic Hypertension in Europe(Syst-Eur) trial. Lancet 352, 1347-1351.

[154] Forette F, Seux ML, Staessen JA, Thijs L, Babarskiene MR,Babeanu S, Bossini A, Fagard R, Gil-Extremera B, Laks T,Kobalava Z, Sarti C, Tuomilehto J, Vanhanen H, WebsterJ, Yodfat Y, Birkenhager WH (2002) The prevention ofdementia with antihypertensive treatment: New evidencefrom the Systolic Hypertension in Europe (Syst-Eur) study.Arch Intern Med 162, 2046-2052.

[155] 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.

[156] O’Neill MJ, Bath CP, Dell CP, Hicks CA, Gilmore J,Ambler SJ, Ward MA, Bleakman D (1997) Effects of Ca2+and Na+ channel inhibitors in vitro and in global cerebralischaemia in vivo. Eur J Pharmacol 332, 121-131.

[157] Paroczai M, Kiss B, Karpati E (1998) Effect of RGH-2716on learning and memory deficits of young and aged rats inwater-labyrinth. Brain Res Bull 45, 475-488.

[158] Fukumoto H, Kakihana M, Kaisho Y, Suno M (1997) Thenovel compound TDN-345 induces synthesis/secretion ofnerve growth factor in C6-10A glioma cells. Brain Res774, 87-93.

[159] Nakayama T, Nagisa Y, Imamoto T, Nagai Y (1997) Ben-eficial effects of TDN-345, a novel Ca2+ antagonist, onischemic brain injury and cerebral glucose metabolism inexperimental animal models with cerebrovascular lesions.Brain Res 762, 203-210.

[160] Bourinet E, Stotz SC, Spaetgens RL, Dayanithi G, LemosJ, Nargeot J, Zamponi GW (2001) Interaction of SNX482with domains III and IV inhibits activation gating ofalpha(1E) (Ca(V)2.3) calcium channels. Biophys J 81,79-88.

[161] Kohlmeier KA, Leonard CS (2006) Transmitter modula-tion of spike-evoked calcium transients in arousal relatedneurons: Muscarinic inhibition of SNX-482-sensitive cal-cium influx. Eur J Neurosci 23, 1151-1162.

[162] Matthews EA, Bee LA, Stephens GJ, Dickenson AH(2007) The Cav2.3 calcium channel antagonist SNX-482reduces dorsal horn neuronal responses in a rat modelof chronic neuropathic pain. Eur J Neurosci 25, 3561-3569.

[163] Miyake N, Wakamori M, Akaike N (1992) A new typeof Ca2+ channel blocker, NC-1100, inhibits the low- andhigh-threshold Ca2+ currents in the rat CNS neurons.Brain Res 598, 215-220.

[164] Miyake N, Fujita R, Ishikawa M, Takayanagi M,Takayanagi Y, Sasaki K (2000) Reversal of multidrug resis-tance in human leukemia K562 by tamolarizine, a novelcalcium antagonist. Jpn J Pharmacol 82, 265-268.

[165] Tamura R, Nakada Y, Nishijo H, Miyake N, Ono T (2000)Ameliorative effects of tamolarizine on place learning

impairment induced by transient forebrain ischemia in rats.Brain Res 853, 81-92.

[166] Zaczek R, Chorvat RJ, Saye JA, Pierdomenico ME,Maciag CM, Logue AR, Fisher BN, Rominger DH,Earl RA (1998) Two new potent neurotransmitterrelease enhancers, 10,10-bis(4-pyridinylmethyl)-9(10H)-anthracenone and 10,10-bis(2-fluoro-4-pyridinylmethyl)-9(10H)-anthracenone: Comparison to linopirdine. JPharmacol Exp Ther 285, 724-730.

[167] MacVinish LJ, Guo Y, Dixon AK, Murrell-Lagnado RD,Cuthbert AW (2001) Xe991 reveals differences in K(+)channels regulating chloride secretion in murine air-way and colonic epithelium. Mol Pharmacol 60, 753-760.

[168] Schindowski K, Belarbi K, Buee L (2008) Neurotrophicfactors in Alzheimer’s disease: Role of axonal transport.Genes Brain Behav 7(Suppl 1), 43-56.

[169] Saragovi HU, Hamel E, Di PA (2009) A neurotrophic ratio-nale for the therapy of neurodegenerative disorders. CurrAlzheimer Res 6, 419-423.

[170] Lu Y, Christian K, Lu B (2008) BDNF: A key regulator forprotein synthesis-dependent LTP and long-term memory?Neurobiol Learn Mem 89, 312-323.

[171] Nagahara AH, Tuszynski MH (2011) Potential therapeuticuses of BDNF in neurological and psychiatric disorders.Nat Rev Drug Discov 10, 209-219.

[172] Fumagalli F, Racagni G, Riva MA (2006) The expand-ing role of BDNF: A therapeutic target for Alzheimer’sdisease? Pharmacogenomics J 6, 8-15.

[173] Zhang F, Kang Z, Li W, Xiao Z, Zhou X (2012) Rolesof brain-derived neurotrophic factor/tropomyosin-relatedkinase B (BDNF/TrkB) signalling in Alzheimer’s disease.J Clin Neurosci 19, 946-949.

[174] Castello NA, Green KN, Laferla FM (2012) Geneticknockdown of brain-derived neurotrophic factor in 3xTg-AD mice does not alter Abeta or tau pathology. PLoS One7, e39566.

[175] Capsoni S, Cattaneo A (2006) On the molecular basis link-ing nerve growth factor (NGF) to Alzheimer’s disease. CellMol Neurobiol 26, 619-633.

[176] Capsoni S, Tiveron C, Amato G, Vignone D, CattaneoA (2010) Peripheral neutralization of nerve growth factorinduces immunosympathectomy and central neurodegen-eration in transgenic mice. J Alzheimers Dis 20, 527-546.

[177] Capsoni S, Brandi R, Arisi I, D’Onofrio M, Catta-neo A (2011) A dual mechanism linking NGF/proNGFimbalance and early inflammation to Alzheimer’s diseaseneurodegeneration in the AD11 anti-NGF mouse model.CNS Neurol Disord Drug Targets 10, 635-647.

[178] Cuello AC (2012) Gangliosides, NGF, brain aging and dis-ease: A mini-review with personal reflections. NeurochemRes 37, 1256-1260.

[179] Cuello AC, Bruno MA, Bell KF (2007) NGF-cholinergicdependency in brain aging, MCI and Alzheimer’s disease.Curr Alzheimer Res 4, 351-358.

[180] Cuello AC, Bruno MA, Allard S, Leon W, Iulita MF (2010)Cholinergic involvement in Alzheimer’s disease. A linkwith NGF maturation and degradation. J Mol Neurosci 40,230-235.

[181] Cuello AC, Ferretti MT, Iulita MF (2012) Preplaque (’pre-clinical’) Abeta-induced inflammation and nerve growthfactor deregulation in transgenic models of Alzheimer’sdisease-like amyloid pathology. Neurodegener Dis 10,104-107.

Page 61: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 61

[182] Jonhagen ME (2000) Nerve growth factor treatmentin dementia. Alzheimer Dis Assoc Disord 14(Suppl 1),S31-S38.

[183] Cattaneo A, Calissano P (2012) Nerve growth factor andAlzheimer’s disease: New facts for an old hypothesis. MolNeurobiol 46, 588-604.

[184] Wang SH, Liao XM, Liu D, Hu J, Yin YY, Wang JZ, ZhuLQ (2012) NGF promotes long-term memory formationby activating poly(ADP-ribose)polymerase-1. Neurophar-macology 63, 1085-1092.

[185] Bishop KM, Hofer EK, Mehta A, Ramirez A, Sun L,Tuszynski M, Bartus RT (2008) Therapeutic potential ofCERE-110 (AAV2-NGF): Targeted, stable, and sustainedNGF delivery and trophic activity on rodent basal forebraincholinergic neurons. Exp Neurol 211, 574-584.

[186] Mandel RJ (2010) CERE-110, an adeno-associated virus-based gene delivery vector expressing human nerve growthfactor for the treatment of Alzheimer’s disease. Curr OpinMol Ther 12, 240-247.

[187] Jain P, Li R, Lama T, Saragovi HU, Cumberlidge G,Meerovitch K (2011) An NGF mimetic, MIM-D3, stim-ulates conjunctival cell glycoconjugate secretion anddemonstrates therapeutic efficacy in a rat model of dryeye. Exp Eye Res 93, 503-512.

[188] Visanji NP, Orsi A, Johnston TH, Howson PA, Dixon K,Callizot N, Brotchie JM, Rees DD (2008) PYM50028, anovel, orally active, nonpeptide neurotrophic factor in-ducer, prevents and reverses neuronal damage inducedby MPP+ in mesencephalic neurons and by MPTP in amouse model of Parkinson’s disease. FASEB J 22, 2488-2497.

[189] Fukushima T, Nakamura A, Iwakami N, Nakada Y, Hat-tori H, Hoki S, Yamaguchi H, Nakagawa M, TerashimaN, Narita H (2011) T-817MA, a neuroprotective agent,attenuates the motor and cognitive impairments associatedwith neuronal degeneration in P301L tau transgenic mice.Biochem Biophys Res Commun 407, 730-734.

[190] Nguyen PT, Kimura T, Ho SA, Tran AH, Ono T, Nishijo H(2007) Ameliorative effects of a neuroprotective agent, T-817MA, on place learning deficits induced by continuousinfusion of amyloid-beta peptide (1-40) in rats. Hippocam-pus 17, 443-455.

[191] Kimura T, Hong Nguyen PT, Ho SA, Tran AH, Ono T,Nishijo H (2009) T-817MA, a neurotrophic agent, amelio-rates the deficits in adult neurogenesis and spatial memoryin rats infused i.c.v. with amyloid-beta peptide. Br J Phar-macol 157, 451-463.

[192] Hirata K, Yamaguchi H, Takamura Y, Takagi A,Fukushima T, Iwakami N, Saitoh A, Nakagawa M, YamadaT (2005) A novel neurotrophic agent, T-817MA [1-{3-[2-(1-benzothiophen-5-yl) ethoxy] propyl}-3-azetidinolmaleate], attenuates amyloid-beta-induced neurotoxicityand promotes neurite outgrowth in rat cultured central ner-vous system neurons. J Pharmacol Exp Ther 314, 252-259.

[193] Wahlberg LU, Lind G, Almqvist PM, Kusk P, Tor-noe J, Juliusson B, Soderman M, Sellden E, Seiger A,Eriksdotter-Jonhagen M, Linderoth B (2012) Targeteddelivery of nerve growth factor via encapsulated cell biode-livery in Alzheimer disease: A technology platform forrestorative neurosurgery. J Neurosurg 117, 340-347.

[194] Voutilainen MH, Back S, Porsti E, Toppinen L, Lind-gren L, Lindholm P, Peranen J, Saarma M, Tuominen RK(2009) Mesencephalic astrocyte-derived neurotrophic fac-tor is neurorestorative in rat model of Parkinson’s disease.J Neurosci 29, 9651-9659.

[195] Glembotski CC, Thuerauf DJ, Huang C, VekichJA, Gottlieb RA, Doroudgar S (2012) Mesencephalicastrocyte-derived neurotrophic factor protects the heartfrom ischemic damage and is selectively secreted uponsarco/endoplasmic reticulum calcium depletion. J BiolChem 287, 25893-25904.

[196] Devi L, Ohno M (2012) 7,8-dihydroxyflavone, a small-molecule TrkB agonist, reverses memory deficits andBACE1 elevation in a mouse model of Alzheimer’s dis-ease. Neuropsychopharmacology 37, 434-444.

[197] Jang SW, Liu X, Yepes M, Shepherd KR, Miller GW, LiuY, Wilson WD, Xiao G, Blanchi B, Sun YE, Ye K (2010) Aselective TrkB agonist with potent neurotrophic activitiesby 7,8-dihydroxyflavone. Proc Natl Acad Sci U S A 107,2687-2692.

[198] Johnson RA, Lam M, Punzo AM, Li H, Lin BR, YeK, Mitchell GS, Chang Q (2012) 7,8-dihydroxyflavoneexhibits therapeutic efficacy in a mouse model of Rettsyndrome. J Appl Physiol 112, 704-710.

[199] Liu X, Chan CB, Jang SW, Pradoldej S, Huang J, He K,Phun LH, France S, Xiao G, Jia Y, Luo HR, Ye K (2010) Asynthetic 7,8-dihydroxyflavone derivative promotes neu-rogenesis and exhibits potent antidepressant effect. J MedChem 53, 8274-8286.

[200] Jang SW, Okada M, Sayeed I, Xiao G, Stein D, Jin P, YeK (2007) Gambogic amide, a selective agonist for TrkAreceptor that possesses robust neurotrophic activity, pre-vents neuronal cell death. Proc Natl Acad Sci U S A 104,16329-16334.

[201] Jang SW, Liu X, Chan CB, France SA, Sayeed I, TangW, Lin X, Xiao G, Andero R, Chang Q, Ressler KJ, YeK (2010) Deoxygedunin, a natural product with potentneurotrophic activity in mice. PLoS One 5, e11528.

[202] Nitta A, Ito M, Fukumitsu H, Ohmiya M, Ito H,Sometani A, Nomoto H, Furukawa Y, Furukawa S (1999)4-methylcatechol increases brain-derived neurotrophicfactor content and mRNA expression in cultured braincells and in rat brain in vivo. J Pharmacol Exp Ther 291,1276-1283.

[203] Sun MK, Alkon DL (2008) Effects of 4-methylcatechol onspatial memory and depression. Neuroreport 19, 355-359.

[204] Labie C, Canolle B, Chatelin S, Lafon C, Fournier J (2006)Effects of paliroden (SR57667B) and xaliproden on adultbrain neurogenesis. Curr Alzheimer Res 3, 35-36.

[205] Douillet P, Orgogozo JM (2009) What we have learnedfrom the Xaliproden Sanofi-aventis trials. J Nutr HealthAging 13, 365-366.

[206] Porzner M, Muller T, Seufferlein T (2009) SR 57746A/xaliproden, a non-peptide neurotrophic compound:Prospects and constraints for the treatment of nervoussystem diseases. Expert Opin Investig Drugs 18, 1765-1772.

[207] Mori T, Town T, TanJ, Yada N, Horikoshi Y, YamamotoJ, Shimoda T, Kamanaka Y, Tateishi N, Asano T (2006)Arundic Acid ameliorates cerebral amyloidosis and gliosisin Alzheimer transgenic mice. J Pharmacol Exp Ther 318,571-578.

[208] Pettigrew LC, Kasner SE, Albers GW, Gorman M, GrottaJC, Sherman DG, Funakoshi Y, Ishibashi H (2006) Safetyand tolerability of arundic acid in acute ischemic stroke. JNeurol Sci 251, 50-56.

[209] Pettigrew LC, Kasner SE, Gorman M, Atkinson RP,Funakoshi Y, Ishibashi H (2006) Effect of arundic acidon serum S-100beta in ischemic stroke. J Neurol Sci 251,57-61.

Page 62: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

62 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[210] Miyamoto Y, Nakahara M, Motoyama K, Ishiguro T,Oda Y, Yamanoi T, Okamoto I, Yagi A, Nishimura H,Hirayama F, Uekama K, Arima H (2011) Improvement ofsome physicochemical properties of arundic acid, (R)-(-)-2-propyloctanonic acid, by complexation with hydrophiliccyclodextrins. Int J Pharm 413, 63-72.

[211] Glasky AJ, Melchior CL, Pirzadeh B, Heydari N, Ritz-mann RF (1994) Effect of AIT-082, a purine analog, onworking memory in normal and aged mice. PharmacolBiochem Behav 47, 325-329.

[212] Glasky AJ, Glasky MS, Ritzmann RF, Rathbone MP(1997) AIT-082, a novel purine derivative with neu-roregenerative properties. Expert Opin Investig Drugs 6,1413-1417.

[213] Middlemiss PJ, Glasky AJ, Rathbone MP, WerstuikE, Hindley S, Gysbers J (1995) AIT-082, a uniquepurine derivative, enhances nerve growth factor mediatedneurite outgrowth from PC12 cells. Neurosci Lett 199,131-134.

[214] Rathbone MP, Middlemiss PJ, Crocker CE, Glasky MS,Juurlink BH, Ramirez JJ, Ciccarelli R, Di IP, Caciagli F(1999) AIT-082 as a potential neuroprotective and regen-erative agent in stroke and central nervous system injury.Expert Opin Investig Drugs 8, 1255-1262.

[215] Lahiri DK, Ge YW, Farlow MR (2000) Effect of a memory-enhancing drug, AIT-082, on the level of synaptophysin.Ann N Y Acad Sci 903, 387-393.

[216] Taylor EM, Yan R, Hauptmann N, Maher TJ, DjahandidehD, Glasky AJ (2000) AIT-082, a cognitive enhancer, istransported into brain by a nonsaturable influx mecha-nism and out of brain by a saturable efflux mechanism.J Pharmacol Exp Ther 293, 813-821.

[217] Westlund KN, Lu Y, Werrbach-Perez K, Hulsebosch CE,Morgan B, Pizzo DP, Eisenberg HM, Perez-Polo JR (1992)Effects of nerve growth factor and acetyl-L-carnitinearginyl amide on the human neuronal line HCN-1A. IntJ Dev Neurosci 10, 361-373.

[218] Taglialatela G, Navarra D, Olivi A, Ramacci MT,Werrbach-Perez K, Perez-Polo JR, Angelucci L (1995)Neurite outgrowth in PC12 cells stimulated by acetyl-L-carnitine arginine amide. Neurochem Res 20, 1-9.

[219] Scorziello A, Meucci O, Calvani M, Schettini G(1997) Acetyl-L-carnitine arginine amide prevents beta25–35-induced neurotoxicity in cerebellar granule cells.Neurochem Res 22, 257-265.

[220] Ono S, Kitamura K, Maekawa M, Hirata K, AnoM, Ukai W, Yamafuji T, Narita H (1993) Protectiveeffect of R(-)-1-(benzo[b]thiophen-5-yl)- 2-[2-(N,N-diethylamino)ethoxy]ethanol hydrochloride (T-588), anovel cerebral activator, against experimental cerebralanoxia. Jpn J Pharmacol 62, 81-86.

[221] Miyazaki H, Murayama T, Ono S, Narita H, NomuraY (1997) Effects of R(-)-1-(benzo[b]thiophen-5-yl)-2-[2-N,N-diethylamino)ethoxy]ethan ol hydrochloride (T-588),a novel cognitive enhancer, on noradrenaline release inrat cerebral cortical slices. Biochem Pharmacol 53, 1263-1269.

[222] Maekawa M, Murayama T, Ono S, Narita H, Nomura Y(1998) The effects of T-588, a novel cognitive enhancer,on noradrenaline uptake and release in rat cerebral corticalslices. Jpn J Pharmacol 77, 155-160.

[223] Iwasaki Y, Ikeda K, Ichikawa Y, Igarashi O, Kinoshita M,Marubuchi S, Ono S (2002) T-588 enhances neurite out-growth and choline acetyltransferase in cultured rat spinalventral horn neurons. Neurochem Res 27, 225-228.

[224] Iwasaki Y, Ichikawa Y, Igarasi O, Aoyagi J, Konno S, IkedaK, Iguchi H, Kawabe S, Marubuchi S, Ono S (2003) T-588protects motor neuron death against glutamate-inducedneurotoxicity. Neurochem Res 28, 1829-1832.

[225] Iwasaki Y, Ichikawa Y, Igarashi O, Konno S, Aoyagi J,Ikeda K, Marabuchi S, Ono S, Iguchi H, Kawabe K,Fujioka T (2004) T-588 protects motor neuron death fol-lowing axotomy. Neurochem Res 29, 403-406.

[226] Yamamuro A, Ago Y, Maeda S, Sakai Y, Baba A, MatsudaT (2003) Protective effect of T-588 on toxic damage byserum deprivation and amyloid-beta protein in culturedneurons. J Pharmacol Sci 92, 153-156.

[227] Phuagphong P, Fukushima T, Hatanaka R, Tanaka K, BabaA, Matsuda T (2004) T-588, a cognitive enhancer, protectsagainst sodium nitroprusside-induced toxicity in culturedastrocytes. J Pharmacol Sci 95, 135-138.

[228] Advokat C (2010) What are the cognitive effectsof stimulant medications? Emphasis on adults withattention-deficit/hyperactivity disorder (ADHD). NeurosciBiobehav Rev 34, 1256-1266.

[229] Berridge CW, Devilbiss DM (2011) Psychostimulantsas cognitive enhancers: The prefrontal cortex, cate-cholamines, and attention-deficit/hyperactivity disorder.Biol Psychiatry 69, e101-e111.

[230] Berridge CW, Shumsky JS, Andrzejewski ME, McGaughyJA, Spencer RC, Devilbiss DM, Waterhouse BD (2012)Differential sensitivity to psychostimulants across pre-frontal cognitive tasks: Differential involvement ofnoradrenergic alpha(1)- and alpha(2)-receptors. Biol Psy-chiatry 71, 467-473.

[231] Dolder CR, Davis LN, McKinsey J (2010) Use of psychos-timulants in patients with dementia. Ann Pharmacother 44,1624-1632.

[232] Arnsten AF, Pliszka SR (2011) Catecholamine influenceson prefrontal cortical function: Relevance to treatment ofattention deficit/hyperactivity disorder and related disor-ders. Pharmacol Biochem Behav 99, 211-216.

[233] Bidwell LC, McClernon FJ, Kollins SH (2011) Cogni-tive enhancers for the treatment of ADHD. PharmacolBiochem Behav 99, 262-274.

[234] Husain M, Mehta MA (2011) Cognitive enhancement bydrugs in health and disease. Trends Cogn Sci 15, 28-36.

[235] Levin ED, Bushnell PJ, Rezvani AH (2011) Attention-modulating effects of cognitive enhancers. PharmacolBiochem Behav 99, 146-154.

[236] Sahakian BJ, Morein-Zamir S (2011) Neuroethical issuesin cognitive enhancement. J Psychopharmacol 25, 197-204.

[237] Ragan CI, Bard I, Singh I (2012) What should we do aboutstudent use of cognitive enhancers? An analysis of currentevidence. Neuropharmacology 64, 588-595.

[238] Wolkenberg SE, Sur C (2010) Recent progress in the dis-covery of non-sarcosine based GlyT1 inhibitors. Curr TopMed Chem 10, 170-186.

[239] Volkow ND, Wang GJ, Fowler JS, Ding YS (2005)Imaging the effects of methylphenidate on braindopamine: New model on its therapeutic actions forattention-deficit/hyperactivity disorder. Biol Psychiatry57, 1410-1415.

[240] Berridge CW, Devilbiss DM, Andrzejewski ME, Arn-sten AF, Kelley AE, Schmeichel B, Hamilton C, SpencerRC (2006) Methylphenidate preferentially increases cate-cholamine neurotransmission within the prefrontal cortexat low doses that enhance cognitive function. Biol Psychi-atry 60, 1111-1120.

Page 63: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 63

[241] Devilbiss DM, Berridge CW (2008) Cognition-enhancingdoses of methylphenidate preferentially increase pre-frontal cortex neuronal responsiveness. Biol Psychiatry 64,626-635.

[242] Volkow ND, Fowler JS, Wang GJ, Telang F, Logan J, WongC, Ma J, Pradhan K, Benveniste H, Swanson JM (2008)Methylphenidate decreased the amount of glucose neededby the brain to perform a cognitive task. PLoS One 3,e2017.

[243] Repantis D, Schlattmann P, Laisney O, Heuser I (2010)Modafinil and methylphenidate for neuroenhancement inhealthy individuals: A systematic review. Pharmacol Res62, 187-206.

[244] Prashad M, Kim HY, Lu Y, Liu Y, Har D, Repic O,Blacklock TJ, Giannousis P (1999) The First Enantios-elective Synthesis of (2R,2’R)-threo-(+)-MethylphenidateHydrochloride. J Org Chem 64, 1750-1753.

[245] Spencer TJ, Bonab AA, Dougherty DD, Mirto T, Martin J,Clarke A, Fischman AJ (2012) Understanding the centralpharmacokinetics of spheroidal oral drug absorption sys-tem (SODAS) dexmethylphenidate: A positron emissiontomography study of dopamine transporter receptor occu-pancy measured with C-11 altropane. J Clin Psychiatry73, 346-352.

[246] Schmitt KC, Reith ME (2011) The atypical stimulant andnootropic modafinil interacts with the dopamine trans-porter in a different manner than classical cocaine-likeinhibitors. PLoS One 6, e25790.

[247] Turner DC, Clark L, Dowson J, Robbins TW, Sahakian BJ(2004) Modafinil improves cognition and response inhibi-tion in adult attention-deficit/hyperactivity disorder. BiolPsychiatry 55, 1031-1040.

[248] Turner DC, Clark L, Pomarol-Clotet E, McKenna P,Robbins TW, Sahakian BJ (2004) Modafinil improvescognition and attentional set shifting in patients withchronic schizophrenia. Neuropsychopharmacology 29,1363-1373.

[249] Scoriels L, Barnett JH, Murray GK, Cherukuru S, Field-ing M, Cheng F, Lennox BR, Sahakian BJ, Jones PB(2011) Effects of modafinil on emotional processing infirst episode psychosis. Biol Psychiatry 69, 457-464.

[250] Scoriels L, Barnett JH, Soma PK, Sahakian BJ, Jones PB(2012) Effects of modafinil on cognitive functions in firstepisode psychosis. Psychopharmacology (Berl) 220, 249-258.

[251] Scoriels L, Jones PB, Sahakian BJ (2013) Modafinil effectson cognition and emotion in schizophrenia and its neuro-chemical modulation in the brain. Neuropharmacology 64,168-184.

[252] Rasetti R, Mattay VS, Stankevich B, Skjei K, Blasi G,Sambataro F, rrillaga-Romany IC, Goldberg TE, Calli-cott JH, Apud JA, Weinberger DR (2010) Modulatoryeffects of modafinil on neural circuits regulating emo-tion and cognition. Neuropsychopharmacology 35, 2101-2109.

[253] Muller U, Rowe JB, Rittman T, Lewis C, Robbins TW,Sahakian BJ (2013) Effects of modafinil on non-verbalcognition, task enjoyment and creative thinking in healthyvolunteers. Neuropharmacology 64, 490-495.

[254] Brady KT, Gray KM, Tolliver BK (2011) Cognitiveenhancers in the treatment of substance use disorders: Clin-ical evidence. Pharmacol Biochem Behav 99, 285-294.

[255] Kalechstein AD, Mahoney JJ III, Yoon JH, Bennett R, DeLa GR (2013) Modafinil, but not escitalopram, improvesworking memory and sustained attention in long-term,

high-dose cocaine users. Neuropharmacology 64, 472-478.

[256] Sutherland SM, Adler LA, Chen C, Smith MD, FeltnerDE (2012) An 8-week, randomized controlled trial of ato-moxetine, atomoxetine plus buspirone, or placebo in adultswith ADHD. J Clin Psychiatry 73, 445-450.

[257] Gehlert DR, Schober DA, Hemrick-Luecke SK, Krushin-ski J, Howbert JJ, Robertson DW, Fuller RW, Wong DT(1995) Novel halogenated analogs of tomoxetine that arepotent and selective inhibitors of norepinephrine uptake inbrain. Neurochem Int 26, 47-52.

[258] Chamberlain SR, Del CN, Dowson J, Muller U, ClarkL, Robbins TW, Sahakian BJ (2007) Atomoxetineimproved response inhibition in adults with attentiondeficit/hyperactivity disorder. Biol Psychiatry 62, 977-984.

[259] Chamberlain SR, Hampshire A, Muller U, Rubia K, DelCN, Craig K, Regenthal R, Suckling J, Roiser JP, GrantJE, Bullmore ET, Robbins TW, Sahakian BJ (2009) Ato-moxetine modulates right inferior frontal activation duringinhibitory control: A pharmacological functional magneticresonance imaging study. Biol Psychiatry 65, 550-555.

[260] Wigal SB, Wong AA, Jun A, Stehli A, Steinberg-Epstein R, Lerner MA (2012) Adverse events inmedication treatment-naive children with attention-deficit/hyperactivity disorder: Results from a small,controlled trial of lisdexamfetamine dimesylate. J ChildAdolesc Psychopharmacol 22, 149-156.

[261] Rothman RB, Baumann MH, Dersch CM, Romero DV,Rice KC, Carroll FI, Partilla JS (2001) Amphetamine-typecentral nervous system stimulants release norepinephrinemore potently than they release dopamine and serotonin.Synapse 39, 32-41.

[262] Kojima T, Niigata K, Fujikura T, Tachikawa S,Nozaki Y, Kagami S, Takahashi K (1985) Synthesesof (+/-)-2-[(inden-7-yloxy)methyl]morpholine hydrochlo-ride (YM-08054, indeloxazine hydrochloride) and itsderivatives with potential cerebral-activating and antide-pressive properties. Chem Pharm Bull (Tokyo) 33,3766-3774.

[263] Yamamoto M, Ooyama M, Ozawa Y, Okada M, TadaS, Yamaguchi T, Endo H (1993) Effects of indeloxazinehydrochloride, a cerebral activator, on passive avoidancelearning impaired by disruption of cholinergic transmis-sion in rats. Neuropharmacology 32, 695-701.

[264] Yamamoto M, Takahashi K, Ohyama M, Sasamata M, Yat-sugi S, Okada M, Endoh H (1994) Possible involvementof central cholinergic system in ameliorating effects ofindeloxazine, a cerebral activator, on disturbance of learn-ing behavior in rats. Prog Neuropsychopharmacol BiolPsychiatry 18, 603-613.

[265] Yamaguchi T, Ozawa Y, Suzuki M, Yamamoto M, Naka-mura T, Yamaura A (1996) Indeloxazine hydrochlorideimproves impairment of passive avoidance performanceafter fluid percussion brain injury in rats. Neuropharma-cology 35, 329-336.

[266] Yamaguchi T, Suzuki M, Yamamoto M (1997) Facili-tation of acetylcholine release in rat frontal cortex byindeloxazine hydrochloride: Involvement of endogenousserotonin and 5-HT4 receptors. Naunyn SchmiedebergsArch Pharmacol 356, 712-720.

[267] Yamaguchi T, Ohyama M, Suzuki M, Ozawa Y, HatanakaK, Hidaka K, Yamamoto M (1998) Neurochemical andbehavioral characterization of potential antidepressantproperties of indeloxazine hydrochloride. Neuropharma-cology 37, 1169-1176.

Page 64: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

64 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[268] Kurosawa Y, Degrauw TJ, Lindquist DM, Blanco VM,Pyne-Geithman GJ, Daikoku T, Chambers JB, Benoit SC,Clark JF (2012) Cyclocreatine treatment improves cogni-tion in mice with creatine transporter deficiency. J ClinInvest 122, 2837-2846.

[269] Sugane T, Tobe T, Hamaguchi W, Shimada I, MaenoK, Miyata J, Suzuki T, Kimizuka T, Kohara A, MoritaT, Doihara H, Saita K, Aota M, Furutani M, Shi-mada Y, Hamada N, Sakamoto S, Tsukamoto S (2011)Synthesis and biological evaluation of 3-biphenyl-4-yl-4-phenyl-4H-1,2,4-triazoles as novel glycine transporter 1inhibitors. J Med Chem 54, 387-391.

[270] Sugane T, Tobe T, Hamaguchi W, Shimada I, Maeno K,Miyata J, Suzuki T, Kimizuka T, Morita T, Sakamoto S,Tsukamoto S (2012) Synthesis and biological evaluationof (4H-1,2,4-triazol-4-yl)isoquinoline derivatives as selec-tive glycine transporter 1 inhibitors. Bioorg Med Chem 20,34-41.

[271] Erdo SL, Kiss B, Rosdy B (1981) Inhibition of dopamineuptake by a new psychostimulant mesocarb (Sydnocarb).Pol J Pharmacol Pharm 33, 141-147.

[272] Ganiev MM, Kharlamov AN, Raevskii KS, Guseinov DI(1987) Effect of sidnocarb on learning and memory. BiullEksp Biol Med 104, 453-454.

[273] Schmeichel BE, Zemlan FP, Berridge CW (2013) Aselective dopamine reuptake inhibitor improves prefrontalcortex-dependent cognitive function: Potential relevanceto attention deficit hyperactivity disorder. Neuropharma-cology 64, 321-328.

[274] Nic Dhonnchadha BA, Kantak KM (2011) Cognitiveenhancers for facilitating drug cue extinction: Insightsfrom animal models. Pharmacol Biochem Behav 99,229-244.

[275] Nic Dhonnchadha BA, Pinard E, Alberati D, WettsteinJG, Spealman RD, Kantak KM (2012) Inhibiting glycinetransporter-1 facilitates cocaine-cue extinction and atten-uates reacquisition of cocaine-seeking behavior. DrugAlcohol Depend 122, 119-126.

[276] Chow TW, Pollock BG, Milgram NW (2007) Potentialcognitive enhancing and disease modification effects ofSSRIs for Alzheimer’s disease. Neuropsychiatr Dis Treat3, 627-636.

[277] Cassano GB, Puca F, Scapicchio PL, Trabucchi M (2002)Paroxetine and fluoxetine effects on mood and cognitivefunctions in depressed nondemented elderly patients. JClin Psychiatry 63, 396-402.

[278] Pollock BG, Mulsant BH, Rosen J, Sweet RA, Mazum-dar S, Bharucha A, Marin R, Jacob NJ, Huber KA,Kastango KB, Chew ML (2002) Comparison of citalo-pram, perphenazine, and placebo for the acute treatmentof psychosis and behavioral disturbances in hospital-ized, demented patients. Am J Psychiatry 159, 460-465.

[279] Krohn M, Lange C, Hofrichter J, Scheffler K, Stenzel J,Steffen J, Schumacher T, Bruning T, Plath AS, Alfen F,Schmidt A, Winter F, Rateitschak K, Wree A, Gsponer J,Walker LC, Pahnke J (2011) Cerebral amyloid-beta pro-teostasis is regulated by the membrane transport proteinABCC1 in mice. J Clin Invest 121, 3924-3931.

[280] Harada K, Nakato K, Yarimizu J, Yamazaki M, Morita M,Takahashi S, Aota M, Saita K, Doihara H, Sato Y, YamajiT, Ni K, Matsuoka N (2012) A novel glycine transporter-1(GlyT1) inhibitor, ASP2535 (4-[3-isopropyl-5-(6-phenyl-3-pyridyl)-4H-1,2,4-triazol-4-yl]-2,1,3-benzoxadiazol e),improves cognition in animal models of cognitive impair-

ment in schizophrenia and Alzheimer’s disease. Eur JPharmacol 685, 59-69.

[281] Murai S, Saito H, Abe E, Masuda Y, Odashima J, ItohT (1994) MKC-231, a choline uptake enhancer, amelio-rates working memory deficits and decreased hippocampalacetylcholine induced by ethylcholine aziridinium ion inmice. J Neural Transm Gen Sect 98, 1-13.

[282] Bessho T, Takashina K, Tabata R, Ohshima C, Chaki H,Yamabe H, Egawa M, Tobe A, Saito K (1996) Effectof the novel high affinity choline uptake enhancer 2-(2-oxopyrrolidin-1-yl)-N-(2,3-dimethyl-5,6,7,8- tetrahydro-furo [2,3-b] quinolin-4-yl)acetoamide on deficits of watermaze learning in rats. Arzneimittelforschung 46, 369-373.

[283] Bessho T, Takashina K, Eguchi J, Komatsu T, SaitoK (2008) MKC-231, a choline-uptake enhancer: (1)long-lasting cognitive improvement after repeated admin-istration in AF64A-treated rats. J Neural Transm 115,1019-1025.

[284] Takashina K, Bessho T, Mori R, Eguchi J, Saito K (2008)MKC-231, a choline uptake enhancer: (2) Effect on syn-thesis and release of acetylcholine in AF64A-treated rats.J Neural Transm 115, 1027-1035.

[285] Takashina K, Bessho T, Mori R, Kawai K, Eguchi J, SaitoK (2008) MKC-231, a choline uptake enhancer: (3) Modeof action of MKC-231 in the enhancement of high-affinitycholine uptake. J Neural Transm 115, 1037-1046.

[286] Akaike A, Maeda T, Kaneko S, Tamura Y (1998) Protectiveeffect of MKC-231, a novel high affinity choline uptakeenhancer, on glutamate cytotoxicity in cultured corticalneurons. Jpn J Pharmacol 76, 219-222.

[287] Uemura K, Yoshioka S, Surina-Baumgartner DM, Tama-gawa T, Miura H, Ueda M, Tamaya N, Iguchi A, Hotta N(1999) Central nervous system-mediated hyperglycemiceffects of NIK-247, a cholinesterase inhibitor, and MKC-231, a choline uptake enhancer, in rats. Jpn J Pharmacol79, 113-115.

[288] Shirayama Y, Yamamoto A, Nishimura T, Katayama S,Kawahara R (2007) Subsequent exposure to the cholineuptake enhancer MKC-231 antagonizes phencyclidine-induced behavioral deficits and reduction in septalcholinergic neurons in rats. Eur Neuropsychopharmacol17, 616-626.

[289] Shimazaki T, Kaku A, Chaki S (2010) D-Serine and aglycine transporter-1 inhibitor enhance social memory inrats. Psychopharmacology (Berl) 209, 263-270.

[290] Boulay D, Pichat P, Dargazanli G, Estenne-Bouhtou G,Terranova JP, Rogacki N, Stemmelin J, Coste A, LanneauC, Desvignes C, Cohen C, Alonso R, Vige X, Biton B,Steinberg R, Sevrin M, Oury-Donat F, George P, BergisO, Griebel G, Avenet P, Scatton B (2008) Characteriza-tion of SSR103800, a selective inhibitor of the glycinetransporter-1 in models predictive of therapeutic activityin schizophrenia. Pharmacol Biochem Behav 91, 47-58.

[291] Boulay D, Bergis O, Avenet P, Griebel G (2010) Theglycine transporter-1 inhibitor SSR103800 displays aselective and specific antipsychotic-like profile in normaland transgenic mice. Neuropsychopharmacology 35, 416-427.

[292] Black MD, Varty GB, Arad M, Barak S, De LA, BoulayD, Pichat P, Griebel G, Weiner I (2009) Procognitive andantipsychotic efficacy of glycine transport 1 inhibitors(GlyT1) in acute and neurodevelopmental models ofschizophrenia: Latent inhibition studies in the rat. Psy-chopharmacology (Berl) 202, 385-396.

Page 65: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 65

[293] Depoortere R, Dargazanli G, Estenne-Bouhtou G, CosteA, Lanneau C, Desvignes C, Poncelet M, Heaulme M,Santucci V, Decobert M, Cudennec A, Voltz C, Boulay D,Terranova JP, StemmelinJ, Roger P, Marabout B, SevrinM, Vige X, Biton B, Steinberg R, Francon D, Alonso R,Avenet P, Oury-Donat F, Perrault G, Griebel G, GeorgeP, Soubrie P, Scatton B (2005) Neurochemical, electro-physiological and pharmacological profiles of the selectiveinhibitor of the glycine transporter-1 SSR504734, a poten-tial new type of antipsychotic. Neuropsychopharmacology30, 1963-1985.

[294] Singer P, Feldon J, Yee BK (2009) The glycine transporter1 inhibitor SSR504734 enhances working memory perfor-mance in a continuous delayed alternation task in C57BL/6mice. Psychopharmacology (Berl) 202, 371-384.

[295] Singer P, Feldon J, Yee BK (2009) Interactions betweenthe glycine transporter 1(GlyT1) inhibitor SSR504734 andpsychoactive drugs in mouse motor behaviour. Eur Neu-ropsychopharmacol 19, 571-580.

[296] Nishikawa H, Inoue T, Izumi T, Nakagawa S, KoyamaT (2010) SSR504734, a glycine transporter-1 inhibitor,attenuates acquisition and expression of contextual con-ditioned fear in rats. Behav Pharmacol 21, 576-579.

[297] Nikiforuk A, Kos T, Rafa D, Behl B, Bespalov A, Popik P(2011) Blockade of glycine transporter 1 by SSR-504734promotes cognitive flexibility in glycine/NMDA receptor-dependent manner. Neuropharmacology 61, 262-267.

[298] (1999) Sufoxazine. Lucelan, Metatone, teniloxazine, Y8894. Drugs R D 2, 66-67.

[299] Anami K, Yamamoto Y, Setoguchi M (1985) Pharma-cological studies on sufoxazine (Y-8894). (I) Effects onexperimental amnesia in mice. Nihon Yakurigaku Zasshi85, 71-77.

[300] Anami K, Yamamoto Y, Setoguchi M, Maruyama Y (1987)Pharmacological studies on Y-8894. (V) Effect on learningand memory in intact and experimentally amnesic rats.Nihon Yakurigaku Zasshi 89, 145-153.

[301] Anami K, Setoguchi M, Senoh H (1988) Pharmacologicalstudies on Y-8894. (VIII). Effects on learning and memoryin the radial maze task in mice. Nihon Yakurigaku Zasshi92, 113-118.

[302] Lehr T, Staab A, Trommeshauser D, Schaefer HG,Kloft C (2010) Quantitative pharmacology approach inAlzheimer’s disease: Efficacy modeling of early clinicaldata to predict clinical outcome of tesofensine. AAPS J12, 117-129.

[303] van de Giessen E, de Bruin K, la Fleur SE, van den BrinkW, Booij J (2012) Triple monoamine inhibitor tesofensinedecreases food intake, body weight, and striatal dopamineD2/D3 receptor availability in diet-induced obese rats. EurNeuropsychopharmacol 22, 290-299.

[304] Adam Y, Edwards RH, Schuldiner S (2008) Expressionand function of the rat vesicular monoamine transporter.Am J Physiol Cell Physiol 294, C1004-C1011.

[305] Koeppe RA, Gilman S, Joshi A, Liu S, Little R, JunckL, Heumann M, Frey KA, Albin RL (2005) 11C-DTBZand 18F-FDG PET measures in differentiating dementias.J Nucl Med 46, 936-944.

[306] Gilman S, Koeppe RA, Little R, An H, Junck L, GiordaniB, Persad C, Heumann M, Wernette K (2004) Stri-atal monoamine terminals in Lewy body dementia andAlzheimer’s disease. Ann Neurol 55, 774-780.

[307] Koeppe RA, Gilman S, Junck L, Wernette K, Frey KA(2008) Differentiating Alzheimer’s disease from demen-tia with Lewy bodies and Parkinson’s disease with

(+)-[11C]dihydrotetrabenazine positron emission tomog-raphy. Alzheimers Dement 4, S67-S76.

[308] Nagren K, Halldin C, Rinne JO (2010) Radiopharmaceu-ticals for positron emission tomography investigations ofAlzheimer’s disease. Eur J Nucl Med Mol Imaging 37,1575-1593.

[309] Zhu L, Liu Y, Plossl K, Lieberman B, Liu J, Kung HF(2010) An improved radiosynthesis of [18F]AV-133: APET imaging agent for vesicular monoamine transporter2. Nucl Med Biol 37, 133-141.

[310] Tsao HH, Lin KJ, Juang JH, Skovronsky DM, Yen TC,Wey SP, Kung MP (2010) Binding characteristics of 9-fluoropropyl-(+)-dihydrotetrabenzazine (AV-133) to thevesicular monoamine transporter type 2 in rats. Nucl MedBiol 37, 413-419.

[311] Wang JL, Oya S, Parhi AK, Lieberman BP, Ploessl K,Hou C, Kung HF (2010) In vivo studies of the SERT-selective [18F]FPBM and VMAT2-selective [18F]AV-133radiotracers in a rat model of Parkinson’s disease. NuclMed Biol 37, 479-486.

[312] Lin KJ, Weng YH, Wey SP, Hsiao IT, Lu CS, Skovron-sky D, Chang HP, Kung MP, Yen TC (2010) Whole-bodybiodistribution and radiation dosimetry of 18F-FP-(+)-DTBZ (18F-AV-133): A novel vesicular monoaminetransporter 2 imaging agent. J Nucl Med 51, 1480-1485.

[313] Okamura N, Villemagne VL, Drago J, Pejoska S, DhamijaRK, Mulligan RS, Ellis JR, Ackermann U, O’Keefe G,Jones G, Kung HF, Pontecorvo MJ, Skovronsky D, RoweCC (2010) In vivo measurement of vesicular monoaminetransporter type 2 density in Parkinson disease with (18)F-AV-133. J Nucl Med 51, 223-228.

[314] Villemagne VL, Okamura N, Pejoska S, Drago J, MulliganRS, Chetelat G, Ackermann U, O’Keefe G, Jones G, GongS, Tochon-Danguy H, Kung HF, Masters CL, Skovron-sky DM, Rowe CC (2011) In vivo assessment of vesicularmonoamine transporter type 2 in dementia with lewy bod-ies and Alzheimer disease. Arch Neurol 68, 905-912.

[315] Lin KJ, Lin WY, Hsieh CJ, Weng YH, Wey SP, Lu CS,Skovronsky D, Yen TC, Chang CJ, Kung MP, Hsiao IT(2011) Optimal scanning time window for 18F-FP-(+)-DTBZ (18F-AV-133) summed uptake measurements. NuclMed Biol 38, 1149-1155.

[316] Toomey JS, Bhatia S, Moon LT, Orchard EA, Tainter KH,Lokitz SJ, Terry T, Mathis JM, Penman AD (2012) PETimaging a MPTP-induced mouse model of Parkinson’s dis-ease using the fluoropropyl-dihydrotetrabenazine analog[(18)F]-DTBZ (AV-133). PLoS One 7, e39041.

[317] Rinne JO, Sahlberg N, Ruottinen H, Nagren K, LehikoinenP (1998) Striatal uptake of the dopamine reuptake ligand[11C]beta-CFT is reduced in Alzheimer’s disease assessedby positron emission tomography. Neurology 50, 152-156.

[318] Rinne JO, Bergman J, Ruottinen H, Haaparanta M, EronenE, Oikonen V, Sonninen P, Solin O (1999) Striatal uptakeof a novel PET ligand, [18F]beta-CFT, is reduced in earlyParkinson’s disease. Synapse 31, 119-124.

[319] Rinne JO, Nagren K (2010) Positron emission tomographyin at risk patients and in the progression of mild cognitiveimpairment to Alzheimer’s disease. J Alzheimers Dis 19,291-300.

[320] McKeith I, O’Brien J, Walker Z, Tatsch K, Booij J, Dar-court J, Padovani A, Giubbini R, Bonuccelli U, VolterraniD, Holmes C, Kemp P, Tabet N, Meyer I, Reininger C(2007) Sensitivity and specificity of dopamine transporterimaging with 123I-FP-CIT SPECT in dementia with Lewy

Page 66: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

66 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

bodies: A phase III, multicentre study. Lancet Neurol 6,305-313.

[321] Walker Z, Jaros E, Walker RW, Lee L, Costa DC, Liv-ingston G, Ince PG, Perry R, McKeith I, Katona CL(2007) Dementia with Lewy bodies: A comparison of clin-ical diagnosis, FP-CIT single photon emission computedtomography imaging and autopsy. J Neurol NeurosurgPsychiatry 78, 1176-1181.

[322] Vlaar AM, de NT, Kessels AG, Vreeling FW, WinogrodzkaA, Mess WH, Tromp SC, van Kroonenburgh MJ, WeberWE (2008) Diagnostic value of 123I-ioflupane and123I-iodobenzamide SPECT scans in 248 patients withparkinsonian syndromes. Eur Neurol 59, 258-266.

[323] Roselli F, Pisciotta NM, Perneczky R, Pennelli M, AnielloMS, De Caro MF, Ferrannini E, Tartaglione B, DefazioG, Rubini G, Livrea P (2009) Severity of neuropsychiatricsymptoms and dopamine transporter levels in dementiawith Lewy bodies: A 123I-FP-CIT SPECT study. MovDisord 24, 2097-2103.

[324] Roselli F, Pisciotta NM, Pennelli M, Aniello MS, GiganteA, De Caro MF, Ferrannini E, Tartaglione B, Niccoli-Asabella A, Defazio G, Livrea P, Rubini G (2010)Midbrain SERT in degenerative parkinsonisms: A 123I-FP-CIT SPECT study. Mov Disord 25, 1853-1859.

[325] Gerasimou G, Costa DC, Papanastasiou E, Bostan-jiopoulou S, Arnaoutoglou M, Moralidis E, AggelopoulouT, Gotzamani-Psarrakou A (2012) SPECT study withI-123-Ioflupane (DaTSCAN) in patients with essentialtremor. Is there any correlation with Parkinson’s disease?Ann Nucl Med 26, 337-344.

[326] Scott R, Bourtchuladze R, Gossweiler S, Dubnau J, TullyT (2002) CREB and the discovery of cognitive enhancers.J Mol Neurosci 19, 171-177.

[327] Saura CA, Valero J (2011) The role of CREB signalingin Alzheimer’s disease and other cognitive disorders. RevNeurosci 22, 153-169.

[328] Saura CA (2012) CREB-regulated transcription coactiva-tor 1-dependent transcription in Alzheimer’s disease mice.Neurodegener Dis 10, 250-252.

[329] Scott Bitner R (2012) Cyclic AMP response element-binding protein (CREB) phosphorylation: A mechanisticmarker in the development of memory enhancingAlzheimer’s disease therapeutics. Biochem Pharmacol 83,705-714.

[330] Oliveira AM, Bading H (2011) Calcium signaling in cog-nition and aging-dependent cognitive decline. Biofactors37, 168-174.

[331] Espana J, Valero J, Minano-Molina AJ, Masgrau R, Mar-tin E, Guardia-Laguarta C, Lleo A, Gimenez-Llort L,Rodriguez-Alvarez J, Saura CA (2010) beta-Amyloid dis-rupts activity-dependent gene transcription required formemory through the CREB coactivator CRTC1. J Neurosci30, 9402-9410.

[332] Helm KA, Haberman RP, Dean SL, Hoyt EC, Melcher T,Lund PK, Gallagher M (2005) GABAB receptor antago-nist SGS742 improves spatial memory and reduces proteinbinding to the cAMP response element (CRE) in the hip-pocampus. Neuropharmacology 48, 956-964.

[333] Florian C, Mons N, Roullet P (2006) CREB antisenseoligodeoxynucleotide administration into the dorsal hip-pocampal CA3 region impairs long- but not short-termspatial memory in mice. Learn Mem 13, 465-472.

[334] Fontan-Lozano A, Romero-Granados R, del-Pozo-MartinY, Suarez-Pereira I, gado-Garcia JM, Penninger JM, Car-rion AM (2009) Lack of DREAM protein enhances

learning and memory and slows brain aging. Curr Biol19, 54-60.

[335] Chow N, Bell RD, Deane R, Streb JW, Chen J, Brooks A,Van NW, Miano JM, Zlokovic BV (2007) Serum responsefactor and myocardin mediate arterial hypercontractil-ity and cerebral blood flow dysregulation in Alzheimer’sphenotype. Proc Natl Acad Sci U S A 104, 823-828.

[336] Bell RD (2012) The imbalance of vascular molecules inAlzheimer’s disease. J Alzheimers Dis 32, 699-709.

[337] McPhee I, Gibson LC, Kewney J, Darroch C, StevensPA, Spinks D, Cooreman A, Mackenzie SJ (2005) Cyclicnucleotide signalling: A molecular approach to drug dis-covery for Alzheimer’s disease. Biochem Soc Trans 33,1330-1332.

[338] Reddy PH, Beal MF (2008) Amyloid beta, mitochondrialdysfunction and synaptic damage: Implications for cogni-tive decline in aging and Alzheimer’s disease. Trends MolMed 14, 45-53.

[339] Di Bona D, Scapagnini G, Candore G, Castiglia L,Colonna-Romano G, Duro G, Nuzzo D, Iemolo F, LioD, Pellicano M, Scafidi V, Caruso C, Vasto S (2010)Immune-inflammatory responses and oxidative stressin Alzheimer’s disease: Therapeutic implications. CurrPharm Des 16, 684-691.

[340] Obrenovich ME, Li Y, Parvathaneni K, Yendluri BB, Pala-cios HH, Leszek J, Aliev G (2011) Antioxidants in health,disease and aging. CNS Neurol Disord Drug Targets 10,192-207.

[341] Pratico D (2008) Oxidative stress hypothesis inAlzheimer’s disease: A reappraisal. Trends Pharmacol Sci29, 609-615.

[342] Pratico D (2008) Evidence of oxidative stress inAlzheimer’s disease brain and antioxidant therapy: Lightsand shadows. Ann N Y Acad Sci 1147, 70-78.

[343] Young KJ, Bennett JP (2010) The mitochondrialsecret(ase) of Alzheimer’s disease. J Alzheimers Dis20(Suppl 2), S381-S400.

[344] Halliwell B (1999) Antioxidant defence mechanisms:From the beginning to the end (of the beginning). FreeRadic Res 31, 261-272.

[345] Halliwell B (2001) Role of free radicals in the neurodegen-erative diseases: Therapeutic implications for antioxidanttreatment. Drugs Aging 18, 685-716.

[346] Halliwell B (2006) Oxidative stress and neurodegenera-tion: Where are we now? J Neurochem 97, 1634-1658.

[347] Halliwell B (2011) Free radicals and antioxidants - quovadis? Trends Pharmacol Sci 32, 125-130.

[348] Lee HP, Zhu X, Casadesus G, Castellani RJ, NunomuraA, Smith MA, Lee HG, Perry G (2010) Antioxidantapproaches for the treatment of Alzheimer’s disease.Expert Rev Neurother 10, 1201-1208.

[349] Vina J, Lloret A, Giraldo E, Badia MC, Alonso MD (2011)Antioxidant pathways in Alzheimer’s disease: Possibilitiesof intervention. Curr Pharm Des 17, 3861-3864.

[350] Baum L, Lam CW, Cheung SK, Kwok T, Lui V, Tsoh J,Lam L, Leung V, Hui E, Ng C, Woo J, Chiu HF, Gog-gins WB, Zee BC, Cheng KF, Fong CY, Wong A, MokH, Chow MS, Ho PC, Ip SP, Ho CS, Yu XW, Lai CY,Chan MH, Szeto S, Chan IH, Mok V (2008) Six-monthrandomized, placebo-controlled, double-blind, pilot clini-cal trial of curcumin in patients with Alzheimer disease. JClin Psychopharmacol 28, 110-113.

[351] Halliwell B (1999) Vitamin C: Poison, prophylactic orpanacea? Trends Biochem Sci 24, 255-259.

Page 67: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 67

[352] Lloret A, Badia MC, Mora NJ, Pallardo FV, Alonso MD,Vina J (2009) Vitamin E paradox in Alzheimer’s disease:It does not prevent loss of cognition and may even bedetrimental. J Alzheimers Dis 17, 143-149.

[353] Brewer GJ (2010) Why vitamin E therapy fails for treat-ment of Alzheimer’s disease. J Alzheimers Dis 19, 27-30.

[354] Usoro OB, Mousa SA (2010) Vitamin E forms inAlzheimer’s disease: A review of controversial and clinicalexperiences. Crit Rev Food Sci Nutr 50, 414-419.

[355] Guan JZ, Guan WP, Maeda T, Makino N (2012) Effectof vitamin E administration on the elevated oxygen stressand the telomeric and subtelomeric status in Alzheimer’sdisease. Gerontology 58, 62-69.

[356] Behl C (1999) Alzheimer’s disease and oxidative stress:Implications for novel therapeutic approaches. Prog Neu-robiol 57, 301-323.

[357] Behl C (2005) Oxidative stress in Alzheimer’s disease:Implications for prevention and therapy. Subcell Biochem38, 65-78.

[358] Aliev G, Smith MA, Seyidov D, Neal ML, Lamb BT,Nunomura A, Gasimov EK, Vinters HV, Perry G, LamannaJC, Friedland RP (2002) The role of oxidative stress in thepathophysiology of cerebrovascular lesions in Alzheimer’sdisease. Brain Pathol 12, 21-35.

[359] Aliev G, Obrenovich ME, Reddy VP, Shenk JC, Mor-eira PI, Nunomura A, Zhu X, Smith MA, Perry G (2008)Antioxidant therapy in Alzheimer’s disease: Theory andpractice. Mini Rev Med Chem 8, 1395-1406.

[360] Aliev G, Palacios HH, Walrafen B, Lipsitt AE, Obren-ovich ME, Morales L (2009) Brain mitochondria as aprimary target in the development of treatment strategiesfor Alzheimer disease. Int J Biochem Cell Biol 41, 1989-2004.

[361] Moreira PI, Honda K, Liu Q, Aliev G, Oliveira CR, San-tos MS, Zhu X, Smith MA, Perry G (2005) Alzheimer’sdisease and oxidative stress: The old problem remainsunsolved. Curr Med Chem Central Nervous System Agents5, 51-62.

[362] Moreira PI, Santos MS, Oliveira CR (2007) Alzheimer’sdisease: A lesson from mitochondrial dysfunction.Antioxid Redox Signal 9, 1621-1630.

[363] Chauhan V, Chauhan A (2006) Oxidative stress inAlzheimer’s disease. Pathophysiology 13, 195-208.

[364] Nunomura A, Castellani RJ, Zhu X, Moreira PI, PerryG, Smith MA (2006) Involvement of oxidative stress inAlzheimer disease. J Neuropathol Exp Neurol 65, 631-641.

[365] Nunomura A, Moreira PI, Lee HG, Zhu X, Castel-lani RJ, Smith MA, Perry G (2007) Neuronal deathand survival under oxidative stress in Alzheimer andParkinson diseases. CNS Neurol Disord Drug Targets 6,411-423.

[366] Onyango IG, Khan SM (2006) Oxidative stress, mito-chondrial dysfunction, and stress signaling in Alzheimer’sdisease. Curr Alzheimer Res 3, 339-349.

[367] Liu Q, Xie F, Rolston R, Moreira PI, Nunomura A, ZhuX, Smith MA, Perry G (2007) Prevention and treatment ofAlzheimer disease and aging: Antioxidants. Mini Rev MedChem 7, 171-180.

[368] Kamat CD, Gadal S, Mhatre M, Williamson KS, Pye QN,Hensley K (2008) Antioxidants in central nervous systemdiseases: Preclinical promise and translational challenges.J Alzheimers Dis 15, 473-493.

[369] Toogood PL (2008) Mitochondrial drugs. Curr Opin ChemBiol 12, 457-463.

[370] Shen L, Ji HF (2010) Insights into the disappointing clin-ical trials of antioxidants in neurodegenerative diseases. JAlzheimers Dis 19, 1141-1142.

[371] Mecocci P, Polidori MC (2012) Antioxidant clinical tri-als in mild cognitive impairment and Alzheimer’s disease.Biochim Biophys Acta 1822, 631-638.

[372] Pocernich CB, Butterfield DA (2012) Elevation of glu-tathione as a therapeutic strategy in Alzheimer disease.Biochim Biophys Acta 1822, 625-630.

[373] Galasko DR, Peskind E, Clark CM, Quinn JF, Ring-man JM, Jicha GA, Cotman C, Cottrell B, MontineTJ, Thomas RG, Aisen P (2012) Antioxidants forAlzheimer disease: A randomized clinical trial with cere-brospinal fluid biomarker measures. Arch Neurol 69, 836-841.

[374] Kitagawa Y (2006) Edaravone in acute ischemic stroke.Intern Med 45, 225-226.

[375] Tsujita K, Shimomura H, Kaikita K, Kawano H, HokamakiJ, Nagayoshi Y, Yamashita T, Fukuda M, Nakamura Y,Sakamoto T, Yoshimura M, Ogawa H (2006) Long-termefficacy of edaravone in patients with acute myocardialinfarction. Circ J 70, 832-837.

[376] Lapchak PA (2010) A critical assessment of edaravoneacute ischemic stroke efficacy trials: Is edaravone an effec-tive neuroprotective therapy? Expert Opin Pharmacother11, 1753-1763.

[377] Kikuchi K, Kawahara K, Miyagi N, Uchikado H,Kuramoto T, Morimoto Y, Tancharoen S, Miura N, Take-nouchi K, Oyama Y, Shrestha B, Matsuda F, Yoshida Y,Arimura S, Mera K, Tada K, Yoshinaga N, MaenosonoR, Ohno Y, Hashiguchi T, Maruyama I, Shigemori M(2010) Edaravone: A new therapeutic approach for thetreatment of acute stroke. Med Hypotheses 75, 583-585.

[378] Gillis JC, Benefield P, McTavish D (1994) Idebenone.A review of its pharmacodynamic and pharmacokineticproperties, and therapeutic use in age-related cognitivedisorders. Drugs Aging 5, 133-152.

[379] Weyer G, Babej-Dolle RM, Hadler D, Hofmann S, Her-rmann WM (1997) A controlled study of 2 doses ofidebenone in the treatment of Alzheimer’s disease. Neu-ropsychobiology 36, 73-82.

[380] Voronkova KV, Meleshkov MN (2009) Use of Noben(idebenone) in the treatment of dementia and memoryimpairments without dementia. Neurosci Behav Physiol39, 501-506.

[381] Haley EC Jr (1998) High-dose tirilazad for acute stroke(RANTTAS II). RANTTAS II Investigators. Stroke 29,1256-1257.

[382] Dorsch NW, Kassell NF, Sinkula MS (2001) Metaanalysisof trials of tirilazad mesylate in aneurysmal SAH. ActaNeurochir Suppl 77, 233-235.

[383] Kavanagh RJ, Kam PC (2001) Lazaroids: Efficacy andmechanism of action of the 21-aminosteroids in neuropro-tection. Br J Anaesth 86, 110-119.

[384] Jang YG, Ilodigwe D, Macdonald RL (2009) Metaanalysisof tirilazad mesylate in patients with aneurysmal subarach-noid hemorrhage. Neurocrit Care 10, 141-147.

[385] Kelso GF, Porteous CM, Hughes G, Ledgerwood EC, GaneAM, Smith RA, Murphy MP (2002) Prevention of mito-chondrial oxidative damage using targeted antioxidants.Ann N Y Acad Sci 959, 263-274.

[386] Murphy MP, Smith RA (2007) Targeting antioxidants tomitochondria by conjugation to lipophilic cations. AnnuRev Pharmacol Toxicol 47, 629-656.

Page 68: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

68 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[387] Smith RA, Murphy MP (2010) Animal and human studieswith the mitochondria-targeted antioxidant MitoQ. Ann NY Acad Sci 1201, 96-103.

[388] Smith RA, Murphy MP (2011) Mitochondria-targetedantioxidants as therapies. Discov Med 11, 106-114.

[389] Reddy PH (2008) Mitochondrial medicine for aging andneurodegenerative diseases. Neuromolecular Med 10, 291-315.

[390] Reddy PH, Reddy TP (2011) Mitochondria as a therapeu-tic target for aging and neurodegenerative diseases. CurrAlzheimer Res 8, 393-409.

[391] Reddy PH, Tripathi R, Troung Q, Tirumala K, Reddy TP,Anekonda V, Shirendeb UP, Calkins MJ, Reddy AP, MaoP, Manczak M (2011) Abnormal mitochondrial dynamicsand synaptic degeneration as early events in Alzheimer’sdisease: Implications to mitochondria-targeted antioxidanttherapeutics. Biochim Biophys Acta 1822, 639-649.

[392] Bonda DJ, Wang X, Gustaw-Rothenberg KA, PerryG, Smith MA, Zhu X (2009) Mitochondrial drugs forAlzheimer disease. Pharmaceuticals (Basel) 2, 287-298.

[393] Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M,Zhu X, Smith MA (2010) Oxidative stress in Alzheimerdisease: A possibility for prevention. Neuropharmacology59, 290-294.

[394] Manczak M, Mao P, Calkins MJ, Cornea A, ReddyAP, Murphy MP, Szeto HH, Park B, Reddy PH(2010) Mitochondria-targeted antioxidants protect againstamyloid-beta toxicity in Alzheimer’s disease neurons. JAlzheimers Dis 20(Suppl 2), S609-S631.

[395] Smith RA, Porteous CM, Coulter CV, Murphy MP (1999)Selective targeting of an antioxidant to mitochondria. EurJ Biochem 263, 709-716.

[396] Reddy PH (2006) Mitochondrial oxidative damage inaging and Alzheimer’s disease: Implications for mito-chondrially targeted antioxidant therapeutics. J BiomedBiotechnol 2006, 31372.

[397] Kotake Y (1999) Pharmacologic properties of phenyl N-tert-butylnitrone. Antioxid Redox Signal 1, 481-499.

[398] Lin S, Rhodes PG, Lei M, Zhang F, Cai Z (2004) alpha-Phenyl-n-tert-butyl-nitrone attenuates hypoxic-ischemicwhite matter injury in the neonatal rat brain. Brain Res1007, 132-141.

[399] Robertson L, Hartley RC (2009) Synthesis of N-arylpyridinium salts bearing a nitrone spin trap aspotential mitochondria-targeted antioxidants. Tetrahedron65, 5284-5292.

[400] Zhao K, Zhao GM, Wu D, Soong Y, Birk AV, SchillerPW, Szeto HH (2004) Cell-permeable peptide antioxidantstargeted to inner mitochondrial membrane inhibit mito-chondrial swelling, oxidative cell death, and reperfusioninjury. J Biol Chem 279, 34682-34690.

[401] Reddy PH (2007) Mitochondrial dysfunction in agingand Alzheimer’s disease: Strategies to protect neurons.Antioxid Redox Signal 9, 1647-1658.

[402] Rocha M, Hernandez-Mijares A, Garcia-Malpartida K,Banuls C, Bellod L, Victor VM (2010) Mitochondria-targeted antioxidant peptides. Curr Pharm Des 16,3124-3131.

[403] Szeto HH, Schiller PW (2011) Novel therapies targetinginner mitochondrial membrane–from discovery to clinicaldevelopment. Pharm Res 28, 2669-2679.

[404] Chyan YJ, Poeggeler B, Omar RA, Chain DG, FrangioneB, Ghiso J, Pappolla MA (1999) Potent neuropro-tective properties against the Alzheimer beta-amyloidby an endogenous melatonin-related indole structure,

indole-3-propionic acid. J Biol Chem 274, 21937-21942.

[405] Liang LP, Huang J, Fulton R, Day BJ, Patel M (2007) Anorally active catalytic metalloporphyrin protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicityin vivo. J Neurosci 27, 4326-4333.

[406] Liang LP, Waldbaum S, Rowley S, Huang TT, Day BJ,Patel M (2012) Mitochondrial oxidative stress and epilepsyin SOD2 deficient mice: Attenuation by a lipophilic met-alloporphyrin. Neurobiol Dis 45, 1068-1076.

[407] Day BJ, Patel M (2008) Metalloporphyrin catalytic antiox-idants for the potential treatment of neurodegenerativediseases. Drugs Future 33, 1025-1032.

[408] Batinic-Haberle I, Reboucas JS, Spasojevic I (2010)Superoxide dismutase mimics: Chemistry, pharmacology,and therapeutic potential. Antioxid Redox Signal 13, 877-918.

[409] Batinic-Haberle I, Rajic Z, Tovmasyan A, Reboucas JS,Ye X, Leong KW, Dewhirst MW, Vujaskovic Z, Benov L,Spasojevic I (2011) Diverse functions of cationic Mn(III)N-substituted pyridylporphyrins, recognized as SOD mim-ics. Free Radic Biol Med 51, 1035-1053.

[410] Sheng H, Batinc-Haberle I, Warner DS (2002) Catalyticantioxidants as novel pharmacologic approaches to treat-ment of ischemic brain injury. Drug News Perspect 15,654-665.

[411] Sheng H, Yang W, Fukuda S, Tse HM, Paschen W, JohnsonK, Batinic-Haberle I, Crapo JD, Pearlstein RD, PiganelliJ, Warner DS (2009) Long-term neuroprotection from apotent redox-modulating metalloporphyrin in the rat. FreeRadic Biol Med 47, 917-923.

[412] Sheng H, Spasojevic I, Tse HM, Jung JY, Hong J,Zhang Z, Piganelli JD, Batinic-Haberle I, WarnerDS (2011) Neuroprotective efficacy from a lipophilicredox-modulating Mn(III) N-Hexylpyridylporphyrin,MnTnHex-2-PyP: Rodent models of ischemic stroke andsubarachnoid hemorrhage. J Pharmacol Exp Ther 338,906-916.

[413] Li L, Zhang B, Tao Y, Wang Y, Wei H, Zhao J, Huang R, PeiZ (2009) DL-3-n-butylphthalide protects endothelial cellsagainst oxidative/nitrosative stress, mitochondrial damageand subsequent cell death after oxygen glucose deprivationin vitro. Brain Res 1290, 91-101.

[414] Xiong N, Huang J, Chen C, Zhao Y, Zhang Z, Jia M, ZhangZ, Hou L, Yang H, Cao X, Liang Z, Zhang Y, Sun S, LinZ, Wang T (2012) Dl-3-n-butylphthalide, a natural antiox-idant, protects dopamine neurons in rotenone models forParkinson’s disease. Neurobiol Aging 33, 1777-1791.

[415] Valgimigli L, Valgimigli M, Gaiani S, Pedulli GF, BolondiL (2000) Measurement of oxidative stress in human liverby EPR spin-probe technique. Free Radic Res 33, 167-178.

[416] D’Aleo V, Del GS, Martano M, Bonamassa B, CanistroD, Soleti A, Valgimigli L, Paolini M, Filipponi F, BoggiU, Del PS, Lupi R (2009) The non-peptidyl low molecularweight radical scavenger IAC protects human pancreaticislets from lipotoxicity. Mol Cell Endocrinol 309, 63-66.

[417] Vasina V, Broccoli M, Ursino MG, Bellot SF, Soleti A,Paolini M, De PF (2009) Effects of the non-peptidyl lowmolecular weight radical scavenger IAC in DNBS-inducedcolitis in rats. Eur J Pharmacol 614, 137-145.

[418] Vasina V, Broccoli M, Ursino MG, Canistro D, ValgimigliL, Soleti A, Paolini M, De PF (2010) Non-peptidyl lowmolecular weight radical scavenger IAC attenuates DSS-induced colitis in rats. World J Gastroenterol 16, 3642-3650.

Page 69: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 69

[419] Puolivali J, Nurmi A, Miettinen TK, Soleti A, RiccardinoF, Kalesnykas G, Heikkinen T, Vartiainen N, PussinenR, Tahtivaara L, Lehtimaki K, Yrjanheikki J, CanistroD, Sapone A, Spisni E, Paolini M (2011) The radi-cal scavenger IAC (bis(1-hydroxy-2,2,6,6-tetramethyl-4-piperidinyl) decantionate) decreases mortality, enhancescognitive functions in water maze and reduces amyloidplaque burden in hAbetaPP transgenic mice. J AlzheimersDis 27, 499-510.

[420] Giles FJ, Fracasso PM, Kantarjian HM, Cortes JE, BrownRA, Verstovsek S, Alvarado Y, Thomas DA, Faderl S,Garcia-Manero G, Wright LP, Samson T, Cahill A, Lam-bert P, Plunkett W, Sznol M, DiPersio JF, Gandhi V(2003) Phase I and pharmacodynamic study of Triapine, anovel ribonucleotide reductase inhibitor, in patients withadvanced leukemia. Leuk Res 27, 1077-1083.

[421] Chen RW, Yao C, Lu XC, Jiang ZG, Whipple R, Liao Z,Ghanbari HA, Almassian B, Tortella FC, Dave JR (2005)PAN-811 (3-aminopyridine-2-carboxaldehyde thiosemi-carbazone), a novel neuroprotectant, elicits its function inprimary neuronal cultures by up-regulating Bcl-2 expres-sion. Neuroscience 135, 191-201.

[422] Gao X, Zheng CY, Qin GW, Tang XC, Zhang HY(2012) S-52, a novel nootropic compound, protects againstbeta-amyloid induced neuronal injury by attenuatingmitochondrial dysfunction. J Neurosci Res 90, 1981-1988.

[423] Zhong SZ, Ge QH, Li Q, Qu R, Ma SP (2009) Peoniflorinattentuates Abeta((1-42))-mediated neurotoxicity by reg-ulating calcium homeostasis and ameliorating oxidativestress in hippocampus of rats. J Neurol Sci 280, 71-78.

[424] Hatanaka M, Nishizawa C, Kakinoki T, Takahashi K,Nakamura S, Mashino T (2008) 2,2′-Pyridoin derivativesprotect HL-60 cells against oxidative stress. Bioorg MedChem Lett 18, 5290-5293.

[425] Xu H, Wang H, Zhuang L, Yan B, Yu Y, Wei Z, Zhang Y,Dyck LE, Richardson SJ, He J, Li X, Kong J, Li XM (2008)Demonstration of an anti-oxidative stress mechanism ofquetiapine: Implications for the treatment of Alzheimer’sdisease. FEBS J 275, 3718-3728.

[426] Han M, Liu Y, Tan Q, Zhang B, Wang W, Liu J, ZhangXJ, Wang YY, Zhang JM (2011) Therapeutic efficacyof stemazole in a beta-amyloid injection rat model ofAlzheimer’s disease. Eur J Pharmacol 657, 104-110.

[427] Choi SJ, Jeong CH, Choi SG, Chun JY, Kim YJ, Lee J,Shin DH, Heo HJ (2009) Zeatin prevents amyloid beta-induced neurotoxicity and scopolamine-induced cognitivedeficits. J Med Food 12, 271-277.

[428] Flaherty DP, Kiyota T, Dong Y, Ikezu T, Vennerstrom JL(2010) Phenolic bis-styrylbenzenes as beta-amyloid bind-ing ligands and free radical scavengers. J Med Chem 53,7992-7999.

[429] Lenhart JA, Ling X, Gandhi R, Guo TL, Gerk PM, BrunzellDH, Zhang S (2010) “Clicked” bivalent ligands contain-ing curcumin and cholesterol as multifunctional abetaoligomerization inhibitors: Design, synthesis, and biolog-ical characterization. J Med Chem 53, 6198-6209.

[430] Jung YS, Kang TS, Yoon JH, Joe BY, Lim HJ, Seong CM,Park WK, Kong JY, Cho J, Park NS (2002) Synthesis andevaluation of 4-hydroxyphenylacetic acid amides and 4-hydroxycinnamamides as antioxidants. Bioorg Med ChemLett 12, 2599-2602.

[431] Weston RM, Jarrott B, Ishizuka Y, Callaway JK (2006)AM-36 modulates the neutrophil inflammatory responseand reduces breakdown of the blood brain barrier after

endothelin-1 induced focal brain ischaemia. Br J Pharma-col 149, 712-723.

[432] Nicolazzo JA, Nguyen TT, Katneni K, Steuten JA,Smith G, Jarrott B, Callaway JK, Charman SA (2008)Pharmacokinetics and brain uptake of AM-36, a novel neu-roprotective agent, following intravenous administration torats. J Pharm Pharmacol 60, 171-178.

[433] Sorbera LA, Leeson PA, Castaner J (2001) CPI-1189.Drugs Future 26, 647-650.

[434] Clifford DB, McArthur JC, Schifitto G, Kieburtz K,McDermott MP, Letendre S, Cohen BA, Marder K, EllisRJ, Marra CM (2002) A randomized clinical trial of CPI-1189 for HIV-associated cognitive-motor impairment.Neurology 59, 1568-1573.

[435] Strid S, Borga O, Edenius C, Jostell KG, Odergren T, WeilA (2002) Pharmacokinetics in renally impaired subjectsof NXY-059, a nitrone-based, free-radical trapping agentdeveloped for the treatment of acute stroke. Eur J ClinPharmacol 58, 409-415.

[436] Green AR, Shuaib A (2006) Therapeutic strategies for thetreatment of stroke. Drug Discov Today 11, 681-693.

[437] Lees KR, Zivin JA, Ashwood T, Davalos A, Davis SM,Diener HC, Grotta J, Lyden P, Shuaib A, HardemarkHG, Wasiewski WW (2006) NXY-059 for acute ischemicstroke. N Engl J Med 354, 588-600.

[438] Lees KR, Davalos A, Davis SM, Diener HC, GrottaJ, Lyden P, Shuaib A, Ashwood T, Hardemark HG,Wasiewski W, Emeribe U, Zivin JA (2006) Additionaloutcomes and subgroup analyses of NXY-059 for acuteischemic stroke in the SAINT I trial. Stroke 37, 2970-2978.

[439] Ginsberg MD (2007) Life after cerovive: A personal per-spective on ischemic neuroprotection in the post-NXY-059era. Stroke 38, 1967-1972.

[440] Lyden PD, Shuaib A, Lees KR, Davalos A, Davis SM,Diener HC, Grotta JC, Ashwood TJ, Hardemark HG,Svensson HH, Rodichok L, Wasiewski WW, AhlbergG (2007) Safety and tolerability of NXY-059 for acuteintracerebral hemorrhage: The CHANT Trial. Stroke 38,2262-2269.

[441] Saver JL (2007) Clinical impact of NXY-059 demonstratedin the SAINT I trial: Derivation of number needed to treatfor benefit over entire range of functional disability. Stroke38, 1515-1518.

[442] Shuaib A, Lees KR, Lyden P, Grotta J, Davalos A, DavisSM, Diener HC, Ashwood T, Wasiewski WW, EmeribeU (2007) NXY-059 for the treatment of acute ischemicstroke. N Engl J Med 357, 562-571.

[443] Cheng YF, Jiang J, Hu P, Reinholdsson I, Guo W, Asen-blad N, Nilsson D (2008) Pharmacokinetics of 8-hourintravenous infusion of NXY-059: A phase I, randomized,double-blind (within dose panels), placebo-controlledstudy in healthy Chinese volunteers. Clin Ther 30, 2342-2353.

[444] Diener HC, Lees KR, Lyden P, Grotta J, Davalos A, DavisSM, Shuaib A, Ashwood T, Wasiewski W, Alderfer V,Hardemark HG, Rodichok L (2008) NXY-059 for the treat-ment of acute stroke: Pooled analysis of the SAINT I andII Trials. Stroke 39, 1751-1758.

[445] Savitz SI, Schabitz WR (2008) A Critique of SAINT II:Wishful thinking, dashed hopes, and the future of neuro-protection for acute stroke. Stroke 39, 1389-1391.

[446] Iwashita A, Maemoto T, Nakada H, Shima I, Mat-suoka N, Hisajima H (2003) A novel potent rad-ical scavenger, 8-(4-fluorophenyl)-2-((2E)-3-phenyl-2-propenoyl)-1,2,3,4-tetrahydropyrazol o[5,1-c] [1,2,4]tri-

Page 70: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

70 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

azine (FR210575), prevents neuronal cell death in culturedprimary neurons and attenuates brain injury after focalischemia in rats. J Pharmacol Exp Ther 307, 961-968.

[447] Dage JL, Ackermann BL, Barbuch RJ, Bernotas RC,Ohlweiler DF, Haegele KD, Thomas CE (1997) Evidencefor a novel pentyl radical adduct of the cyclic nitrone spintrap MDL 101,002. Free Radic Biol Med 22, 807-812.

[448] Johnson MP, McCarty DR, Velayo NL, Markgraf CG,Chmielewski PA, Ficorilli JV, Cheng HC, Thomas CE(1998) MDL 101,002, a free radical spin trap, is effica-cious in permanent and transient focal ischemia models.Life Sci 63, 241-253.

[449] Galeano M, Torre V, Deodato B, Campo GM, Colonna M,Sturiale A, Squadrito F, Cavallari V, Cucinotta D, BuemiM, Altavilla D (2001) Raxofelast, a hydrophilic vitamin E-like antioxidant, stimulates wound healing in geneticallydiabetic mice. Surgery 129, 467-477.

[450] Altavilla D, Galeano M, Bitto A, Minutoli L, SquadritoG, Seminara P, Venuti FS, Torre V, Calo M, ColonnaM, Lo CP, Giugliano G, Scuderi N, Mioni C, LeoneS, Squadrito F (2005) Lipid peroxidation inhibition byraxofelast improves angiogenesis and wound healing inexperimental burn wounds. Shock 24, 85-91.

[451] Altavilla D, Marini H, Seminara P, Squadrito G, Minu-toli L, Passaniti M, Bitto A, Calapai G, Calo M, CaputiAP, Squadrito F (2005) Protective effects of antioxidantraxofelast in alcohol-induced liver disease in mice. Phar-macology 74, 6-14.

[452] Bitto A, Minutoli L, Squadrito F, Polito F, Altavilla D(2007) Raxofelast, (+/–)5-(acetyloxy)-2,3-dihydro-4,6,7-trimethyl-2-benzofuranacetic acid: A new antioxidant tomodulate the inflammatory response during ischemia-reperfusion injury and impaired wound healing. Mini RevMed Chem 7, 339-343.

[453] Dorrell S (2001) Alzheimer’s disease - a metallic problem.Drug Discov Today 6, 61-62.

[454] Perry G, Cash AD, Srinivas R, Smith MA (2002) Metalsand oxidative homeostasis in Alzheimer’s disease. DrugDev Res 56, 293-299.

[455] Bush AI (2003) The metallobiology of Alzheimer’s dis-ease. Trends Neurosci 26, 207-214.

[456] Crouch PJ, Barnham KJ, Bush AI, White AR (2006) Ther-apeutic treatments for Alzheimer’s disease based on metalbioavailability. Drug News Perspect 19, 469-474.

[457] Schugar H, Green DE, Bowen ML, Scott LE, StorrT, Bohmerle K, Thomas F, Allen DD, Lockman PR,Merkel M, Thompson KH, Orvig C (2007) Combat-ing Alzheimer’s disease with multifunctional moleculesdesigned for metal passivation. Angew Chem Int Ed Engl46, 1716-1718.

[458] Shcherbatykh I, Carpenter DO (2007) The role of metalsin the etiology of Alzheimer’s disease. J Alzheimers Dis11, 191-205.

[459] Storr T, Merkel M, Song-Zhao GX, Scott LE, Green DE,Bowen ML, Thompson KH, Patrick BO, Schugar HJ,Orvig C (2007) Synthesis, characterization, and metalcoordinating ability of multifunctional carbohydrate-containing compounds for Alzheimer’s therapy. J AmChem Soc 129, 7453-7463.

[460] Barnham KJ, Bush AI (2008) Metals in Alzheimer’s andParkinson’s diseases. Curr Opin Chem Biol 12, 222-228.

[461] Bush AI (2008) Drug development based on the metalshypothesis of Alzheimer’s disease. J Alzheimers Dis 15,223-240.

[462] Bush AI, Tanzi RE (2008) Therapeutics for Alzheimer’sdisease based on the metal hypothesis. Neurotherapeutics5, 421-432.

[463] Bolognin S, Messori L, Zatta P (2009) Metal ionphysiopathology in neurodegenerative disorders. Neuro-molecular Med 11, 223-238.

[464] Bolognin S, Drago D, Messori L, Zatta P (2009) Chelationtherapy for neurodegenerative diseases. Med Res Rev 29,547-570.

[465] Rodriguez-Rodriguez C, Sanchez de GN, Rimola A,Avarez-Larena A, Lloveras V, Vidal-Gancedo J, Ventura S,Vendrell J, Sodupe M, Gonzalez-Duarte P (2009) Design,selection, and characterization of thioflavin-based inter-calation compounds with metal chelating properties forapplication in Alzheimer’s disease. J Am Chem Soc 131,1436-1451.

[466] Storr T, Scott LE, Bowen ML, Green DE, Thompson KH,Schugar HJ, Orvig C (2009) Glycosylated tetrahydrosalensas multifunctional molecules for Alzheimer’s therapy. Dal-ton Trans, 3034-3043.

[467] Zatta P, Drago D, Bolognin S, Sensi SL (2009) Alzheimer’sdisease, metal ions and metal homeostatic therapy. TrendsPharmacol Sci 30, 346-355.

[468] Bandyopadhyay S, Huang X, Lahiri DK, Rogers JT (2010)Novel drug targets based on metallobiology of Alzheimer’sdisease. Expert Opin Ther Targets 14, 1177-1197.

[469] Choi JS, Braymer JJ, Nanga RP, Ramamoorthy A, LimMH (2010) Design of small molecules that target metal-A{beta} species and regulate metal-induced A{beta}aggregation and neurotoxicity. Proc Natl Acad Sci U SA 107, 21990-21995.

[470] Braymer JJ, DeToma AS, Choi JS, Ko KS, Lim MH (2010)Recent development of bifunctional small molecules tostudy metal-amyloid-beta species in Alzheimer’s disease.Int J Alzheimers Dis 2011, 623051.

[471] Duce JA, Bush AI (2010) Biological metals andAlzheimer’s disease: Implications for therapeutics anddiagnostics. Prog Neurobiol 92, 1-18.

[472] Rodriguez-Rodriguez C, Rimola A, Rodriguez-SantiagoL, Ugliengo P, Avarez-Larena A, Gutierrez-de-Teran H,Sodupe M, Gonzalez-Duarte P (2010) Crystal structureof thioflavin-T and its binding to amyloid fibrils: Insightsat the molecular level. Chem Commun (Camb) 46, 1156-1158.

[473] Braymer JJ, Choi JS, DeToma AS, Wang C, Nam K,Kampf JW, Ramamoorthy A, Lim MH (2011) Develop-ment of bifunctional stilbene derivatives for targeting andmodulating metal-amyloid-beta species. Inorg Chem 50,10724-10734.

[474] Choi JS, Braymer JJ, Park SK, Mustafa S, Chae J, Lim MH(2011) Synthesis and characterization of IMPY derivativesthat regulate metal-induced amyloid-beta aggregation.Metallomics 3, 284-291.

[475] Rimola A, Ali-Torres J, Rodriguez-Rodriguez C, PoaterJ, Matito E, Sola M, Sodupe M (2011) Ab initio designof chelating ligands relevant to Alzheimer’s disease:Influence of metalloaromaticity. J Phys Chem A 115,12659-12666.

[476] Kenche VB, Barnham KJ (2011) Alzheimer’s disease &metals: Therapeutic opportunities. Br J Pharmacol 163,211-219.

[477] Tomljenovic L (2011) Aluminum and Alzheimer’s disease:After a century of controversy, is there a plausible link?J Alzheimers Dis 23, 567-598.

Page 71: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 71

[478] Manso Y, Comes G, Hidalgo J, Bush AI, Adlard PA (2011)Copper modulation as a therapy for Alzheimer’s disease?Int J Alzheimers Dis 2011, 370345.

[479] Crouch PJ, Barnham KJ (2012) Therapeutic redistributionof metal ions to treat Alzheimer’s disease. Acc Chem Res45, 1604-1611.

[480] Grasso G, Giuffrida ML, Rizzarelli E (2012) Metallostasisand amyloid beta-degrading enzymes. Metallomics 4, 937-949.

[481] Hung LW, Barnham KJ (2012) Modulating metals as atherapeutic strategy for Alzheimer’s disease. Future MedChem 4, 955-969.

[482] Squitti R (2012) Metals in Alzheimer’s disease: A systemicperspective. Front Biosci 17, 451-472.

[483] Watt AD, Villemagne VL, Barnham KJ (2012) Metals,membranes, and amyloid-beta oligomers: Key pieces inthe Alzheimer’s disease puzzle? J Alzheimers Dis, doi:10.3233/JAD-2012-129017 [Epub ahead of print].

[484] Craddock TJ, Tuszynski JA, Chopra D, Casey N, GoldsteinLE, Hameroff SR, Tanzi RE (2012) The zinc dyshome-ostasis hypoothesis of Alzheimer’s disease. PloS ONE, 7,3.

[485] Wise-Scira O, Xu L, Perry G, Coskuner O (2012) Struc-tures and free energy landscapes of aqueous zinc(II)-boundamyloid-beta(1-40) and zinc(II)-bound amyloid-beta(1-42) with dynamics. J Biol Inorg Chem 17, 927-938.

[486] Rosenberg G, Angel I, Kozak A (2005) Clinicalpharmacology of DP-b99 in healthy volunteers: Firstadministration to humans. Br J Clin Pharmacol 60, 7-16.

[487] Diener HC, Schneider D, Lampl Y, Bornstein NM, KozakA, Rosenberg G (2008) DP-b99, a membrane-activatedmetal ion chelator, as neuroprotective therapy in ischemicstroke. Stroke 39, 1774-1778.

[488] Rosenberg G, Bornstein N, Diener HC, Gorelick PB,Shuaib A, Lees K (2011) The Membrane-Activated Chela-tor Stroke Intervention (MACSI) Trial of DP-b99 in acuteischemic stroke: A randomized, double-blind, placebo-controlled, multinational pivotal phase III study. Int JStroke 6, 362-367.

[489] Angel I, Bar A, Horovitz T, Taler G, Krakovsky M, Resnit-sky D, Rosenberg G, Striem S, Friedman JE, Kozak A(2002) Metal ion chelation in neurodegenerative disorders.Drug Dev Res 56, 300-309.

[490] Yeung PK (2004) DP-b99 (D-Pharm). Curr Opin InvestigDrugs 5, 90-94.

[491] Barkalifa R, Hershfinkel M, Friedman JE, Kozak A, SeklerI (2009) The lipophilic zinc chelator DP-b99 prevents zincinduced neuronal death. Eur J Pharmacol 618, 15-21.

[492] Duce JA, Tsatsanis A, Cater MA, James SA, Robb E,Wikhe K, Leong SL, Perez K, Johanssen T, GreenoughMA, Cho HH, Galatis D, Moir RD, Masters CL, McLeanC, Tanzi RE, Cappai R, Barnham KJ, Ciccotosto GD,Rogers JT, Bush AI (2010) Iron-export ferroxidase activ-ity of beta-amyloid precursor protein is inhibited by zincin Alzheimer’s disease. Cell 142, 857-867.

[493] Lannfelt L, Blennow K, Zetterberg H, Batsman S, AmesD, Harrison J, Masters CL, Targum S, Bush AI, Mur-doch R, Wilson J, Ritchie CW (2008) Safety, efficacy,and biomarker findings of PBT2 in targeting Abeta as amodifying therapy for Alzheimer’s disease: A phase IIa,double-blind, randomised, placebo-controlled trial. LancetNeurol 7, 779-786.

[494] Faux NG, Ritchie CW, Gunn A, Rembach A, TsatsanisA, Bedo J, Harrison J, Lannfelt L, Blennow K, ZetterbergH, Ingelsson M, Masters CL, Tanzi RE, Cummings JL,

Herd CM, Bush AI (2010) PBT2 rapidly improves cogni-tion in Alzheimer’s Disease: Additional phase II analyses.J Alzheimers Dis 20, 509-516.

[495] Adlard PA, James SA, Bush AI, Masters CL (2009) beta-Amyloid as a molecular therapeutic target in Alzheimer’sdisease. Drugs Today (Barc) 45, 293-304.

[496] Adlard PA, Bica L, White AR, Nurjono M, Filiz G, CrouchPJ, Donnelly PS, Cappai R, Finkelstein DI, Bush AI (2011)Metal ionophore treatment restores dendritic spine den-sity and synaptic protein levels in a mouse model ofAlzheimer’s disease. PLoS One 6, e17669.

[497] Crouch PJ, Savva MS, Hung LW, Donnelly PS, Mot AI,Parker SJ, Greenough MA, Volitakis I, Adlard PA, ChernyRA, Masters CL, Bush AI, Barnham KJ, White AR (2011)The Alzheimer’s therapeutic PBT2 promotes amyloid-betadegradation and GSK3 phosphorylation via a metal chap-erone activity. J Neurochem 119, 220-230.

[498] Lee JY, Friedman JE, Angel I, Kozak A, Koh JY (2004) Thelipophilic metal chelator DP-109 reduces amyloid pathol-ogy in brains of human beta-amyloid precursor proteintransgenic mice. Neurobiol Aging 25, 1315-1321.

[499] Petri S, Calingasan NY, Alsaied OA, Wille E, Kiaei M,Friedman JE, Baranova O, Chavez JC, Beal MF (2007) Thelipophilic metal chelators DP-109 and DP-460 are neuro-protective in a transgenic mouse model of amyotrophiclateral sclerosis. J Neurochem 102, 991-1000.

[500] Fine JM, Baillargeon AM, Renner DB, Hoerster NS,Tokarev J, Colton S, Pelleg A, Andrews A, Sparley KA,Krogh KM, Frey WH, Hanson LR (2012) Intranasal defer-oxamine improves performance in radial arm water maze,stabilizes HIF-1alpha, and phosphorylates GSK3beta inP301L tau transgenic mice. Exp Brain Res 219, 381-390.

[501] 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.

[502] Zheng H, Youdim MB, Fridkin M (2009) Site-activatedmultifunctional chelator with acetylcholinesterase andneuroprotective-neurorestorative moieties for Alzheimer’stherapy. J Med Chem 52, 4095-4098.

[503] Jones MR, Service EL, Thompson JR, Wang MC, KimseyIJ, Detoma AS, Ramamoorthy A, Lim MH, Storr T (2012)Dual-function triazole-pyridine derivatives as inhibitors ofmetal-induced amyloid-beta aggregation. Metallomics 4,910-920.

[504] Youdim MB, Fridkin M, Zheng H (2005) Bifunctional drugderivatives of MAO-B inhibitor rasagiline and iron chela-tor VK-28 as a more effective approach to treatment ofbrain ageing and ageing neurodegenerative diseases. MechAgeing Dev 126, 317-326.

[505] 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.

[506] 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.J Neural Transm Suppl 70, 447-456.

[507] 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,

Page 72: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

72 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

for the treatment of Alzheimer’s disease. J Neurochem 100,490-502.

[508] 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.

[509] Weinreb O, Mandel S, Bar-Am O, Yogev-Falach M,Avramovich-Tirosh Y, Amit T, Youdim MB (2009) Mul-tifunctional neuroprotective derivatives of rasagiline asanti-Alzheimer’s disease drugs. Neurotherapeutics 6, 163-174.

[510] 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.

[511] 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.

[512] 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.

[513] 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.

[514] Kupershmidt L, Amit T, Bar-Am O, Weinreb O,Youdim MB (2012) Multi-target, neuroprotective and neu-rorestorative M30 improves cognitive impairment andreduces Alzheimer’s-like neuropathology and age-relatedalterations in mice. Mol Neurobiol 46, 217-220.

[515] Kupershmidt L, Weinreb O, Amit T, Mandel S, Bar-AmO, Youdim MB (2011) Novel molecular targets of the neu-roprotective/neurorescue multimodal iron chelating drugM30 in the mouse brain. Neuroscience 189, 345-358.

[516] Kupershmidt L, Amit T, Bar-Am O, Youdim MB, WeinrebO (2012) Neuroprotection by the multitarget iron chelatorM30 on age-related alterations in mice. Mech Ageing Dev133, 267-274.

[517] Kupershmidt L, Amit T, Bar-Am O, Youdim MB, Wein-reb O (2012) The novel multi-target iron chelating-radicalscavenging compound M30 possesses beneficial effects onmajor hallmarks of Alzheimer’s disease. Antioxid RedoxSignal 17, 860-877.

[518] Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY,Smith WL, Kalow W, Andrews DF (1991) Intramuscu-lar desferrioxamine in patients with Alzheimer’s disease.Lancet 337, 1304-1308.

[519] Cuajungco MP, Faget KY, Huang X, Tanzi RE, BushAI (2000) Metal chelation as a potential therapy forAlzheimer’s disease. Ann N Y Acad Sci 920, 292-304.

[520] de Lima MN, Presti-Torres J, Caldana F, Grazziotin MM,Scalco FS, Guimaraes MR, Bromberg E, Franke SI, Hen-riques JA, Schroder N (2007) Desferoxamine reversesneonatal iron-induced recognition memory impairment inrats. Eur J Pharmacol 570, 111-114.

[521] Cherny RA, Atwood CS, Xilinas ME, Gray DN, JonesWD, McLean CA, Barnham KJ, Volitakis I, FraserFW, Kim Y, Huang X, Goldstein LE, Moir RD, Lim

JT, Beyreuther K, Zheng H, Tanzi RE, Masters CL,Bush AI (2001) Treatment with a copper-zinc chelatormarkedly and rapidly inhibits beta-amyloid accumulationin Alzheimer’s disease transgenic mice. Neuron 30, 665-676.

[522] Ibach B, Haen E, Marienhagen J, Hajak G (2005) Clio-quinol treatment in familiar early onset of Alzheimer’sdisease: A case report. Pharmacopsychiatry 38, 178-179.

[523] Shachar DB, Kahana N, Kampel V, Warshawsky A,Youdim MB (2004) Neuroprotection by a novel brain per-meable iron chelator, VK-28, against 6-hydroxydopaminelession in rats. Neuropharmacology 46, 254-263.

[524] Youdim MB, Stephenson G, Ben SD (2004) Ironingiron out in Parkinson’s disease and other neurodegen-erative diseases with iron chelators: A lesson from6-hydroxydopamine and iron chelators, desferal and VK-28. Ann N Y Acad Sci 1012, 306-325.

[525] Bandyopadhyay S, Huang X, Cho H, Greig NH,Youdim MB, Rogers JT (2006) Metal specificity ofan iron-responsive element in Alzheimer’s APP mRNA5’untranslated region, tolerance of SH-SY5Y and H4neural cells to desferrioxamine, clioquinol, VK-28, anda piperazine chelator. J Neural Transm Suppl 71, 237-247.

[526] 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.

[527] Akhondzadeh S, Abbasi SH (2006) Herbal medicine inthe treatment of Alzheimer’s disease. Am J Alzheimers DisOther Demen 21, 113-118.

[528] Man SC, Durairajan SS, Kum WF, Lu JH, Huang JD,Cheng CF, Chung V, Xu M, Li M (2008) Systematicreview on the efficacy and safety of herbal medicines forAlzheimer’s disease. J Alzheimers Dis 14, 209-223.

[529] Kim J, Lee HJ, Lee KW (2010) Naturally occurring phy-tochemicals for the prevention of Alzheimer’s disease.J Neurochem 112, 1415-1430.

[530] Kennedy DO, Wightman EL (2011) Herbal extractsand phytochemicals: Plant secondary metabolites andthe enhancement of human brain function. Adv Nutr(Bethesda) 2, 32-50.

[531] Perry E, Howes MJ (2011) Medicinal plants and dementiatherapy: Herbal hopes for brain aging? CNS Neurosci Ther17, 683-698.

[532] Kim HG, Oh MS (2012) Herbal medicines for the preven-tion and treatment of Alzheimer’s disease. Curr PharmDes 18, 57-75.

[533] Mancuso C, Siciliano R, Barone E, Preziosi P (2012) Nat-ural substances and Alzheimer’s disease: From preclinicalstudies to evidence based medicine. Biochim Biophys Acta1822, 616-624.

[534] Yoshitake T, Yoshitake S, Kehr J (2010) The Ginkgo bilobaextract EGb 761(R) and its main constituent flavonoids andginkgolides increase extracellular dopamine levels in therat prefrontal cortex. Br J Pharmacol 159, 659-668.

[535] Han CK, Park YH, Jin DQ, Hwang YK, Oh KB, Han JS(2007) SK-PC-B70M from Pulsatilla koreana improvesscopolamine-induced impairments of memory consoli-dation and spatial working memory. Brain Res 1184,254-259.

[536] Han CK, Choi WR, Oh KB (2007) Cognition-enhancingand neuroprotective effects of hederacolchiside-E fromPulsatilla koreana. Planta Med 73, 665-669.

Page 73: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 73

[537] Seo JS, Kim TK, Leem YH, Lee KW, Park SK, Baek IS,Kim KS, Im GJ, Lee SM, Park YH, Han PL (2009) SK-PC-B70M confers anti-oxidant activity and reduces Abetalevels in the brain of Tg2576 mice. Brain Res 1261, 100-108.

[538] Seo JS, Baek IS, Leem YH, Kim TK, Cho Y, Lee SM, ParkYH, Han PL (2011) SK-PC-B70M alleviates neurologicsymptoms in G93A-SOD1 amyotrophic lateral sclerosismice. Brain Res 1368, 299-307.

[539] Baur JA, Sinclair DA (2006) Therapeutic potential ofresveratrol: The in vivo evidence. Nat Rev Drug Discov5, 493-506.

[540] 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, LakattaEG, Le CD, Shaw RJ, Navas P, Puigserver P, Ingram DK,de CR, Sinclair DA (2006) Resveratrol improves healthand survival of mice on a high-calorie diet. Nature 444,337-342.

[541] Baur JA (2010) Resveratrol, sirtuins, and the promise of aDR mimetic. Mech Ageing Dev 131, 261-269.

[542] Baur JA, Chen D, Chini EN, Chua K, Cohen HY, deCR, 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.

[543] Kennedy DO, Wightman EL, Reay JL, Lietz G, OkelloEJ, Wilde A, Haskell CF (2010) Effects of resveratrol oncerebral blood flow variables and cognitive performancein humans: A double-blind, placebo-controlled, crossoverinvestigation. Am J Clin Nutr 91, 1590-1597.

[544] Quideau S, Deffieux D, Pouysegu L (2012) Resveratrolstill has something to say about aging! Angew Chem IntEd Engl 51, 6824-6826.

[545] Marambaud P, Zhao H, Davies P (2005) Resveratrolpromotes clearance of Alzheimer’s disease amyloid-betapeptides. J Biol Chem 280, 37377-37382.

[546] 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.

[547] 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.

[548] Albani D, Polito L, Signorini A, Forloni G (2010)Neuroprotective properties of resveratrol in different neu-rodegenerative disorders. Biofactors 36, 370-376.

[549] 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.

[550] Beher D, Wu J, Cumine S, Kim KW, Lu SC, Atangan L,Wang M (2009) Resveratrol is not a direct activator ofSIRT1 enzyme activity. Chem Biol Drug Des 74, 619-624.

[551] 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.

[552] 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.

[553] Zhang Y, Li SZ, Li J, Pan X, Cahoon RE, Jaworski JG,Wang X, Jez JM, Chen F, Yu O (2006) Using unnaturalprotein fusions to engineer resveratrol biosynthesis in yeastand Mammalian cells. J Am Chem Soc 128, 13030-13031.

[554] 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.

[555] Wang Y, Xia Z, Xu JR, Wang YX, Hou LN, QiuY, Chen HZ (2012) Alpha-mangostin, a polyphenolicxanthone derivative from mangosteen, attenuates beta-amyloid oligomers-induced neurotoxicity by inhibitingamyloid aggregation. Neuropharmacology 62, 871-881.

[556] Paris D, Beaulieu-Abdelahad D, Bachmeier C, Reed J, it-Ghezala G, Bishop A, Chao J, Mathura V, Crawford F,Mullan M (2011) Anatabine lowers Alzheimer’s Abetaproduction in vitro and in vivo. Eur J Pharmacol 670,384-391.

[557] Radhika P, Annapurna A, Rao SN (2012) Immunos-timulant, cerebroprotective & nootropic activities ofAndrographis paniculata leaves extract in normal & type2 diabetic rats. Indian J Med Res 135, 636-641.

[558] Lashuel HA, Hartley DM, Balakhaneh D, Aggarwal A,Teichberg S, Callaway DJ (2002) New class of inhibitors ofamyloid-beta fibril formation. Implications for the mecha-nism of pathogenesis in Alzheimer’s disease. J Biol Chem277, 42881-42890.

[559] Himeno E, Ohyagi Y, Ma L, Nakamura N, MiyoshiK, Sakae N, Motomura K, Soejima N, Yamasaki R,Hashimoto T, Tabira T, LaFerla FM, Kira J (2011)Apomorphine treatment in Alzheimer mice promotingamyloid-beta degradation. Ann Neurol 69, 248-256.

[560] Steele JW, Gandy S (2011) Apomorphine and AlzheimerAbeta: Roles for regulated alpha cleavage, autophagy, andantioxidation? Ann Neurol 69, 221-225.

[561] Howland RH (2010) Drug therapies for cognitive impair-ment and dementia. J Psychosoc Nurs Ment Health Serv48, 11-14.

[562] Roman MW (2010) Axona (Accera, Inc): A new medicalfood therapy for persons with Alzheimer’s disease. IssuesMent Health Nurs 31, 435-436.

[563] Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD,Watson GS, Hyde K, Chapman D, Craft S (2004) Effectsof beta-hydroxybutyrate on cognition in memory-impairedadults. Neurobiol Aging 25, 311-314.

[564] Costantini LC, Barr LJ, Vogel JL, Henderson ST (2008)Hypometabolism as a therapeutic target in Alzheimer’sdisease. BMC Neurosci 9(Suppl 2), S16.

[565] Pocernich CB, Lange ML, Sultana R, Butterfield DA(2011) Nutritional approaches to modulate oxidative stressin Alzheimer’s disease. Curr Alzheimer Res 8, 452-469.

[566] Pase MP, Kean J, Sarris J, Neale C, Scholey AB, StoughC (2012) The cognitive-enhancing effects of Bacopamonnieri: A systematic review of randomized, controlled

Page 74: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

74 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

human clinical trials. J Altern Complement Med 18, 647-652.

[567] Saini N, Singh D, Sandhir R (2012) Neuroprotectiveeffects of Bacopa monnieri in experimental model ofdementia. Neurochem Res 37, 1928-1937.

[568] Lebeau A, Esclaire F, Rostene W, Pelaprat D (2001)Baicalein protects cortical neurons from beta-amyloid (25-35) induced toxicity. Neuroreport 12, 2199-2202.

[569] Wang SY, Wang HH, Chi CW, Chen CF, Liao JF (2004)Effects of baicalein on beta-amyloid peptide-(25-35)-induced amnesia in mice. Eur J Pharmacol 506, 55-61.

[570] Yin F, Liu J, Ji X, Wang Y, Zidichouski J, Zhang J (2011)Baicalin prevents the production of hydrogen peroxideand oxidative stress induced by Abeta aggregation in SH-SY5Y cells. Neurosci Lett 492, 76-79.

[571] Gauci AJ, Caruana M, Giese A, Scerri C, Vassallo N (2011)Identification of polyphenolic compounds and black teaextract as potent inhibitors of lipid membrane desta-bilization by abeta42 aggregates. J Alzheimers Dis 27,767-779.

[572] Lu JH, Ardah MT, Durairajan SS, Liu LF, Xie LX, FongWF, Hasan MY, Huang JD, El-Agnaf OM, Li M (2011)Baicalein inhibits formation of alpha-synuclein oligomerswithin living cells and prevents Abeta peptide fibrillationand oligomerisation. Chembiochem 12, 615-624.

[573] Geng Y, Li C, Liu J, Xing G, Zhou L, Dong M, Li X,Niu Y (2010) Beta-asarone improves cognitive functionby suppressing neuronal apoptosis in the beta-amyloidhippocampus injection rats. Biol Pharm Bull 33, 836-843.

[574] Li C, Xing G, Dong M, Zhou L, Li J, Wang G, Zou D, WangR, Liu J, Niu Y (2010) Beta-asarone protection againstbeta-amyloid-induced neurotoxicity in PC12 cells via JNKsignaling and modulation of Bcl-2 family proteins. Eur JPharmacol 635, 96-102.

[575] Liu J, Li C, Xing G, Zhou L, Dong M, Geng Y, Li X,Li J, Wang G, Zou D, Niu Y (2010) Beta-asarone atten-uates neuronal apoptosis induced by Beta amyloid in rathippocampus. Yakugaku Zasshi 130, 737-746.

[576] Lee JY, Kim KY, Shin KY, Won BY, Jung HY, Suh YH(2009) Effects of BT-11 on memory in healthy humans.Neurosci Lett 454, 111-114.

[577] Shin KY, Won BY, Heo C, Kim HJ, Jang DP, Park CH,Kim S, Kim HS, Kim YB, Lee HG, Lee SH, Cho ZH,Suh YH (2009) BT-11 improves stress-induced memoryimpairments through increment of glucose utilization andtotal neural cell adhesion molecule levels in rat brains.J Neurosci Res 87, 260-268.

[578] Shin KY, Lee JY, Won BY, Jung HY, Chang KA, KoppulaS, Suh YH (2009) BT-11 is effective for enhancing cog-nitive functions in the elderly humans. Neurosci Lett 465,157-159.

[579] Wang J, Ho L, Zhao Z, Seror I, Humala N, DicksteinDL, Thiyagarajan M, Percival SS, Talcott ST, PasinettiGM (2006) Moderate consumption of Cabernet Sauvi-gnon attenuates Abeta neuropathology in a mouse modelof Alzheimer’s disease. FASEB J 20, 2313-2320.

[580] Azizi Z, Ebrahimi S, Saadatfar E, Kamalinejad M, Maj-lessi N (2012) Cognitive-enhancing activity of thymol andcarvacrol in two rat models of dementia. Behav Pharmacol23, 241-249.

[581] Ono K, Yoshiike Y, Takashima A, Hasegawa K, Naiki H,Yamada M (2003) Potent anti-amyloidogenic and fibril-destabilizing effects of polyphenols in vitro: Implicationsfor the prevention and therapeutics of Alzheimer’s disease.J Neurochem 87, 172-181.

[582] Mandel SA, Amit T, Kalfon L, Reznichenko L, YoudimMB (2008) Targeting multiple neurodegenerative diseasesetiologies with multimodal-acting green tea catechins.J Nutr 138, 1578S-1583S.

[583] Ferruzzi MG, Lobo JK, Janle EM, Cooper B, Simon JE,Wu QL, Welch C, Ho L, Weaver C, Pasinetti GM (2009)Bioavailability of gallic acid and catechins from grapeseed polyphenol extract is improved by repeated dosingin rats: Implications for treatment in Alzheimer’s disease.J Alzheimers Dis 18, 113-124.

[584] Paris D, Ganey NJ, Laporte V, Patel NS, Beaulieu-Abdelahad D, Bachmeier C, March A, it-Ghezala G,Mullan MJ (2010) Reduction of beta-amyloid pathologyby celastrol in a transgenic mouse model of Alzheimer’sdisease. J Neuroinflammation 7, 17.

[585] Bhanumathy M, Harish MS, Shivaprasad HN, Sushma G(2010) Nootropic activity of Celastrus paniculatus seed.Pharm Biol 48, 324-327.

[586] Frydman-Marom A, Levin A, Farfara D, Benromano T,Scherzer-Attali R, Peled S, Vassar R, Segal D, Gazit E,Frenkel D, Ovadia M (2011) Orally administrated cin-namon extract reduces beta-amyloid oligomerization andcorrects cognitive impairment in Alzheimer’s disease ani-mal models. PLoS One 6, e16564.

[587] Marumoto S, Miyazawa M (2012) Structure-activityrelationships for naturally occurring coumarins as beta-secretase inhibitor. Bioorg Med Chem 20, 784-788.

[588] Anand P, Singh B, Singh N (2012) A review on coumarinsas acetylcholinesterase inhibitors for Alzheimer’s disease.Bioorg Med Chem 20, 1175-1180.

[589] Mei Z, Yan P, Situ B, Mou Y, Liu P (2012) Cryptotan-shinione inhibits beta-amyloid aggregation and protectsdamage from beta-amyloid in SH-SY5Y cells. NeurochemRes 37, 622-628.

[590] Mei Z, Situ B, Tan X, Zheng S, Zhang F, Yan P, Liu P(2010) Cryptotanshinione upregulates alpha-secretase byactivation PI3K pathway in cortical neurons. Brain Res1348, 165-173.

[591] Jiang T, Zhi XL, Zhang YH, Pan LF, Zhou P (2012)Inhibitory effect of curcumin on the Al(III)-inducedAbeta(42) aggregation and neurotoxicity in vitro. BiochimBiophys Acta 1822, 1207-1215.

[592] Lim GP, Chu T, Yang F, Beech W, Frautschy SA, ColeGM (2001) The curry spice curcumin reduces oxidativedamage and amyloid pathology in an Alzheimer transgenicmouse. J Neurosci 21, 8370-8377.

[593] Ono K, Hasegawa K, Naiki H, Yamada M (2004)Curcumin has potent anti-amyloidogenic effects forAlzheimer’s beta-amyloid fibrils in vitro. J Neurosci Res75, 742-750.

[594] Ringman JM, Frautschy SA, Cole GM, Masterman DL,Cummings JL (2005) A potential role of the curry spicecurcumin in Alzheimer’s disease. Curr Alzheimer Res 2,131-136.

[595] Yang F, Lim GP, Begum AN, Ubeda OJ, SimmonsMR, Ambegaokar SS, Chen PP, Kayed R, Glabe CG,Frautschy SA, Cole GM (2005) Curcumin inhibits forma-tion of amyloid beta oligomers and fibrils, binds plaques,and reduces amyloid in vivo. J Biol Chem 280, 5892-5901.

[596] Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT,Bacskai BJ (2007) Curcumin labels amyloid pathology invivo, disrupts existing plaques, and partially restores dis-torted neurites in an Alzheimer mouse model. J Neurochem102, 1095-1104.

Page 75: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 75

[597] Begum AN, Jones MR, Lim GP, Morihara T, Kim P, HeathDD, Rock CL, Pruitt MA, Yang F, Hudspeth B, Hu S,Faull KF, Teter B, Cole GM, Frautschy SA (2008) Cur-cumin structure-function, bioavailability, and efficacy inmodels of neuroinflammation and Alzheimer’s disease.J Pharmacol Exp Ther 326, 196-208.

[598] Park SY, Kim HS, Cho EK, Kwon BY, Phark S, HwangKW, Sul D (2008) Curcumin protected PC12 cells againstbeta-amyloid-induced toxicity through the inhibition ofoxidative damage and tau hyperphosphorylation. FoodChem Toxicol 46, 2881-2887.

[599] Hamaguchi T, Ono K, Murase A, Yamada M (2009) Phe-nolic compounds prevent Alzheimer’s pathology throughdifferent effects on the amyloid-beta aggregation pathway.Am J Pathol 175, 2557-2565.

[600] Hamaguchi T, Ono K, Yamada M (2010) REVIEW: Cur-cumin and Alzheimer’s disease. CNS Neurosci Ther 16,285-297.

[601] Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, GantDJ, Chen PP, Hudspeth B, Chen C, Zhao Y, Vinters HV,Frautschy SA, Cole GM (2009) Beta-amyloid oligomersinduce phosphorylation of tau and inactivation of insulinreceptor substrate via c-Jun N-terminal kinase signaling:Suppression by omega-3 fatty acids and curcumin. J Neu-rosci 29, 9078-9089.

[602] Ray B, Lahiri DK (2009) Neuroinflammation inAlzheimer’s disease: Different molecular targets andpotential therapeutic agents including curcumin. CurrOpin Pharmacol 9, 434-444.

[603] Zhang C, Browne A, Child D, Tanzi RE (2010) Curcumindecreases amyloid-beta peptide levels by attenuating thematuration of amyloid-beta precursor protein. J Biol Chem285, 28472-28480.

[604] Ferrari E, Pignedoli F, Imbriano C, Marverti G, Basile V,Venturi E, Saladini M (2011) Newly synthesized curcuminderivatives: Crosstalk between chemico-physical proper-ties and biological activity. J Med Chem 54, 8066-8077.

[605] Gupta SC, Prasad S, Kim JH, Patchva S, Webb LJ,Priyadarsini IK, Aggarwal BB (2011) Multitargeting bycurcumin as revealed by molecular interaction studies. NatProd Rep 28, 1937-1955.

[606] Mancuso C, Siciliano R, Barone E (2011) Curcumin andAlzheimer disease: This marriage is not to be performed.J Biol Chem 286, le3.

[607] Ray B, Bisht S, Maitra A, Maitra A, Lahiri DK (2011) Neu-roprotective and neurorescue effects of a novel polymericnanoparticle formulation of curcumin (NanoCurc) in theneuronal cell culture and animal model: Implications forAlzheimer’s disease. J Alzheimers Dis 23, 61-77.

[608] Mourtas S, Canovi M, Zona C, Aurilia D, Niarakis A,La FB, Salmona M, Nicotra F, Gobbi M, AntimisiarisSG (2011) Curcumin-decorated nanoliposomes with veryhigh affinity for amyloid-beta1-42 peptide. Biomaterials32, 1635-1645.

[609] Reinke AA, Gestwicki JE (2007) Structure-activity rela-tionships of amyloid beta-aggregation inhibitors based oncurcumin: Influence of linker length and flexibility. ChemBiol Drug Des 70, 206-215.

[610] Narlawar R, Pickhardt M, Leuchtenberger S, Baumann K,Krause S, Dyrks T, Weggen S, Mandelkow E, SchmidtB (2008) Curcumin-derived pyrazoles and isoxazoles:Swiss army knives or blunt tools for Alzheimer’s disease?ChemMedChem 3, 165-172.

[611] Orlando RA, Gonzales AM, Royer RE, Deck LM, Van-der Jagt DL (2012) A chemical analog of curcumin as

an improved inhibitor of amyloid Abeta oligomerization.PLoS One 7, e31869.

[612] Zhang Y, Saito H, Nishiyama N, Abe K (1994) Effectsof DX-9386, a traditional Chinese medicinal prescription,on long-term potentiation in the dentate gyrus in rats. BiolPharm Bull 17, 1337-1340

[613] Yang SG, Zhang X, Sun XS, Ling TJ, Feng Y, Du XY,Zhao M, Yang Y, Xue D, Wang L, Liu RT (2010) Diverseecdysterones show different effects on amyloid-beta42aggregation but all uniformly inhibit amyloid-beta42-induced cytotoxicity. J Alzheimers Dis 22, 107-117.

[614] Rezai-Zadeh K, Shytle D, Sun N, Mori T, Hou H, Jeanni-ton D, Ehrhart J, Townsend K, Zeng J, Morgan D, Hardy J,Town T, Tan J (2005) Green tea epigallocatechin-3-gallate(EGCG) modulates amyloid precursor protein cleavageand reduces cerebral amyloidosis in Alzheimer transgenicmice. J Neurosci 25, 8807-8814.

[615] Rezai-Zadeh K, Arendash GW, Hou H, Fernandez F,Jensen M, Runfeldt M, Shytle RD, Tan J (2008) Green teaepigallocatechin-3-gallate (EGCG) reduces beta-amyloidmediated cognitive impairment and modulates tau pathol-ogy in Alzheimer transgenic mice. Brain Res 1214,177-187.

[616] Rezai-Zadeh K, Douglas SR, Bai Y, Tian J, Hou H,Mori T, Zeng J, Obregon D, Town T, Tan J (2009)Flavonoid-mediated presenilin-1 phosphorylation reducesAlzheimer’s disease beta-amyloid production. J Cell MolMed 13, 574-588.

[617] Lee JW, Lee YK, Ban JO, Ha TY, Yun YP, Han SB,Oh KW, Hong JT (2009) Green tea (–)-epigallocatechin-3-gallate inhibits beta-amyloid-induced cognitive dys-function through modification of secretase activity viainhibition of ERK and NF-kappaB pathways in mice.J Nutr 139, 1987-1993.

[618] Bieschke J, Russ J, Friedrich RP, Ehrnhoefer DE, Wobst H,Neugebauer K, Wanker EE (2010) EGCG remodels maturealpha-synuclein and amyloid-beta fibrils and reduces cel-lular toxicity. Proc Natl Acad Sci U S A 107, 7710-7715.

[619] Mandel SA, Avramovich-Tirosh Y, Reznichenko L, ZhengH, Weinreb O, Amit T, Youdim MB (2005) Multifunc-tional activities of green tea catechins in neuroprotection.Modulation of cell survival genes, iron-dependent oxida-tive stress and PKC signaling pathway. Neurosignals 14,46-60.

[620] Mandel SA, Amit T, Weinreb O, Reznichenko L, YoudimMB (2008) Simultaneous manipulation of multiple braintargets by green tea catechins: A potential neuroprotec-tive strategy for Alzheimer and Parkinson diseases. CNSNeurosci Ther 14, 352-365.

[621] Mandel SA, Amit T, Kalfon L, Reznichenko L, Wein-reb O, Youdim MB (2008) Cell signaling pathways andiron chelation in the neurorestorative activity of green teapolyphenols: Special reference to epigallocatechin gallate(EGCG). J Alzheimers Dis 15, 211-222.

[622] Mandel SA, Amit T, Weinreb O, Youdim MB (2011)Understanding the broad-spectrum neuroprotective actionprofile of green tea polyphenols in aging and neurodegen-erative diseases. J Alzheimers Dis 25, 187-208.

[623] Mandel SA, Weinreb O, Amit T, Youdim MB (2012)The importance of the multiple target action of green teapolyphenols for neuroprotection. Front Biosci (Schol Ed)4, 581-598.

[624] Kalfon L, Youdim MB, Mandel SA (2007) Greentea polyphenol (–) -epigallocatechin-3-gallate promotesthe rapid protein kinase C- and proteasome-mediated

Page 76: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

76 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

degradation of Bad: Implications for neuroprotection.J Neurochem 100, 992-1002.

[625] Lopez Del Amo JM, Fink U, Dasari M, Grelle G, WankerEE, Bieschke J, Reif B (2012) Structural propertiesof EGCG-induced, nontoxic Alzheimer’s disease Abetaoligomers. J Mol Biol 421, 517-524.

[626] Fernandez JW, Rezai-Zadeh K, Obregon D, Tan J(2010) EGCG functions through estrogen receptor-mediated activation of ADAM10 in the promotion ofnon-amyloidogenic processing of APP. FEBS Lett 584,4259-4267.

[627] Obregon DF, Rezai-Zadeh K, Bai Y, Sun N, Hou H, EhrhartJ, Zeng J, Mori T, Arendash GW, Shytle D, Town T,Tan J (2006) ADAM10 activation is required for greentea (–)-epigallocatechin-3-gallate-induced alpha-secretasecleavage of amyloid precursor protein. J Biol Chem 281,16419-16427.

[628] Smith A, Giunta B, Bickford PC, Fountain M, TanJ, Shytle RD (2010) Nanolipidic particles improve thebioavailability and alpha-secretase inducing ability ofepigallocatechin-3-gallate (EGCG) for the treatment ofAlzheimer’s disease. Int J Pharm 389, 207-212.

[629] Kang SY, Lee KY, Koo KA, Yoon JS, Lim SW, Kim YC,Sung SH (2005) ESP-102, a standardized combined extractof Angelica gigas, Saururus chinensis and Schizandrachinensis, significantly improved scopolamine-inducedmemory impairment in mice. Life Sci 76, 1691-1705.

[630] Kim DH, Jung WY, Park SJ, Kim JM, Lee S, KimYC, Ryu JH (2010) Anti-amnesic effect of ESP-102 onAbeta(1-42)-induced memory impairment in mice. Phar-macol Biochem Behav 97, 239-248.

[631] Kwon SH, Lee HK, Kim JA, Hong SI, Kim SY, Jo TH,Park YI, Lee CK, Kim YB, Lee SY, Jang CG (2011)Neuroprotective effects of Eucommia ulmoides Oliv. Barkon amyloid beta(25-35)-induced learning and memoryimpairments in mice. Neurosci Lett 487, 123-127.

[632] Williams RJ, Spencer JP (2012) Flavonoids, cognition, anddementia: Actions, mechanisms, and potential therapeuticutility for Alzheimer disease. Free Radic Biol Med 52,35-45.

[633] Spencer JP, Vafeiadou K, Williams RJ, Vauzour D (2012)Neuroinflammation: Modulation by flavonoids and mech-anisms of action. Mol Aspects Med 33, 83-97.

[634] Paris D, Mathura V, it-Ghezala G, Beaulieu-AbdelahadD, Patel N, Bachmeier C, Mullan M (2011) Flavonoidslower Alzheimer’s Abeta production via an NFkappaBdependent mechanism. Bioinformation 6, 229-236.

[635] Zhu JT, Choi RC, Chu GK, Cheung AW, Gao QT, LiJ, Jiang ZY, Dong TT, Tsim KW (2007) Flavonoidspossess neuroprotective effects on cultured pheochromo-cytoma PC12 cells: A comparison of different flavonoidsin activating estrogenic effect and in preventing beta-amyloid-induced cell death. J Agric Food Chem 55,2438-2445.

[636] Uriarte-Pueyo I, Calvo MI (2011) Flavonoids as acetyl-cholinesterase inhibitors. Curr Med Chem 18, 5289-5302.

[637] Thapa A, Woo ER, Chi EY, Sharoar MG, Jin HG,Shin SY, Park IS (2011) Biflavonoids are superior tomonoflavonoids in inhibiting amyloid-beta toxicity andfibrillogenesis via accumulation of nontoxic oligomer-likestructures. Biochemistry 50, 2445-2455.

[638] Broersen LM, Kuipers AA, Balvers M, van WN, SavelkoulPJ, de Wilde MC, van der Beek EM, Sijben JW, HagemanRJ, Kamphuis PJ, Kiliaan AJ (2012) A specific multi-nutrient diet reduces Alzheimer-like pathology in young

adult AbetaPPswe/PS1dE9 mice. J Alzheimers Dis, doi:10.3233/JAD-2012-112039 [Epub ahead of print].

[639] Cornejo A, Jimenez JM, Caballero L, Melo F, MaccioniRB (2011) Fulvic acid inhibits aggregation and promotesdisassembly of tau fibrils associated with Alzheimer’s dis-ease. J Alzheimers Dis 27, 143-153.

[640] Selassie M, Griffin B, Gwebu N, Gwebu ET (1999) Agedgarlic extract attenuates the cytotoxicity of beta-amyloidon undifferentiated PC12 cells. In Vitro Cell Dev Biol Anim35, 369-370.

[641] Griffin B, Selassie M, Gwebu ET (2000) Aged garlicextract suppresses lipid peroxidation induced by beta-amyloid in PC12 cells. In Vitro Cell Dev Biol Anim 36,279-280.

[642] Peng Q, Buz’Zard AR, Lau BH (2002) Neuropro-tective effect of garlic compounds in amyloid-betapeptide-induced apoptosis in vitro. Med Sci Monit 8,BR328-BR337.

[643] Gupta VB, Indi SS, Rao KS (2009) Garlic extract exhibitsantiamyloidogenic activity on amyloid-beta fibrillogene-sis: Relevance to Alzheimer’s disease. Phytother Res 23,111-115.

[644] Chauhan NB (2006) Effect of aged garlic extract on APPprocessing and tau phosphorylation in Alzheimer’s trans-genic model Tg2576. J Ethnopharmacol 108, 385-394.

[645] Chauhan NB, Sandoval J (2007) Amelioration of earlycognitive deficits by aged garlic extract in Alzheimer’stransgenic mice. Phytother Res 21, 629-640.

[646] Ray B, Chauhan NB, Lahiri DK (2011) The “aged garlicextract:” (AGE) and one of its active ingredients S-allyl-L-cysteine (SAC) as potential preventive and therapeuticagents for Alzheimer’s disease (AD). Curr Med Chem 18,3306-3313.

[647] Stackman RW, Eckenstein F, Frei B, Kulhanek D, NowlinJ, Quinn JF (2003) Prevention of age-related spatial mem-ory deficits in a transgenic mouse model of Alzheimer’sdisease by chronic Ginkgo biloba treatment. Exp Neurol184, 510-520.

[648] Tchantchou F, Xu Y, Wu Y, Christen Y, Luo Y (2007)EGb 761 enhances adult hippocampal neurogenesis andphosphorylation of CREB in transgenic mouse model ofAlzheimer’s disease. FASEB J 21, 2400-2408.

[649] Defeudis FV, Drieu K (2000) Ginkgo biloba extract (EGb761) and CNS functions: Basic studies and clinical appli-cations. Curr Drug Targets 1, 25-58.

[650] Defeudis FV (2003) A brief history of EGb 761 and itstherapeutic uses. Pharmacopsychiatry 36(Suppl 1), S2-S7.

[651] Allain H, Raoul P, Lieury A, LeCoz F, Gandon JM,D’Arbigny P (1993) Effect of two doses of ginkgo bilobaextract (EGb 761) on the dual-coding test in elderly sub-jects. Clin Ther 15, 549-558.

[652] Le Bars PL, Katz MM, Berman N, Itil TM, FreedmanAM, Schatzberg AF (1997) A placebo-controlled, double-blind, randomized trial of an extract of Ginkgo biloba fordementia. North American EGb Study Group. JAMA 278,1327-1332.

[653] Le Bars PL, Kieser M, Itil KZ (2000) A 26-week analysisof a double-blind, placebo-controlled trial of the ginkgobiloba extract EGb 761 in dementia. Dement Geriatr CognDisord 11, 230-237.

[654] Le Bars PL, Velasco FM, Ferguson JM, Dessain EC, KieserM, Hoerr R (2002) Influence of the severity of cognitiveimpairment on the effect of the Gnkgo biloba extract EGb761 in Alzheimer’s disease. Neuropsychobiology 45, 19-26.

Page 77: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 77

[655] Le Bars PL (2003) Magnitude of effect and specialapproach to Ginkgo biloba extract EGb 761 in cognitivedisorders. Pharmacopsychiatry 36(Suppl 1), S44-S49.

[656] Le Bars PL (2003) Response patterns of EGb 761 inAlzheimer’s disease: Influence of neuropsychological pro-files. Pharmacopsychiatry 36(Suppl 1), S50-S55.

[657] Oken BS, Storzbach DM, Kaye JA (1998) The efficacy ofGinkgo biloba on cognitive function in Alzheimer disease.Arch Neurol 55, 1409-1415.

[658] Mix JA, Crews WD Jr (2000) An examination of theefficacy of Ginkgo biloba extract EGb761 on the neuropsy-chologic functioning of cognitively intact older adults.J Altern Complement Med 6, 219-229.

[659] Mix JA, Crews WD Jr (2002) A double-blind, placebo-controlled, randomized trial of Ginkgo biloba extractEGb 761 in a sample of cognitively intact older adults:Neuropsychological findings. Hum Psychopharmacol 17,267-277.

[660] Dekosky ST, Fitzpatrick A, Ives DG, Saxton J, WilliamsonJ, Lopez OL, Burke G, Fried L, Kuller LH, RobbinsJ, Tracy R, Woolard N, Dunn L, Kronmal R, Nahin R,Furberg C (2006) The Ginkgo Evaluation of Memory(GEM) study: Design and baseline data of a randomizedtrial of Ginkgo biloba extract in prevention of dementia.Contemp Clin Trials 27, 238-253.

[661] Dekosky ST, Furberg CD (2008) Turning over a new leaf:Ginkgo biloba in prevention of dementia? Neurology 70,1730-1731.

[662] Mazza M, Capuano A, Bria P, Mazza S (2006) Ginkgobiloba and donepezil: A comparison in the treatment ofAlzheimer’s dementia in a randomized placebo-controlleddouble-blind study. Eur J Neurol 13, 981-985.

[663] Snitz BE, O’Meara ES, Carlson MC, Arnold AM, IvesDG, Rapp SR, Saxton J, Lopez OL, Dunn LO, Sink KM,Dekosky ST (2009) Ginkgo biloba for preventing cognitivedecline in older adults: A randomized trial. JAMA 302,2663-2670.

[664] Yancheva S, Ihl R, Nikolova G, Panayotov P, Schlae-fke S, Hoerr R (2009) Ginkgo biloba extract EGb761(R), donepezil or both combined in the treatment ofAlzheimer’s disease with neuropsychiatric features: Arandomised, double-blind, exploratory trial. Aging MentHealth 13, 183-190.

[665] Janssen IM, Sturtz S, Skipka G, Zentner A, Velasco GM,Busse R (2010) Ginkgo biloba in Alzheimer’s disease:A systematic review. Wien Med Wochenschr 160, 539-546.

[666] Wang J, Ho L, Zhao W, Ono K, Rosensweig C, ChenL, Humala N, Teplow DB, Pasinetti GM (2008) Grape-derived polyphenolics prevent Abeta oligomerization andattenuate cognitive deterioration in a mouse model ofAlzheimer’s disease. J Neurosci 28, 6388-6392.

[667] Ksiezak-Reding H, Ho L, Santa-Maria I, az-Ruiz C,Wang J, Pasinetti GM (2012) Ultrastructural alterations ofAlzheimer’s disease paired helical filaments by grape seed-derived polyphenols. Neurobiol Aging 33, 1427-1439.

[668] Tarozzi A, Merlicco A, Morroni F, Bolondi C, Di IP, Cic-carelli R, Romano S, Giuliani P, Hrelia P (2010) Guanosineprotects human neuroblastoma cells from oxidative stressand toxicity induced by Amyloid-beta peptide oligomers.J Biol Regul Homeost Agents 24, 297-306.

[669] Bao Q, Luo Y, Li W, Sun X, Zhu C, Li P, Huang ZX, Tan X(2011) The mechanism for heme to prevent Abeta(1-40)aggregation and its cytotoxicity. J Biol Inorg Chem 16,809-816.

[670] Dong X, Geng M, Guan H (2003) Effects of the marineacidic oligose on dopamine release from striatum andamygdala in the rat model of Parkinson’s disease. ChinJ Marine Drugs 22, 9-12.

[671] Zeng KW, Wang XM, Ko H, Kwon HC, Cha JW, YangHO (2011) Hyperoside protects primary rat cortical neu-rons from neurotoxicity induced by amyloid beta-proteinvia the PI3K/Akt/Bad/Bcl(XL)-regulated mitochondrialapoptotic pathway. Eur J Pharmacol 672, 45-55.

[672] Nie J, Luo Y, Huang XN, Gong QH, Wu Q, Shi JS(2010) Icariin inhibits beta-amyloid peptide segment25-35 induced expression of beta-secretase in rat hip-pocampus. Eur J Pharmacol 626, 213-218.

[673] Zeng KW, Ko H, Yang HO, Wang XM (2010) Icariinattenuates beta-amyloid-induced neurotoxicity by inhibi-tion of tau protein hyperphosphorylation in PC12 cells.Neuropharmacology 59, 542-550.

[674] Zeng KW, Fu H, Liu GX, Wang XM (2010) Icariinattenuates lipopolysaccharide-induced microglial acti-vation and resultant death of neurons by inhibitingTAK1/IKK/NF-kappaB and JNK/p38 MAPK pathways.Int Immunopharmacol 10, 668-678.

[675] Urano T, Tohda C (2010) Icariin improves memory impair-ment in Alzheimer’s disease model mice (5xFAD) andattenuates amyloid beta-induced neurite atrophy. Phy-tother Res 24, 1658-1663.

[676] Zheng M, Qu L, Lou Y (2008) Effects of icariin combinedwith Panax notoginseng saponins on ischemia reperfusion-induced cognitive impairments related with oxidativestress and CA1 of hippocampal neurons in rat. PhytotherRes 22, 597-604.

[677] Carvajal FJ, Inestrosa NC (2011) Interactions of AChEwith Abeta aggregates in Alzheimer’s brain: Therapeuticrelevance of IDN 5706. Front Mol Neurosci 4, 19.

[678] Roth A, Schaffner W, Hertel C (1999) Phytoestrogenkaempferol (3,4′,5,7-tetrahydroxyflavone) protects PC12and T47D cells from beta-amyloid-induced toxicity. J Neu-rosci Res 57, 399-404.

[679] Kim JK, Choi SJ, Cho HY, Hwang HJ, Kim YJ, Lim ST,Kim CJ, Kim HK, Peterson S, Shin DH (2010) Protec-tive effects of kaempferol (3,4′,5,7-tetrahydroxyflavone)against amyloid beta peptide (Abeta)-induced neurotoxic-ity in ICR mice. Biosci Biotechnol Biochem 74, 397-401.

[680] Lee KY, Sung SH, Kim SH, Jang YP, Oh TH, Kim YC(2009) Cognitive-enhancing activity of loganin isolatedfrom Cornus officinalis in scopolamine-induced amnesicmice. Arch Pharm Res 32, 677-683.

[681] Tsai FS, Peng WH, Wang WH, Wu CR, Hsieh CC, Lin YT,Feng IC, Hsieh MT (2007) Effects of luteolin on learningacquisition in rats: involvement of the central cholinergicsystem. Life Sci 80, 1692-1698.

[682] Tsai FS, Cheng HY, Hsieh MT, Wu CR, Lin YC, PengWH (2010) The ameliorating effects of luteolin on beta-amyloid-induced impairment of water maze performanceand passive avoidance in rats. Am J Chin Med 38, 279-291.

[683] Cheng HY, Hsieh MT, Tsai FS, Wu CR, Chiu CS, Lee MM,Xu HX, Zhao ZZ, Peng WH (2010) Neuroprotective effectof luteolin on amyloid beta protein (25-35)-induced toxic-ity in cultured rat cortical neurons. Phytother Res 24(Suppl1), S102-S108.

[684] Xu B, Li XX, He GR, Hu JJ, Mu X, Tian S, Du GH (2010)Luteolin promotes long-term potentiation and improvescognitive functions in chronic cerebral hypoperfused rats.Eur J Pharmacol 627, 99-105.

Page 78: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

78 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[685] Parachikova A, Green KN, Hendrix C, LaFerla FM (2010)Formulation of a medical food cocktail for Alzheimer’sdisease: Beneficial effects on cognition and neuropathol-ogy in a mouse model of the disease. PLoS One 5,e14015.

[686] Pappolla MA, Sos M, Omar RA, Bick RJ, Hickson-BickDL, Reiter RJ, Efthimiopoulos S, Robakis NK (1997)Melatonin prevents death of neuroblastoma cells exposedto the Alzheimer amyloid peptide. J Neurosci 17, 1683-1690.

[687] Pappolla M, Bozner P, Soto C, Shao H, Robakis NK,Zagorski M, Frangione B, Ghiso J (1998) Inhibition ofAlzheimer beta-fibrillogenesis by melatonin. J Biol Chem273, 7185-7188.

[688] Pappolla MA, Chyan YJ, Poeggeler B, Frangione B,Wilson G, Ghiso J, Reiter RJ (2000) An assessment ofthe antioxidant and the antiamyloidogenic properties ofmelatonin: Implications for Alzheimer’s disease. J NeuralTransm 107, 203-231.

[689] Pappolla MA, Simovich MJ, Bryant-Thomas T, ChyanYJ, Poeggeler B, Dubocovich M, Bick R, Perry G,Cruz-Sanchez F, Smith MA (2002) The neuroprotectiveactivities of melatonin against the Alzheimer beta-proteinare not mediated by melatonin membrane receptors.J Pineal Res 32, 135-142.

[690] Wang JZ, Wang ZF (2006) Role of melatonin inAlzheimer-like neurodegeneration. Acta Pharmacol Sin27, 41-49.

[691] Cheng Y, Feng Z, Zhang QZ, Zhang JT (2006) Beneficialeffects of melatonin in experimental models of Alzheimerdisease. Acta Pharmacol Sin 27, 129-139.

[692] Srinivasan V, Kaur C, Pandi-Perumal S, Brown GM, Car-dinali DP (2010) Melatonin and its agonist ramelteonin Alzheimer’s disease: Possible therapeutic value. Int JAlzheimers Dis 2011, 741974.

[693] Srinivasan V, Cardinali DP, Srinivasan US, Kaur C, BrownGM, Spence DW, Hardeland R, Pandi-Perumal SR (2011)Therapeutic potential of melatonin and its analogs inParkinson’s disease: Focus on sleep and neuroprotection.Ther Adv Neurol Disord 4, 297-317.

[694] Argyriou A, Prast H, Philippu A (1998) Melatoninfacilitates short-term memory. Eur J Pharmacol 349,159-162.

[695] Bhadania M, Joshi H, Patel P, Kulkarni VH (2012) Pro-tective effect of menthol on beta-amyloid peptide inducedcognitive deficits in mice. Eur J Pharmacol 681, 50-54.

[696] Lemkul JA, Bevan DR (2010) Destabilizing Alzheimer’sAbeta(42) protofibrils with morin: Mechanistic insightsfrom molecular dynamics simulations. Biochemistry 49,3935-3946.

[697] Lemkul JA, Bevan DR (2012) Morin inhibits the earlystages of amyloid beta-peptide aggregation by altering ter-tiary and quaternary interactions to produce “off-pathway”structures. Biochemistry 51, 5990-6009.

[698] Gong EJ, Park HR, Kim ME, Piao S, Lee E, Jo DG, ChungHY, Ha NC, Mattson MP, Lee J (2011) Morin attenuates tauhyperphosphorylation by inhibiting GSK3beta. NeurobiolDis 44, 223-230.

[699] DeToma AS, Choi JS, Braymer JJ, Lim MH (2011)Myricetin: A naturally occurring regulator of metal-induced amyloid-beta aggregation and neurotoxicity.Chembiochem 12, 1198-1201.

[700] Jones JR, Lebar MD, Jinwal UK, Abisambra JF, KorenJ III, Blair L, O’Leary JC, Davey Z, Trotter J, John-son AG, Weeber E, Eckman CB, Baker BJ, Dickey CA

(2011) The diarylheptanoid (+)-aR,11S-myricanol andtwo flavones from bayberry (Myrica cerifera) destabilizethe microtubule-associated protein tau. J Nat Prod 74,38-44.

[701] Maratha SR, Mahadevan N (2012) Memory enhancingactivity of naringin in unstressed and stressed mice: Possi-ble cholinergic and nitriergic modulation. Neurochem Res37, 2206-2212.

[702] Ono K, Hasegawa K, Yoshiike Y, Takashima A, YamadaM, Naiki H (2002) Nordihydroguaiaretic acid potentlybreaks down pre-formed Alzheimer’s beta-amyloid fibrilsin vitro. J Neurochem 81, 434-440.

[703] Moss MA, Varvel NH, Nichols MR, Reed DK, RosenberryTL (2004) Nordihydroguaiaretic acid does not disag-gregate beta-amyloid(1-40) protofibrils but does inhibitgrowth arising from direct protofibril association. MolPharmacol 66, 592-600.

[704] Bieschke J, Herbst M, Wiglenda T, Friedrich RP, Boed-drich A, Schiele F, Kleckers D, Lopez Del Amo JM,Gruning BA, Wang Q, Schmidt MR, Lurz R, Anwyl R,Schnoegl S, Fandrich M, Frank RF, Reif B, Gunther S,Walsh DM, Wanker EE (2012) Small-molecule conversionof toxic oligomers to nontoxic beta-sheet-rich amyloidfibrils. Nat Chem Biol 8, 93-101.

[705] Choi DY, Lee YJ, Lee SY, Lee YM, Lee HH, Choi IS, OhKW, Han SB, Nam SY, Hong JT (2012) Attenuation ofscopolamine-induced cognitive dysfunction by obovatol.Arch Pharm Res 35, 1279-1286.

[706] Choi DY, Lee JW, Peng J, Lee YJ, Han JY, Lee YH, ChoiIS, Han SB, Jung JK, Lee WS, Lee SH, Kwon BM, Oh KW,Hong JT (2012) Obovatol improves cognitive functions inanimal models for Alzheimer’s disease. J Neurochem 120,1048-1059.

[707] Choi DY, Lee JW, Lin G, Lee YK, Lee YH, Choi IS,Han SB, Jung JK, Kim YH, Kim KH, Oh KW, Hong JT,Lee MS (2012) Obovatol attenuates LPS-induced memoryimpairments in mice via inhibition of NF-kappaB signal-ing pathway. Neurochem Int 60, 68-77.

[708] Choi DY, Lee YJ, Hong JT, Lee HJ (2012) Antioxidantproperties of natural polyphenols and their therapeuticpotentials for Alzheimer’s disease. Brain Res Bull 87, 144-153.

[709] Daccache A, Lion C, Sibille N, Gerard M, Slomianny C,Lippens G, Cotelle P (2011) Oleuropein and derivativesfrom olives as Tau aggregation inhibitors. Neurochem Int58, 700-707.

[710] Yu CJ, Zheng MF, Kuang CX, Huang WD, Yang Q(2010) Oren-gedoku-to and its constituents with therapeu-tic potential in Alzheimer’s disease inhibit indoleamine2, 3-dioxygenase activity in vitro. J Alzheimers Dis 22,257-266.

[711] Kim DH, Jeon SJ, Son KH, Jung JW, Lee S, Yoon BH,Choi JW, Cheong JH, Ko KH, Ryu JH (2006) Effect of theflavonoid, oroxylin A, on transient cerebral hypoperfusion-induced memory impairment in mice. Pharmacol BiochemBehav 85, 658-668.

[712] Fujiwara H, Tabuchi M, Yamaguchi T, Iwasaki K,Furukawa K, Sekiguchi K, Ikarashi Y, Kudo Y, HiguchiM, Saido TC, Maeda S, Takashima A, Hara M,Yaegashi N, Kase Y, Arai H (2009) A traditionalmedicinal herb Paeonia suffruticosa and its active con-stituent 1,2,3,4,6-penta-O-galloyl-beta-D-glucopyranosehave potent anti-aggregation effects on Alzheimer’s amy-loid beta proteins in vitro and in vivo. J Neurochem 109,1648-1657.

Page 79: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 79

[713] Kim HJ, Lee KW, Lee HJ (2007) Protective effects ofpiceatannol against beta-amyloid-induced neuronal celldeath. Ann N Y Acad Sci 1095, 473-482.

[714] Liu R, Wu CX, Zhou D, Yang F, Tian S, ZhangL, Zhang TT, Du GH (2012) Pinocembrin protectsagainst beta-amyloid-induced toxicity in neurons throughinhibiting receptor for advanced glycation end products(RAGE)-independent signaling pathways and regulatingmitochondrion-mediated apoptosis. BMC Med 10, 105.

[715] Gazit E (2002) A possible role for pi-stacking in the self-assembly of amyloid fibrils. FASEB J 16, 77-83.

[716] Smid SD, Maag JL, Musgrave IF (2012) Dietarypolyphenol-derived protection against neurotoxic beta-amyloid protein: From molecular to clinical. Food Funct3, 1242-1250.

[717] Xu J, Rong S, Xie B, Sun Z, Deng Q, Wu H, Bao W, WangD, Yao P, Huang F, Liu L (2010) Memory impairment incognitively impaired aged rats associated with decreasedhippocampal CREB phosphorylation: Reversal by pro-cyanidins extracted from the lotus seedpod. J Gerontol ABiol Sci Med Sci 65, 933-940.

[718] Xu J, Rong S, Xie B, Sun Z, Deng Q, Bao W, Wang D, YaoP, Huang F, Liu L (2011) Changes in the nitric oxide systemcontribute to effect of procyanidins extracted from the lotusseedpod ameliorating memory impairment in cognitivelyimpaired aged rats. Rejuvenation Res 14, 33-43.

[719] Bithu BS, Reddy NR, Prasad SK, Sairam K, HemalathaS (2012) Prosopis cineraria: A potential nootropic agent.Pharm Biol 50, 1241-1247.

[720] Li J, Wang G, Liu J, Zhou L, Dong M, Wang R, Li X,Li X, Lin C, Niu Y (2010) Puerarin attenuates amyloid-beta-induced cognitive impairment through suppression ofapoptosis in rat hippocampus in vivo. Eur J Pharmacol649, 195-201.

[721] Wang G, Zhou L, Zhang Y, Dong M, Li X, Liu J, NiuY (2011) Implication of the c-Jun-NH2-terminal kinasepathway in the neuroprotective effect of puerarin against1-methyl-4-phenylpyridinium (MPP+)-induced apoptosisin PC-12 cells. Neurosci Lett 487, 88-93.

[722] Xing G, Dong M, Li X, Zou Y, Fan L, Wang X, Cai D,Li C, Zhou L, Liu J, Niu Y (2011) Neuroprotective effectsof puerarin against beta-amyloid-induced neurotoxicity inPC12 cells via a PI3K-dependent signaling pathway. BrainRes Bull 85, 212-218.

[723] Lin F, Xie B, Cai F, Wu G (2012) Protective effect ofPuerarin on beta-amyloid-induced neurotoxicity in rat hip-pocampal neurons. Arzneimittelforschung 62, 187-193.

[724] Peng QL, Buz’Zard AR, Lau BH (2002) Pycnogenolprotects neurons from amyloid-beta peptide-inducedapoptosis. Brain Res Mol Brain Res 104, 55-65.

[725] Ansari MA, Abdul HM, Joshi G, Opii WO, Butterfield DA(2009) Protective effect of quercetin in primary neuronsagainst Abeta(1-42): Relevance to Alzheimer’s disease.J Nutr Biochem 20, 269-275.

[726] Iuvone T, De FD, Esposito G, D’Amico A, Izzo AA (2006)The spice sage and its active ingredient rosmarinic acidprotect PC12 cells from amyloid-beta peptide-inducedneurotoxicity. J Pharmacol Exp Ther 317, 1143-1149.

[727] 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.

[728] Wang SW, Wang YJ, Su YJ, Zhou WW, Yang SG, ZhangR, Zhao M, Li YN, Zhang ZP, Zhan DW, Liu RT (2012)Rutin inhibits beta-amyloid aggregation and cytotoxicity,

attenuates oxidative stress, and decreases the productionof nitric oxide and proinflammatory cytokines. Neurotox-icology 33, 482-490.

[729] Papandreou MA, Kanakis CD, Polissiou MG,Efthimiopoulos S, Cordopatis P, Margarity M, Lamari FN(2006) Inhibitory activity on amyloid-beta aggregationand antioxidant properties of Crocus sativus stigmasextract and its crocin constituents. J Agric Food Chem 54,8762-8768.

[730] Papandreou MA, Tsachaki M, Efthimiopoulos S, Cor-dopatis P, Lamari FN, Margarity M (2011) Memoryenhancing effects of saffron in aged mice are correlatedwith antioxidant protection. Behav Brain Res 219, 197-204.

[731] Akhondzadeh S, Sabet MS, Harirchian MH, Togha M,Cheraghmakani H, Razeghi S, Hejazi SS, Yousefi MH,Alimardani R, Jamshidi A, Zare F, Moradi A (2010) Saf-fron in the treatment of patients with mild to moderateAlzheimer’s disease: A 16-week, randomized and placebo-controlled trial. J Clin Pharm Ther 35, 581-588.

[732] Akhondzadeh S, Shafiee SM, Harirchian MH, Togha M,Cheraghmakani H, Razeghi S, Hejazi SS, Yousefi MH, Ali-mardani R, Jamshidi A, Rezazadeh SA, Yousefi A, ZareF, Moradi A, Vossoughi A (2010) A 22-week, multicen-ter, randomized, double-blind controlled trial of Crocussativus in the treatment of mild-to-moderate Alzheimer’sdisease. Psychopharmacology (Berl) 207, 637-643.

[733] Geromichalos GD, Lamari FN, Papandreou MA, TrafalisDT, Margarity M, Papageorgiou A, Sinakos Z (2012) Saf-fron as a source of novel acetylcholinesterase inhibitors:Molecular docking and in vitro enzymatic studies. J AgricFood Chem 60, 6131-6138.

[734] Zhang L, Yu H, Zhao X, Lin X, Tan C, Cao G, Wang Z(2010) Neuroprotective effects of salidroside against beta-amyloid-induced oxidative stress in SH-SY5Y humanneuroblastoma cells. Neurochem Int 57, 547-555.

[735] Ito Y, Kosuge Y, Sakikubo T, Horie K, Ishikawa N,Obokata N, Yokoyama E, Yamashina K, Yamamoto M,Saito H, Arakawa M, Ishige K (2003) Protective effectof S-allyl-L-cysteine, a garlic compound, on amyloidbeta-protein-induced cell death in nerve growth factor-differentiated PC12 cells. Neurosci Res 46, 119-125.

[736] Ito Y, Ito M, Takagi N, Saito H, Ishige K (2003) Neurotox-icity induced by amyloid beta-peptide and ibotenic acid inorganotypic hippocampal cultures: Protection by S-allyl-L-cysteine, a garlic compound. Brain Res 985, 98-107.

[737] Lu P, Mamiya T, Lu LL, Mouri A, Zou L, Nagai T,Hiramatsu M, Ikejima T, Nabeshima T (2009) Silibininprevents amyloid beta peptide-induced memory impair-ment and oxidative stress in mice. Br J Pharmacol 157,1270-1277.

[738] Lee HE, Kim DH, Park SJ, Kim JM, Lee YW, Jung JM,Lee CH, Hong JG, Liu X, Cai M, Park KJ, Jang DS, Ryu JH(2012) Neuroprotective effect of sinapic acid in a mousemodel of amyloid beta(1-42) protein-induced Alzheimer’sdisease. Pharmacol Biochem Behav 103, 260-266.

[739] Scheltens P, Kamphuis PJ, Verhey FR, Olde Rikkert MG,Wurtman RJ, Wilkinson D, Twisk JW, Kurz A (2010) Effi-cacy of a medical food in mild Alzheimer’s disease: Arandomized, controlled trial. Alzheimers Dement 6, 1-10.

[740] Alzheimer Research Forum (2011) Nutrient formula-tion appears to grease memory function, http://www.alzforum.org/new/detail.asp?id=2957.

[741] Cummings JL (2012) Food for thought: Souvenaid(R) inmild Alzheimer’s disease. J Alzheimers Dis 31, 237-238.

Page 80: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

80 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[742] Pieri M, Amadoro G, Carunchio I, Ciotti MT, QuaresimaS, Florenzano F, Calissano P, Possenti R, Zona C, Sev-erini C (2010) SP protects cerebellar granule cells againstbeta-amyloid-induced apoptosis by down-regulation andreduced activity of Kv4 potassium channels. Neurophar-macology 58, 268-276.

[743] Yang EJ, Kim SI, Ku HY, Lee DS, Lee JW, Kim YS, SeongYH, Song KS (2010) Syringin from stem bark of Fraxinusrhynchophylla protects Abeta(25-35)-induced toxicity inneuronal cells. Arch Pharm Res 33, 531-538.

[744] Ono K, Hasegawa K, Naiki H, Yamada M (2004) Anti-amyloidogenic activity of tannic acid and its activityto destabilize Alzheimer’s beta-amyloid fibrils in vitro.Biochim Biophys Acta 1690, 193-202.

[745] Inbar P, Bautista MR, Takayama SA, Yang J (2008) Assayto screen for molecules that associate with Alzheimer’srelated beta-amyloid fibrils. Anal Chem 80, 3502-3506.

[746] Mori T, Rezai-Zadeh K, Koyama N, Arendash GW, Yam-aguchi H, Kakuda N, Horikoshi-Sakuraba Y, Tan J, Town T(2012) Tannic acid is a natural beta-secretase inhibitor thatprevents cognitive impairment and mitigates Alzheimer-like pathology in transgenic mice. J Biol Chem 287,6912-6927.

[747] Yang SG, Wang WY, Ling TJ, Feng Y, Du XT, ZhangX, Sun XX, Zhao M, Xue D, Yang Y, Liu RT (2010)alpha-Tocopherol quinone inhibits beta-amyloid aggrega-tion and cytotoxicity, disaggregates preformed fibrils anddecreases the production of reactive oxygen species, NOand inflammatory cytokines. Neurochem Int 57, 914-922.

[748] Yang ZQ, Yang SF, Yang JQ, Zhou QX, Li SL (2007)Protective effects and mechanism of total coptis alkaloidson a beta 25-35 induced learning and memory dysfunctionin rats. Chin J Integr Med 13, 50-54.

[749] Hong SY, Jeong WS, Jun M (2012) Protective effects of thekey compounds isolated from Corni fructus against beta-amyloid-induced neurotoxicity in PC12 cells. Molecules17, 10831-10845.

[750] Takasaki J, Ono K, Yoshiike Y, Hirohata M, Ikeda T,Morinaga A, Takashima A, Yamada M (2011) Vitamin Ahas anti-oligomerization effects on amyloid-beta in vitro.J Alzheimers Dis 27, 271-280.

[751] Kulkarni SK, Dhir A (2008) Withania somnifera: AnIndian ginseng. Prog Neuropsychopharmacol Biol Psychi-atry 32, 1093-1105.

[752] Sehgal N, Gupta A, Valli RK, Joshi SD, Mills JT, HamelE, Khanna P, Jain SC, Thakur SS, Ravindranath V (2012)Withania somnifera reverses Alzheimer’s disease pathol-ogy by enhancing low-density lipoprotein receptor-relatedprotein in liver. Proc Natl Acad Sci U S A 109, 3510-3515.

[753] Wang XM, Fu H, Liu GX, Zhu W, Li L, Yang JX (2007)Effect of modified wuzi yanzong granule on patients withmild cognitive impairment from oxidative damage aspect.Chin J Integr Med 13, 258-263.

[754] Hayashi Y, Ishida Y, Inoue T, Udagawa M, Takeuchi K,Yoshimuta H, Kiue K, Ninomiya Y, Kawano J, SameshimaT, Kawahara T, Goto I, Shudo K, Kurayama S, Naka-mura J, Okahara K, Mitsuyama Y (2010) Treatment ofbehavioral and psychological symptoms of Alzheimer-type dementia with Yokukansan in clinical practice. ProgNeuropsychopharmacol Biol Psychiatry 34, 541-545.

[755] Uchida N, Takasaki K, Sakata Y, Nogami A, OishiH, Watanabe T, Shindo T, Egashira N, Kubota K,Katsurabayashi S, Mishima K, Fujiwara M, Nishimura R,Iwasaki K (2012) Cholinergic involvement and synapticdynamin 1 expression in yokukansan-mediated improve-

ment of spatial memory in a rat model of early Alzheimer’sdisease. Phytother Res, doi: 10.1002/ptr.4818 [Epub aheadof print].

[756] Tohda C, Tamura T, Komatsu K (2003) Repair of amyloidbeta(25-35)-induced memory impairment and synapticloss by a Kampo formula, Zokumei-to. Brain Res 990,141-147.

[757] Takahashi K, Matsumura T, Ishihara J, Hatakeyama S(2007) A highly stereocontrolled total synthesis of dysi-herbaine. Chem Commun (Camb) 4158-4160.

[758] Nakanishi K (2005) Terpene trilactones from Gingkobiloba: From ancient times to the 21st century. Bioorg MedChem 13, 4987-5000.

[759] Keowkase R, Aboukhatwa M, Adam BL, Beach JW, TerryAV Jr, Buccafussco JJ, Luo Y (2010) Neuroprotectiveeffects and mechanism of cognitive-enhancing cholineanalogs JWB 1-84-1 and JAY 2-22-33 in neuronal cultureand Caenorhabditis elegans. Mol Neurodegener 5, 59.

[760] Sood A, Warren BJ, Webster SJ, Terry AV, Buccafusco JJ(2007) The effects of JWB1-84-1 on memory-related taskperformance by amyloid Abeta transgenic mice and byyoung and aged monkeys. Neuropharmacology 53, 588-600.

[761] Shin KY, Lee GH, Park CH, Kim HJ, Park SH, Kim S,Kim HS, Lee KS, Won BY, Lee HG, Choi JH, Suh YH(2007) A novel compound, maltolyl p-coumarate, attenu-ates cognitive deficits and shows neuroprotective effectsin vitro and in vivo dementia models. J Neurosci Res 85,2500-2511.

[762] Guan J, Zhang R, le-Gandar L, Hodgkinson S, VickersMH (2010) NNZ-2591, a novel diketopiperazine, pre-vented scopolamine-induced acute memory impairment inthe adult rat. Behav Brain Res 210, 221-228.

[763] Wang HY, Bakshi K, Frankfurt M, Stucky A, GoberdhanM, Shah SM, Burns LH (2012) Reducing amyloid-relatedAlzheimer’s disease pathogenesis by a small molecule tar-geting filamin a. J Neurosci 32, 9773-9784.

[764] Zs-Nagy I, Ohta M, Kitani K (1989) Effect of centro-phenoxine and BCE-001 treatment on lateral diffusion ofproteins in the hepatocyte plasma membrane as revealedby fluorescence recovery after photobleaching in rat liversmears. Exp Gerontol 24, 317-330.

[765] Zs-Nagy I (1994) A survey of the available data on a newnootropic drug, BCE-001. Ann N Y Acad Sci 717, 102-114.

[766] Nagy K, Dajko G, Uray I, Zs-Nagy I (1994) Compara-tive studies on the free radical scavenger properties of twonootropic drugs, CPH and BCE-001. Ann N Y Acad Sci717, 115-121.

[767] Scopes D, O’Hare E, Jeggo R, Whyment A, Spanswick D,Kim EM, Gannon J, Amijee H, Treherne J (2012) Abetaoligomer toxicity inhibitor protects memory in models ofsynaptic toxicity. Br J Pharmacol 167, 383-392.

[768] Pierce JE, Smith DH, Eison MS, McIntosh TK (1993) Thenootropic compound BMY-21502 improves spatial learn-ing ability in brain injured rats. Brain Res 624, 199-208.

[769] Saletu B, Schulz H, Herrmann WM, Anderer P, ShrotriyaRC, Vanbrabant E (1994) BMS-181168 for protection ofthe human brain against hypoxia: Double-blind, placebo-controlled EEG mapping studies. Pharmacopsychiatry 27,189-197.

[770] Shrotriya RC, Cutler NR, Sramek JJ, Veroff AE, Hiron-aka DY (1996) Efficacy and safety of BMY 21,502 inAlzheimer disease. Ann Pharmacother 30, 1376-1380.

[771] Lishko PV, Maximyuk OP, Chatterjee SS, Noldner M,Krishtal OA (1998) The putative cognitive enhancer KA-

Page 81: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 81

672.HCl is an uncompetitive voltage-dependent NMDAreceptor antagonist. Neuroreport 9, 4193-4197.

[772] Sourgens H, Hoerr R, Biber A, Steinbrede H, Derendorf H(1998) KA 672-HCl, a neuronal activator against demen-tia: Tolerability, safety, and preliminary pharmacokineticsafter single and multiple oral doses in healthy male andfemale volunteers. J Clin Pharmacol 38, 373-381.

[773] Winter JC, Helsley SE, Rabin RA (1998) The discrim-inative stimulus effects of KA 672, a putative cognitiveenhancer: Evidence for a 5-HT1A component. PharmacolBiochem Behav 60, 703-707.

[774] Reisner E, Noldner M, Rossner S, Chatterjee SS, Bigl V,Schliebs R (1999) Acute effect of KA-672, a putative cog-nitive enhancer, on neurotransmitter receptor binding inmouse brain. Neurosci Lett 274, 187-190.

[775] Teismann P, Ferger B (2000) In vivo effects of the putativecognitive enhancer KA-672.HCl in comparison with 8-hydroxy-2-(di-N-propylamino) tetralin and haloperidol ondopamine, 3,4-dihydroxyphenylacetic acid, serotonin and5-hydroxyindoleacetic acid levels in striatal and corticalbrain regions. Prog Neuropsychopharmacol Biol Psychia-try 24, 337-348.

[776] Teismann P, Ferger B (2000) Effects of ensaculin ondopamine metabolite levels and K(+)-induced glutamaterelease. Eur J Pharmacol 398, 247-250.

[777] Hoerr R, Noeldner M (2002) Ensaculin (KA-672 HCl):a multitransmitter approach to dementia treatment. CNSDrug Rev 8, 143-158.

[778] Sramek JJ, Cutler NR (2000) Ongoing trials in Alzheimer’sdisease. Expert Opin Investig Drugs 9, 899-915.

[779] Inozemtsev AN, Garibova TL, Khromova IV, Alvarez R,Voronina TA, Tushmalova NA (1993) [The effect of noog-lutil and piracetam on different forms of operant learning].Eksp Klin Farmakol 56, 6-8.

[780] Mondadori C, Ducret T, Borkowski J (1991) How longdoes ‘memory consolidation’ take? New compounds canimprove retention performance, even if administered upto 24 hours after the learning experience. Brain Res 555,107-111.

[781] Mondadori C, Buerki H, Borkowski J, Radeke E, DucretT, Glatt A (1992) CGS 5649 B, a new compound, reversesage-related cognitive dysfunctions in rats. Behav NeuralBiol 57, 149-156.

[782] Nilsson E, Schneider T, Hoffmann E, Schmidt EK (1991)Urinary excretion of CGS 5,649 and its conjugatedglucuronide and sulfate in man. Eur J Drug Metab Phar-macokinet Spec No 3, 201-204.

[783] Molloy DW, Guyatt GH, Standish T, Willan A, McIlroyW, D’Souza J, Brown G, Mondadori C (1993) Effect of anew nootropic agent, CGS 5649B, on cognition, function,and behavior in dementia. J Gen Intern Med 8, 444-447.

[784] Clincke GH, Tritsmans L, Idzikowski C, Amery WK,Janssen PA (1988) The effect of R 58 735 (Sabeluzole)on memory functions in healthy elderly volunteers. Psy-chopharmacology (Berl) 94, 52-57.

[785] Clincke GH, Tritsmans L (1988) Sabeluzole (R58 735)increases consistent retrieval during serial learning andrelearning of nonsense syllables. Psychopharmacology(Berl) 96, 309-310.

[786] Tritsmans L, Clincke G, Amery WK (1988) The effectof sabeluzole (R 58735) on memory retrieval functions.Psychopharmacology (Berl) 94, 527-531.

[787] Geerts H, Nuydens R, Nuyens R, Cornelissen F (1992)Sabeluzole accelerates neurite outgrowth in different neu-ronal cell lines. Restor Neurol Neurosci 4, 21-32.

[788] Geerts H, Nuydens R, Nuyens R, Cornelissen F, De BM,Pauwels P, Janssen PA, Song YH, Mandelkow EM (1992)Sabeluzole, a memory-enhancing molecule, increases fastaxonal transport in neuronal cell cultures. Exp Neurol 117,36-43.

[789] Geerts H, Nuydens R, de JM, Cornelissen F, NuyensR, Wouters L (1996) Sabeluzole stabilizes the neuronalcytoskeleton. Neurobiol Aging 17, 573-581.

[790] Wong YN, Quon CY, Holm KA, Burcham DL, FreyNL, Huang SM, Lam GN (1996) Pharmacokinetics andmetabolism of EXP921, a novel cognitive enhancer, in rats.Drug Metab Dispos 24, 172-179.

[791] Saletu B, Semlitsch HV, Anderer P, Resch F, PresslichO, Schuster P (1989) Psychophysiological research inpsychiatry and neuropsychopharmacology. II. The investi-gation of antihypoxidotic/nootropic drugs (tenilsetam andco-dergocrine-mesylate) in elderlies with the ViennesePsychophysiological Test-System (VPTS). Methods FindExp Clin Pharmacol 11, 43-55.

[792] Webster J, Urban C, Berbaum K, Loske C, Alpar A,Gartner U, de Arriba SG, Arendt T, Munch G (2005)The carbonyl scavengers aminoguanidine and tenilsetamprotect against the neurotoxic effects of methylglyoxal.Neurotox Res 7, 95-101.

[793] Rainer M, Brunnbauer M, Dunky A, Ender F, GoldsteinerH, Holl O, Kotlan P, Paulitsch G, Reiner C, Stossl J,Zachhuber C, Mossler H (1997) Therapeutic results withCerebrolysin in the treatment of dementia. Wien MedWochenschr 147, 426-431.

[794] Xiong H, Baskys A, Wojtowicz JM (1996) Brain-derivedpeptides inhibit synaptic transmission via presynapticGABAB receptors in CA1 area of rat hippocampal slices.Brain Res 737, 188-194.

[795] Kofler B, Erhart C, Erhart P, Harrer G (1990) A mul-tidimensional approach in testing nootropic drug effects(Cerebrolysin). Arch Gerontol Geriatr 10, 129-140.

[796] Hutter-Paier B, Eggenreich U, Windisch M (1996)Effects of two protein-free peptide derivatives on passiveavoidance behaviour of 24-month-old rats. Arzneimit-telforschung 46, 237-241.

[797] Hutter-Paier B, Eggenreich U, Windisch M (1996) Dose-dependent behavioural effects of two protein-free peptidederivatives on the passive avoidance reaction of rats.Arzneimittelforschung 46, 242-246.

[798] Gschanes A, Windisch M (1996) The influence of cere-brolysin and E021 on spatial navigation of young rats. JNeural Transm Suppl 47, 278.

[799] Gschanes A, Windisch M (1998) The influence of Cere-brolysin and E021 on spatial navigation of 24-month-oldrats. J Neural Transm Suppl 53, 313-321.

[800] Hutter-Paier B, Fruhwirth M, Grygar E, Windisch M(1996) Cerebrolysin protects neurons from ischemia-induced loss of microtubule–associated protein 2. J NeuralTransm Suppl 47, 276.

[801] Rockenstein E, Mallory M, Mante M, Alford M, WindischM, Moessler H, Masliah E (2002) Effects of Cerebrolysinon amyloid-beta deposition in a transgenic model ofAlzheimer’s disease. J Neural Transm Suppl 62, 327-336.

[802] Rockenstein E, Adame A, Mante M, Moessler H, WindischM, Masliah E (2003) The neuroprotective effects of Cere-brolysin in a transgenic model of Alzheimer’s disease areassociated with improved behavioral performance. J Neu-ral Transm 110, 1313-1327.

[803] Rockenstein E, Adame A, Mante M, Larrea G, Crews L,Windisch M, Moessler H, Masliah E (2005) Ameliora-

Page 82: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

82 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

tion of the cerebrovascular amyloidosis in a transgenicmodel of Alzheimer’s disease with the neurotrophiccompound cerebrolysin. J Neural Transm 112, 269-282.

[804] Rockenstein E, Torrance M, Mante M, Adame A, PaulinoA, Rose JB, Crews L, Moessler H, Masliah E (2006)Cerebrolysin decreases amyloid-beta production by regu-lating amyloid protein precursor maturation in a transgenicmodel of Alzheimer’s disease. J Neurosci Res 83, 1252-1261.

[805] Rockenstein E, Mante M, Adame A, Crews L, Moessler H,Masliah E (2007) Effects of Cerebrolysin on neurogenesisin an APP transgenic model of Alzheimer’s disease. ActaNeuropathol 113, 265-275.

[806] Rockenstein E, Ubhi K, Pham E, Michael S, Doppler E,Novak P, Inglis C, Mante M, Adame A, Alvarez XA,Moessler H, Masliah E (2011) Beneficial effects of aneurotrophic peptidergic mixture persist for a prolongedperiod following treatment interruption in a transgenicmodel of Alzheimer’s disease. J Neurosci Res 89, 1812-1821.

[807] Rockenstein E, Ubhi K, Doppler E, Novak P, Moessler H,Li B, Blanchard J, Grundke-Iqbal I, Iqbal K, Mante M,Adame A, Crews L, Masliah E (2011) Regional compar-ison of the neurogenic effects of CNTF-derived peptidesand cerebrolysin in AbetaPP transgenic mice. J AlzheimersDis 27, 743-752.

[808] Masliah E, Diez-Tejedor E (2012) The pharmacologyof neurotrophic treatment with Cerebrolysin: Brain pro-tection and repair to counteract pathologies of acuteand chronic neurological disorders. Drugs Today (Barc)48(Suppl A), 3-24.

[809] Panisset M, Gauthier S, Moessler H, Windisch M (2002)Cerebrolysin in Alzheimer’s disease: A randomized,double-blind, placebo-controlled trial with a neurotrophicagent. J Neural Transm 109, 1089-1104.

[810] Molloy DW, Standish TI (2000) Clinical experience withCerebrolysin. J Neural Transm Suppl 59, 293-300.

[811] Ruether E, Alvarez XA, Rainer M, Moessler H (2002)Sustained improvement of cognition and global functionin patients with moderately severe Alzheimer’s disease:A double-blind, placebo-controlled study with the neu-rotrophic agent Cerebrolysin. J Neural Transm Suppl 62,265-275.

[812] Ladurner G, Kalvach P, Moessler H (2005) Neuropro-tective treatment with cerebrolysin in patients with acutestroke: A randomised controlled trial. J Neural Transm112, 415-428.

[813] Alvarez XA, Cacabelos R, Laredo M, Couceiro V, Sampe-dro C, Varela M, Corzo L, Fernandez-Novoa L, VargasM, Aleixandre M, Linares C, Granizo E, Muresanu D,Moessler H (2006) A 24-week, double-blind, placebo-controlled study of three dosages of Cerebrolysin inpatients with mild to moderate Alzheimer’s disease. Eur JNeurol 13, 43-54.

[814] Alvarez XA, Sampedro C, Figueroa J, Tellado I, Gon-zalez A, Garcia-Fantini M, Cacabelos R, Muresanu D,Moessler H (2008) Reductions in qEEG slowing over 1year and after treatment with Cerebrolysin in patients withmoderate-severe traumatic brain injury. J Neural Transm115, 683-692.

[815] Alvarez XA, Sampedro C, Cacabelos R, Linares C,Aleixandre M, Garcia-Fantini M, Moessler H (2009)Reduced TNF-alpha and increased IGF-I levels in theserum of Alzheimer’s disease patients treated with the neu-

rotrophic agent cerebrolysin. Int J Neuropsychopharmacol12, 867-872.

[816] Alvarez XA, Cacabelos R, Sampedro C, Aleixandre M,Linares C, Granizo E, Doppler E, Moessler H (2011)Efficacy and safety of Cerebrolysin in moderate to moder-ately severe Alzheimer’s disease: Results of a randomized,double-blind, controlled trial investigating three dosagesof Cerebrolysin. Eur J Neurol 18, 59-68.

[817] Muresanu DF, Alvarez XA, Moessler H, Buia M, Stan A,Pintea D, Moldovan F, Popescu BO (2008) A pilot study toevaluate the effects of Cerebrolysin on cognition and qEEGin vascular dementia: Cognitive improvement correlateswith qEEG acceleration. J Neurol Sci 267, 112-119.

[818] Muresanu DF, Alvarez XA, Moessler H, Novak PH, StanA, Buzoianu A, Bajenaru O, Popescu BO (2010) Per-sistence of the effects of Cerebrolysin on cognition andqEEG slowing in vascular dementia patients: Results of a3-month extension study. J Neurol Sci 299, 179-183.

[819] Plosker GL, Gauthier S (2009) Cerebrolysin: A review ofits use in dementia. Drugs Aging 26, 893-915.

[820] Plosker GL, Gauthier S (2010) Spotlight on cerebrolysinin dementia. CNS Drugs 24, 263-266.

[821] Bajenaru O, Tiu C, Moessler H, Antochi F, Muresanu D,Popescu BO, Novak P (2010) Efficacy and safety of Cere-brolysin in patients with hemorrhagic stroke. J Med Life3, 137-143.

[822] Guekht AB, Moessler H, Novak PH, Gusev EI (2011)Cerebrolysin in vascular dementia: Improvement ofclinical outcome in a randomized, double-blind, placebo-controlled multicenter trial. J Stroke Cerebrovasc Dis 20,310-318.

[823] Allegri RF, Guekht A (2012) Cerebrolysin improves symp-toms and delays progression in patients with Alzheimer’sdisease and vascular dementia. Drugs Today (Barc) 48Suppl A, 25-41.

[824] Alvarez XA, Cacabelos R, Sampedro C, Couceiro V,Aleixandre M, Vargas M, Linares C, Granizo E, Garcia-Fantini M, Baurecht W, Doppler E, Moessler H (2011)Combination treatment in Alzheimer’s disease: Resultsof a randomized, controlled trial with cerebrolysin anddonepezil. Curr Alzheimer Res 8, 583-591.

[825] Anton Alvarez X, Fuentes P (2011) Cerebrolysin inAlzheimer’s disease. Drugs Today (Barc) 47, 487-513.

[826] Thome J, Doppler E (2012) Safety profile of Cerebrolysin:Clinical experience from dementia and stroke trials. DrugsToday (Barc)48 Suppl A, 63-69.

[827] Leker RR, Teichner A, Grigoriadis N, Ovadia H, Bren-neman DE, Fridkin M, Giladi E, Romano J, Gozes I(2002) NAP, a femtomolar-acting peptide, protects thebrain against ischemic injury by reducing apoptotic death.Stroke 33, 1085-1092.

[828] Ashur-Fabian O, Segal-Ruder Y, Skutelsky E, BrennemanDE, Steingart RA, Giladi E, Gozes I (2003) The neuro-protective peptide NAP inhibits the aggregation of thebeta-amyloid peptide. Peptides 24, 1413-1423.

[829] Alcalay RN, Giladi E, Pick CG, Gozes I (2004)Intranasal administration of NAP, a neuroprotective pep-tide, decreases anxiety-like behavior in aging mice in theelevated plus maze. Neurosci Lett 361, 128-131.

[830] Gozes I, Zaltzman R, Hauser J, Brenneman DE, ShohamiE, Hill JM (2005) The expression of activity-dependentneuroprotective protein (ADNP) is regulated by brain dam-age and treatment of mice with the ADNP derived peptide,NAP, reduces the severity of traumatic head injury. CurrAlzheimer Res 2, 149-153.

Page 83: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 83

[831] Matsuoka Y, Gray AJ, Hirata-Fukae C, Minami SS, Water-house EG, Mattson MP, LaFerla FM, Gozes I, Aisen PS(2007) Intranasal NAP administration reduces accumula-tion of amyloid peptide and tau hyperphosphorylation ina transgenic mouse model of Alzheimer’s disease at earlypathological stage. J Mol Neurosci 31, 165-170.

[832] Matsuoka Y, Jouroukhin Y, Gray AJ, Ma L, Hirata-FukaeC, Li HF, Feng L, Lecanu L, Walker BR, Planel E, ArancioO, Gozes I, Aisen PS (2008) A neuronal microtubule-interacting agent, NAPVSIPQ, reduces tau pathologyand enhances cognitive function in a mouse model ofAlzheimer’s disease. J Pharmacol Exp Ther 325, 146-153.

[833] Shiryaev N, Jouroukhin Y, Giladi E, Polyzoidou E, Grigo-riadis NC, Rosenmann H, Gozes I (2009) NAP protectsmemory, increases soluble tau and reduces tau hyper-phosphorylation in a tauopathy model. Neurobiol Dis 34,381-388.

[834] Gozes I, Divinski I (2004) The femtomolar-acting NAPinteracts with microtubules: Novel aspects of astrocyteprotection. J Alzheimers Dis 6, S37-S41.

[835] Gozes I, Divinski I (2007) NAP, a neuroprotective drugcandidate in clinical trials, stimulates microtubule assem-bly in the living cell. Curr Alzheimer Res 4, 507-509.

[836] Merenlender-Wagner A, Pikman R, Giladi E, AndrieuxA, Gozes I (2010) NAP (davunetide) enhances cognitivebehavior in the STOP heterozygous mouse–a microtubule-deficient model of schizophrenia. Peptides 31, 1368-1373.

[837] Fleming SM, Mulligan CK, Richter F, Mortazavi F,Lemesre V, Frias C, Zhu C, Stewart A, Gozes I, Mori-moto B, Chesselet MF (2011) A pilot trial of themicrotubule-interacting peptide (NAP) in mice overex-pressing alpha-synuclein shows improvement in motorfunction and reduction of alpha-synuclein inclusions. MolCell Neurosci 46, 597-606.

[838] Gozes I (2007) Activity-dependent neuroprotective pro-tein: From gene to drug candidate. Pharmacol Ther 114,146-154.

[839] Gozes I (2011) NAP (davunetide) provides functional andstructural neuroprotection. Curr Pharm Des 17, 1040-1044.

[840] Gozes I (2011) Microtubules, schizophrenia and cognitivebehavior: Preclinical development of davunetide (NAP) asa peptide-drug candidate. Peptides 32, 428-431.

[841] Gozes I (2011) Microtubules (tau) as an emerging ther-apeutic target: NAP (davunetide). Curr Pharm Des 17,3413-3417.

[842] Gozes I, Spivak-Pohis I (2006) Neurotrophic effects ofthe peptide NAP: A novel neuroprotective drug candidate.Curr Alzheimer Res 3, 197-199.

[843] Gozes I, Steingart RA, Spier AD (2004) NAP mechanismsof neuroprotection. J Mol Neurosci 24, 67-72.

[844] Gozes I, Morimoto BH, Tiong J, Fox A, Sutherland K,Dangoor D, Holser-Cochav M, Vered K, Newton P, AisenPS, Matsuoka Y, van Dyck CH, Thal L (2005) NAP:Research and development of a peptide derived fromactivity-dependent neuroprotective protein (ADNP). CNSDrug Rev 11, 353-368.

[845] Gozes I, Stewart A, Morimoto B, Fox A, SutherlandK, Schmeche D (2009) Addressing Alzheimer’s diseasetangles: From NAP to AL-108. Curr Alzheimer Res 6,455-460.

[846] Geerts H (2008) AL-108 and AL-208, formulations of theneuroprotective NAP fragment of activity-dependent neu-roprotective protein, for cognitive disorders. Curr OpinInvestig Drugs 9, 800-811.

[847] Shiryaev N, Pikman R, Giladi E, Gozes I (2011) Protectionagainst tauopathy by the drug candidates NAP (davune-tide) and D-SAL: Biochemical, cellular and behavioralaspects. Curr Pharm Des 17, 2603-2612.

[848] Javitt DC, Buchanan RW, Keefe RS, Kern R, McMahonRP, Green MF, Lieberman J, Goff DC, Csernansky JG,McEvoy JP, Jarskog F, Seidman LJ, Gold JM, KimhyD, Nolan KS, Barch DS, Ball MP, Robinson J, MarderSR (2012) Effect of the neuroprotective peptide davune-tide (AL-108) on cognition and functional capacity inschizophrenia. Schizophr Res 136, 25-31.

[849] Borsello T, Clarke PG, Hirt L, Vercelli A, Repici M,Schorderet DF, Bogousslavsky J, Bonny C (2003) A pep-tide inhibitor of c-Jun N-terminal kinase protects againstexcitotoxicity and cerebral ischemia. Nat Med 9, 1180-1186.

[850] Hirt L, Badaut J, Thevenet J, Granziera C, Regli L, Mau-rer F, Bonny C, Bogousslavsky J (2004) D-JNKI1, acell-penetrating c-Jun-N-terminal kinase inhibitor, pro-tects against cell death in severe cerebral ischemia. Stroke35, 1738-1743.

[851] Suckfuell M, Canis M, Strieth S, Scherer H, Haisch A(2007) Intratympanic treatment of acute acoustic traumawith a cell-permeable JNK ligand: A prospective random-ized phase I/II study. Acta Otolaryngol 127, 938-942.

[852] Wiegler K, Bonny C, Coquoz D, Hirt L (2008) The JNKinhibitor XG-102 protects from ischemic damage withdelayed intravenous administration also in the presenceof recombinant tissue plasminogen activator. CerebrovascDis 26, 360-366.

[853] Tayal V, Kalra BS (2008) Cytokines and anti-cytokines astherapeutics–an update. Eur J Pharmacol 579, 1-12.

[854] Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP(2008) Tumor necrosis factor antagonist mechanisms ofaction: A comprehensive review. Pharmacol Ther 117,244-279.

[855] Tobinick EL (2007) Perispinal etanercept for the treatmentof Alzheimer’s disease. Curr Alzheimer Res 4, 550-552.

[856] Tobinick EL, Gross H (2008) Rapid improvement in ver-bal fluency and aphasia following perispinal etanercept inAlzheimer’s disease. BMC Neurol 8, 27.

[857] Tobinick EL, Gross H (2008) Rapid cognitive improve-ment in Alzheimer’s disease following perispinal etaner-cept administration. J Neuroinflammation 5, 2.

[858] Griffin WS (2008) Perispinal etanercept: Potential as anAlzheimer therapeutic. J Neuroinflammation 5, 3.

[859] Sara VR, Carlsson-Skwirut C, Bergman T, Jornvall H,Roberts PJ, Crawford M, Hakansson LN, Civalero I, Nord-berg A (1989) Identification of Gly-Pro-Glu (GPE), theaminoterminal tripeptide of insulin-like growth factor 1which is truncated in brain, as a novel neuroactive peptide.Biochem Biophys Res Commun 165, 766-771.

[860] Sara VR, Carlsson-Skwirut C, Drakenberg K, GiacobiniMB, Hakansson L, Mirmiran M, Nordberg A, Olson L,Reinecke M, Stahlbom PA (1993) The biological role oftruncated insulin-like growth factor-1 and the tripeptideGPE in the central nervous system. Ann N Y Acad Sci 692,183-191.

[861] Saura J, Curatolo L, Williams CE, Gatti S, Benatti L,Peeters C, Guan J, Dragunow M, Post C, Faull RL, Gluck-man PD, Skinner SJ (1999) Neuroprotective effects ofGly-Pro-Glu, the N-terminal tripeptide of IGF-1, in thehippocampus in vitro. Neuroreport 10, 161-164.

[862] Aguado-Llera D, Martin-Martinez M, Garcia-Lopez MT,rilla-Ferreiro E, Barrios V (2004) Gly-Pro-Glu protects

Page 84: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

84 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

beta-amyloid-induced somatostatin depletion in the ratcortex. Neuroreport 15, 1979-1982.

[863] Burgos-Ramos E, Martos-Moreno GA, Lopez MG,Herranz R, Aguado-Llera D, Egea J, Frechilla D, Cenar-ruzabeitia E, Leon R, Arilla-Ferreiro E, Argente J, BarriosV (2009) The N-terminal tripeptide of insulin-like growthfactor-I protects against beta-amyloid-induced somato-statin depletion by calcium and glycogen synthase kinase3 beta modulation. J Neurochem 109, 360-370.

[864] Bickerdike MJ, Thomas GB, Batchelor DC, SirimanneES, Leong W, Lin H, Sieg F, Wen J, Brimble MA, Har-ris PW, Gluckman PD (2009) NNZ-2566: A Gly-Pro-Gluanalogue with neuroprotective efficacy in a rat model ofacute focal stroke. J Neurol Sci 278, 85-90.

[865] Lu XC, Chen RW, Yao C, Wei H, Yang X, Liao Z, DaveJR, Tortella FC (2009) NNZ-2566, a glypromate analog,improves functional recovery and attenuates apoptosis andinflammation in a rat model of penetrating ballistic-typebrain injury. J Neurotrauma 26, 141-154.

[866] Lu XC, Si Y, Williams AJ, Hartings JA, Gryder D, TortellaFC (2009) NNZ-2566, a glypromate analog, attenuatesbrain ischemia-induced non-convulsive seizures in rats.J Cereb Blood Flow Metab 29, 1924-1932.

[867] Wei HH, Lu XC, Shear DA, Waghray A, Yao C, TortellaFC, Dave JR (2009) NNZ-2566 treatment inhibits neu-roinflammation and pro-inflammatory cytokine expressioninduced by experimental penetrating ballistic-like braininjury in rats. J Neuroinflammation 6, 19.

[868] Alonso De Diego SA, Munoz P, Gonzalez-Muniz R, Her-ranz R, Martin-Martinez M, Cenarruzabeitia E, FrechillaD, Del RJ, Jimeno ML, Garcia-Lopez MT (2005) Ana-logues of the neuroprotective tripeptide Gly-Pro-Glu(GPE): Synthesis and structure-activity relationships.Bioorg Med Chem Lett 15, 2279-2283.

[869] Alonso De Diego SA, Gutierrez-Rodriguez M, Perez dV, Casabona D, Cativiela C, Gonzalez-Muniz R, Her-ranz R, Cenarruzabeitia E, Frechilla D, Del RJ, JimenoML, Garcia-Lopez MT (2006) New Gly-Pro-Glu (GPE)analogues: Expedite solid-phase synthesis and biologicalactivity. Bioorg Med Chem Lett 16, 1392-1396.

[870] Alonso De Diego SA, Gutierrez-Rodriguez M, Perez dV, Gonzalez-Muniz R, Herranz R, Martin-Martinez M,Cenarruzabeitia E, Frechilla D, Del RJ, Jimeno ML,Garcia-Lopez MT (2006) The neuroprotective activity ofGPE tripeptide analogues does not correlate with gluta-mate receptor binding affinity. Bioorg Med Chem Lett 16,3396-3400.

[871] Lai MY, Brimble MA, Callis DJ, Harris PW, Levi MS,Sieg F (2005) Synthesis and pharmacological evaluationof glycine-modified analogues of the neuroprotective agentglycyl-L-prolyl-L-glutamic acid (GPE). Bioorg Med Chem13, 533-548.

[872] Holtje M, Djalali S, Hofmann F, Munster-Wandowski A,Hendrix S, Boato F, Dreger SC, Grosse G, HennebergerC, Grantyn R, Just I, Ahnert-Hilger G (2009) A 29-amino acid fragment of Clostridium botulinum C3 proteinenhances neuronal outgrowth, connectivity, and reinner-vation. FASEB J 23, 1115-1126.

[873] Boato F, Hendrix S, Huelsenbeck SC, Hofmann F, GrosseG, Djalali S, Klimaschewski L, Auer M, Just I, hnert-Hilger G, Holtje M (2010) C3 peptide enhances recoveryfrom spinal cord injury by improved regenerative growthof descending fiber tracts. J Cell Sci 123, 1652-1662.

[874] Just I, Rohrbeck A, Huelsenbeck SC, Hoeltje M (2011)Therapeutic effects of Clostridium botulinum C3 exoen-

zyme. Naunyn Schmiedebergs Arch Pharmacol 383,247-252.

[875] Loske P, Boato F, Hendrix S, Piepgras J, Just I, hnert-Hilger G, Holtje M (2012) Minimal essential length ofClostridium botulinum C3 peptides to enhance neuronalregenerative growth and connectivity in a non-enzymaticmode. J Neurochem 120, 1084-1096.

[876] Jiang Y, Brody DL (2012) Administration of COG1410reduces axonal amyloid precursor protein immunoreac-tivity and microglial activation after controlled corticalimpact in mice. J Neurotrauma 29, 2332-2341.

[877] Ghosal K, Stathopoulos A, Thomas D, Phenis D, VitekMP, Pimplikar SW (2012) The apolipoprotein-E-mimeticCOG112 protects amyloid precursor protein intracel-lular domain-overexpressing animals from Alzheimer’sdisease-like pathological features. Neurodegener Dis,DOI: 10.1159-000341299 [Epub ahead of print].

[878] 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.

[879] Hofmann SG, Smits JA, Asnaani A, Gutner CA, Otto MW(2011) Cognitive enhancers for anxiety disorders. Phar-macol Biochem Behav 99, 275-284.

[880] Marwarha G, Dasari B, Prasanthi JR, Schommer J, GhribiO (2010) Leptin reduces the accumulation of Abeta andphosphorylated tau induced by 27-hydroxycholesterol inrabbit organotypic slices. J Alzheimers Dis 19, 1007-1019.

[881] Marwarha G, Dasari B, Prabhakara JP, Schommer J, GhribiO (2010) beta-Amyloid regulates leptin expression and tauphosphorylation through the mTORC1 signaling pathway.J Neurochem 115, 373-384.

[882] Marwarha G, Prasanthi JR, Schommer J, Dasari B, GhribiO (2011) Molecular interplay between leptin, insulin-likegrowth factor-1, and beta-amyloid in organotypic slicesfrom rabbit hippocampus. Mol Neurodegener 6, 41.

[883] Harvey J, Shanley LJ, O’Malley D, Irving AJ (2005) Lep-tin: A potential cognitive enhancer? Biochem Soc Trans33, 1029-1032.

[884] Farr SA, Banks WA, Morley JE (2006) Effects of leptin onmemory processing. Peptides 27, 1420-1425.

[885] Beccano-Kelly D, Harvey J (2012) Leptin: A novel thera-peutic target in Alzheimer’s disease? Int J Alzheimers Dis2012, 594137.

[886] Perez-Gonzalez R, Antequera D, Vargas T, Spuch C, BolosM, Carro E (2011) Leptin induces proliferation of neu-ronal progenitors and neuroprotection in a mouse model ofAlzheimer’s disease. J Alzheimers Dis 24(Suppl 2), 17-25.

[887] Lourenco FC, Galvan V, Fombonne J, Corset V, LlambiF, Muller U, Bredesen DE, Mehlen P (2009) Netrin-1interacts with amyloid precursor protein and regulatesamyloid-beta production. Cell Death Differ 16, 655-663.

[888] Rama N, Goldschneider D, Corset V, Lambert J, PaysL, Mehlen P (2012) Amyloid precursor protein regulatesnetrin-1-mediated commissural axon outgrowth. J BiolChem 287, 30014-30023.

[889] Gorba T, Bradoo P, Antonic A, Marvin K, Liu DX, LobiePE, Reymann KG, Gluckman PD, Sieg F (2006) Neu-ral regeneration protein is a novel chemoattractive and

Page 85: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 85

neuronal survival-promoting factor. Exp Cell Res 312,3060-3074.

[890] Xue D, Zhao M, Wang YJ, Wang L, Yang Y, WangSW, Zhang R, Zhao Y, Liu RT (2012) A multifunctionalpeptide rescues memory deficits in Alzheimer’s diseasetransgenic mice by inhibiting Abeta42-induced cytotoxic-ity and increasing microglial phagocytosis. Neurobiol Dis46, 701-709.

[891] Schuster D, Rajendran A, Hui SW, Nicotera T, SrikrishnanT, Kruzel ML (2005) Protective effect of colostrinin onneuroblastoma cell survival is due to reduced aggregationof beta-amyloid. Neuropeptides 39, 419-426.

[892] Bourhim M, Kruzel M, Srikrishnan T, Nicotera T (2007)Linear quantitation of Abeta aggregation using ThioflavinT: Reduction in fibril formation by colostrinin. J NeurosciMethods 160, 264-268.

[893] Froud KE, Wardhaugh T, Banks D, Saffrey MJ, StewartMG (2010) Colostrinin alleviates amyloid-beta inducedtoxicity in rat primary hippocampal cultures. J AlzheimersDis 20, 423-426.

[894] Popik P, Bobula B, Janusz M, Lisowski J, Vetulani J (1999)Colostrinin, a polypeptide isolated from early milk, facil-itates learning and memory in rats. Pharmacol BiochemBehav 64, 183-189.

[895] Stewart MG, Banks D (2006) Enhancement of long-termmemory retention by Colostrinin in one-day-old chickstrained on a weak passive avoidance learning paradigm.Neurobiol Learn Mem 86, 66-71.

[896] Szaniszlo P, German P, Hajas G, Saenz DN, WoodberryMW, Kruzel ML, Boldogh I (2009) Effects of Colostrininon gene expression-transcriptomal network analysis. IntImmunopharmacol 9, 181-193.

[897] Leszek J, Inglot AD, Janusz M, Byczkiewicz F, Kiejna A,Georgiades J, Lisowski J (2002) Colostrinin proline-richpolypeptide complex from ovine colostrum–a long-termstudy of its efficacy in Alzheimer’s disease. Med Sci Monit8, I93-I96.

[898] Bilikiewicz A, Gaus W (2004) Colostrinin (a naturallyoccurring, proline-rich, polypeptide mixture) in the treat-ment of Alzheimer’s disease. J Alzheimers Dis 6, 17-26.

[899] Boldogh I, Kruzel ML (2008) Colostrinintrade mark: Anoxidative stress modulator for prevention and treatment ofage-related disorders. J Alzheimers Dis 13, 303-321.

[900] Szaniszlo P, German P, Hajas G, Saenz DN, Kruzel M,Boldogh I (2009) New insights into clinical trial forColostrinin in Alzheimer’s disease. J Nutr Health Aging13, 235-241.

[901] Janusz M, Zablocka A (2010) Colostral proline-richpolypeptides–immunoregulatory properties and prospectsof therapeutic use in Alzheimer’s disease. Curr AlzheimerRes 7, 323-333.

[902] Stewart MG (2008) Colostrinin: A naturally occurringcompound derived from mammalian colostrum with effi-cacy in treatment of neurodegenerative diseases, includingAlzheimer’s. Expert Opin Pharmacother 9, 2553-2559.

[903] Zimecki M (2008) A proline-rich polypeptide from ovinecolostrum: Colostrinin with immunomodulatory activity.Adv Exp Med Biol 606, 241-250.

[904] Taddei K, Laws SM, Verdile G, Munns S, D’Costa K, Har-vey AR, Martins IJ, Hill F, Levy E, Shaw JE, MartinsRN (2010) Novel phage peptides attenuate beta amyloid-42 catalysed hydrogen peroxide production and associatedneurotoxicity. Neurobiol Aging 31, 203-214.

[905] Hock FJ, Gerhards HJ, Wiemer G, Usinger P, Geiger R(1988) Learning and memory processes of an ACTH4-9

analog (ebiratide; Hoe 427) in mice and rats. Peptides 9,575-581.

[906] Wiemer G, Gerhards HJ, Hock FJ, Usinger P, Von RW,Geiger R (1988) Neurochemical effects of the syntheticACTH4-9-analog Hoe 427 (Ebiratide) in rat brain. Pep-tides 9, 1081-1087.

[907] Siegfried KR (1991) First clinical impressions with anACTH analog (HOE 427) in the treatment of Alzheimer’sdisease. Ann N Y Acad Sci 640, 280-283.

[908] Manso Y, Adlard PA, Carrasco J, Vasak M, Hidalgo J(2011) Metallothionein and brain inflammation. J BiolInorg Chem 16, 1103-1113.

[909] Manso Y, Carrasco J, Comes G, Meloni G, Adlard PA,Bush AI, Vasak M, Hidalgo J (2012) Characterizationof the role of metallothionein-3 in an animal model ofAlzheimer’s disease. Cell Mol Life Sci 69, 3683-3700.

[910] Pelsman A, Hoyo-Vadillo C, Gudasheva TA, SeredeninSB, Ostrovskaya RU, Busciglio J (2003) GVS-111 pre-vents oxidative damage and apoptosis in normal andDown’s syndrome human cortical neurons. Int J Dev Neu-rosci 21, 117-124.

[911] Ostrovskaya RU, Romanova GA, Barskov IV, ShaninaEV, Gudasheva TA, Victorov IV, Voronina TA, Sere-denin SB (1999) Memory restoring and neuroprotectiveeffects of the proline-containing dipeptide, GVS-111, in aphotochemical stroke model. Behav Pharmacol 10, 549-553.

[912] Ostrovskaya RU, Mirsoev TK, Romanova GA, GudashevaTA, Kravchenko EV, Trofimov CC, Voronina TA, Sere-denin SB (2001) Proline-containing dipeptide GVS-111retains nootropic activity after oral administration. BullExp Biol Med 132, 959-962.

[913] Ostrovskaya RU, Gruden MA, Bobkova NA, Sewell RD,Gudasheva TA, Samokhin AN, Seredinin SB, Noppe W,Sherstnev VV, Morozova-Roche LA (2007) The nootropicand neuroprotective proline-containing dipeptide noopeptrestores spatial memory and increases immunoreactivity toamyloid in an Alzheimer’s disease model. J Psychophar-macol 21, 611-619.

[914] Ostrovskaya RU, Belnik AP, Storozheva ZI (2008)Noopept efficiency in experimental Alzheimer disease(cognitive deficiency caused by beta-amyloid25-35 injec-tion into Meynert basal nuclei of rats). Bull Exp Biol Med146, 77-80.

[915] Ostrovskaya RU, Gudasheva TA, Zaplina AP, Vahitova JV,Salimgareeva MH, Jamidanov RS, Seredenin SB (2008)Noopept stimulates the expression of NGF and BDNF inrat hippocampus. Bull Exp Biol Med 146, 334-337.

[916] Kovalenko LP, Shipaeva EV, Alekseeva SV, Pronin AV,Durnev AD, Gudasheva TA, Ostrovskaja RU, SeredeninSB (2007) Immunopharmacological properties of noopept.Bull Exp Biol Med 144, 49-52.

[917] Neznamov GG, Teleshova ES (2009) Comparative stud-ies of Noopept and piracetam in the treatment of patientswith mild cognitive disorders in organic brain diseases ofvascular and traumatic origin. Neurosci Behav Physiol 39,311-321.

[918] Kondratenko RV, Derevyagin VI, Skrebitsky VG (2010)Novel nootropic dipeptide Noopept increases inhibitorysynaptic transmission in CA1 pyramidal cells. NeurosciLett 476, 70-73.

[919] Vorobyov V, Kaptsov V, Kovalev G, Sengpiel F (2011)Effects of nootropics on the EEG in conscious rats andtheir modification by glutamatergic inhibitors. Brain ResBull 85, 123-132.

Page 86: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

86 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[920] Jia X, Gharibyan AL, Ohman A, Liu Y, OlofssonA, Morozova-Roche LA (2011) Neuroprotective andnootropic drug noopept rescues alpha-synuclein amyloidcytotoxicity. J Mol Biol 414, 699-712.

[921] Hirano M, Hirate K, Miyake N (1998) NC-1900, a[pGlu4,Cyt6]AVP(4-9) analogue, facilitates the learningperformance of rats on delayed nonmatching to sampletask in Y-water maze. Methods Find Exp Clin Pharmacol20, 567-573.

[922] Tanaka T, Yamada K, Senzaki K, Narimatsu H, NishimuraK, Kameyama T, Nabeshima T (1998) NC-1900, an activefragment analog of arginine vasopressin, improves learn-ing and memory deficits induced by beta-amyloid proteinin rats. Eur J Pharmacol 352, 135-142.

[923] Hori E, Uwano T, Tamura R, Miyake N, Nishijo H, Ono T(2002) Effects of a novel arginine-vasopressin derivative,NC-1900, on the spatial memory impairment of rats withtransient forebrain ischemia. Brain Res Cogn Brain Res13, 1-15.

[924] Ishida T, Sato T, Irifune M, Tanaka K, Hirate K, NakamuraN, Nishikawa T (2007) Inhibitory effect of cyclooxy-genase inhibitors on the step-through passive avoidanceperformance in mice treated with NC-1900, an arginine-vasopressin fragment analog. Methods Find Exp ClinPharmacol 29, 315-320.

[925] Sato T, Ishida T, Irifune M, Tanaka K, Hirate K, Naka-mura N, Nishikawa T (2007) Effect of NC-1900, an activefragment analog of arginine vasopressin, and inhibitors ofarachidonic acid metabolism on performance of a passiveavoidance task in mice. Eur J Pharmacol 560, 36-41.

[926] Fotinopoulou A, Tsachaki M, Vlavaki M, PoulopoulosA, Rostagno A, Frangione B, Ghiso J, EfthimiopoulosS (2005) BRI2 interacts with amyloid precursor protein(APP) and regulates amyloid beta (Abeta) production.J Biol Chem 280, 30768-30772.

[927] Peng S, Fitzen M, Jornvall H, Johansson J (2010) Theextracellular domain of Bri2 (ITM2B) binds the ABripeptide (1-23) and amyloid beta-peptide (Abeta1-40):Implications for Bri2 effects on processing of amyloid pre-cursor protein and Abeta aggregation. Biochem BiophysRes Commun 393, 356-361.

[928] Matsuda S, Giliberto L, Matsuda Y, Davies P, McGowan E,Pickford F, Ghiso J, Frangione B, D’Adamio L (2005) Thefamilial dementia BRI2 gene binds the Alzheimer geneamyloid-beta precursor protein and inhibits amyloid-betaproduction. J Biol Chem 280, 28912-28916.

[929] Matsuda S, Giliberto L, Matsuda Y, McGowan EM,D’Adamio L (2008) BRI2 inhibits amyloid beta-peptideprecursor protein processing by interfering with thedocking of secretases to the substrate. J Neurosci 28, 8668-8676.

[930] Matsuda S, Matsuda Y, Snapp EL, D’Adamio L(2011) Maturation of BRI2 generates a specific inhibitorthat reduces APP processing at the plasma membraneand in endocytic vesicles. Neurobiol Aging 32, 1400-1408.

[931] Kim J, Miller VM, Levites Y, West KJ, Zwizinski CW,Moore BD, Troendle FJ, Bann M, Verbeeck C, Price RW,Smithson L, Sonoda L, Wagg K, Rangachari V, Zou F,Younkin SG, Graff-Radford N, Dickson D, Rosenberry T,Golde TE (2008) BRI2 (ITM2b) inhibits Abeta depositionin vivo. J Neurosci 28, 6030-6036.

[932] Tsachaki M, Ghiso J, Efthimiopoulos S (2008) BRI2 as acentral protein involved in neurodegeneration. BiotechnolJ 3, 1548-1554.

[933] Tsachaki M, Serlidaki D, Fetani A, Zarkou V, Rozani I,Ghiso J, Efthimiopoulos S (2011) Glycosylation of BRI2on asparagine 170 is involved in its trafficking to the cellsurface but not in its processing by furin or ADAM10.Glycobiology 21, 1382-1388.

[934] Findeis MA (2007) The role of amyloid beta peptide 42 inAlzheimer’s disease. Pharmacol Ther 116, 266-286.

[935] Gunstad J, Spitznagel MB, Glickman E, Alexander T,Juvancic-Heltzel J, Walter K, Murray L (2008) beta-Amyloid is associated with reduced cognitive function inhealthy older adults. J Neuropsychiatry Clin Neurosci 20,327-330.

[936] Cosentino SA, Stern Y, Sokolov E, Scarmeas N, ManlyJJ, Tang MX, Schupf N, Mayeux RP (2010) Plasmabeta-amyloid and cognitive decline. Arch Neurol 67, 1485-1490.

[937] Parihar MS, Brewer GJ (2010) Amyloid-beta as a modu-lator of synaptic plasticity. J Alzheimers Dis 22, 741-763.

[938] Koyama A, Okereke OI, Yang T, Blacker D, Selkoe DJ,Grodstein F (2012) Plasma amyloid-beta as a predictor ofdementia and cognitive decline: A systematic review andmeta-analysis. Arch Neurol 69, 824-831.

[939] Koivunen J, Karrasch M, Scheinin NM, Aalto S, VahlbergT, Nagren K, Helin S, Viitanen M, Rinne JO (2012) Cog-nitive decline and amyloid accumulation in patients withmild cognitive impairment. Dement Geriatr Cogn Disord34, 31-37.

[940] Chetelat G, Villemagne VL, Bourgeat P, Pike KE, JonesG, Ames D, Ellis KA, Szoeke C, Martins RN, O’KeefeGJ, Salvado O, Masters CL, Rowe CC (2010) Rela-tionship between atrophy and beta-amyloid deposition inAlzheimer disease. Ann Neurol 67, 317-324.

[941] Chetelat G, Villemagne VL, Villain N, Jones G, EllisKA, Ames D, Martins RN, Masters CL, Rowe CC (2012)Accelerated cortical atrophy in cognitively normal elderlywith high beta-amyloid deposition. Neurology 78, 477-484.

[942] Chetelat G, Villemagne VL, Pike KE, Ellis KA, AmesD, Masters CL, Rowe CC (2012) Relationship betweenmemory performance and beta-amyloid deposition at dif-ferent stages of Alzheimer’s disease. Neurodegener Dis10, 141-144.

[943] Klyubin I, Cullen WK, Hu NW, Rowan MJ (2012)Alzheimer’s disease Abeta assemblies mediating rapid dis-ruption of synaptic plasticity and memory. Mol Brain 5,25.

[944] Cavallucci V, D’Amelio M, Cecconi F (2012) Abeta toxi-city in Alzheimer’s disease. Mol Neurobiol 45, 366-378.

[945] Mucke L, Selkoe DJ (2012) Neurotoxicity of amyloid beta-protein: Synaptic and network dysfunction. Cold SpringHarb Perspect Med 2, a006338.

[946] Reitz C (2012) Alzheimer’s disease and the amyloid cas-cade hypothesis: A critical review. Int J Alzheimers Dis2012, 369808.

[947] Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H,Cairns NJ, Castellani RJ, Crain BJ, Davies P, Del TK, Duy-ckaerts C, Frosch MP, Haroutunian V, Hof PR, HuletteCM, Hyman BT, Iwatsubo T, Jellinger KA, Jicha GA,Kovari E, Kukull WA, Leverenz JB, Love S, MackenzieIR, Mann DM, Masliah E, McKee AC, Montine TJ, Mor-ris JC, Schneider JA, Sonnen JA, Thal DR, TrojanowskiJQ, Troncoso JC, Wisniewski T, Woltjer RL, Beach TG(2012) Correlation of Alzheimer disease neuropathologicchanges with cognitive status: A review of the literature. JNeuropathol Exp Neurol 71, 362-381.

Page 87: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 87

[948] Kline A (2012) Apolipoprotein E, amyloid-beta clear-ance and therapeutic opportunities in Alzheimer’s disease.Alzheimers Res Ther 4, 32.

[949] Soto C, Kindy MS, Baumann M, Frangione B (1996) Inhi-bition of Alzheimer’s amyloidosis by peptides that preventbeta-sheet conformation. Biochem Biophys Res Commun226, 672-680.

[950] Soto C, Sigurdsson EM, Morelli L, Kumar RA, Cas-tano EM, Frangione B (1998) Beta-sheet breaker peptidesinhibit fibrillogenesis in a rat brain model of amyloido-sis: Implications for Alzheimer’s therapy. Nat Med 4, 822-826.

[951] Zagorski MG (1998) Amyloid aggregation inhibitors.IDrugs 1, 17-18.

[952] Findeis MA (1999) �-Amyloid aggregation inhibitors.Current Opin CNS Invest Druigs 1, 333-339.

[953] Findeis MA, Musso GM, rico-Muendel CC, BenjaminHW, Hundal AM, Lee JJ, Chin J, Kelley M, WakefieldJ, Hayward NJ, Molineaux SM (1999) Modified-peptideinhibitors of amyloid beta-peptide polymerization. Bio-chemistry 38, 6791-6800.

[954] Soto C (1999) Plaque busters: Strategies to inhibit amyloidformation in Alzheimer’s disease. Mol Med Today 5, 343-350.

[955] Soto C (1999) b-Amyloid disrupting drugs. CNS Drugs12, 347-356.

[956] Moore CI, Wolfe MS (1999) Inhibition of �-amyloid for-mation as a therapeutic strategy. Exp Opin Ther Patents 9,135-146.

[957] Findeis MA, Lee JJ, Kelley M, Wakefield JD, Zhang MH,Chin J, Kubasek W, Molineaux SM (2001) Characteriza-tion of cholyl-leu-val-phe-phe-ala-OH as an inhibitor ofamyloid beta-peptide polymerization. Amyloid 8, 231-241.

[958] Adessi C, Soto C (2002) Beta-sheet breaker strategy forthe treatment of Alzheimer’s disease. Drug Dev Res 56,184-193.

[959] Findeis MA (2002) Peptide inhibitors of beta amyloidaggregation. Curr Top Med Chem 2, 417-423.

[960] Levine H (2002) The challenge of inhibiting Abeta poly-merization. Curr Med Chem 9, 1121-1133.

[961] Levine H III (2005) Multiple ligand binding sites on Abeta(1-40) fibrils. Amyloid 12, 5-14.

[962] Soto C, Estrada L (2005) Amyloid inhibitors and beta-sheet breakers. Subcell Biochem 38, 351-364.

[963] Lansbury PT, Lashuel HA (2006) A century-old debateon protein aggregation and neurodegeneration enters theclinic. Nature 443, 774-779.

[964] Levine H III (2007) Small molecule inhibitors of Abetaassembly. Amyloid 14, 185-197.

[965] Stains CI, Mondal K, Ghosh I (2007) Molecules that targetbeta-amyloid. ChemMedChem 2, 1674-1692.

[966] Hawkes CA, Ng V, McLaurin J (2009) Small moleculeinhibitors of A�-aggregation and neurotoxicity. Drug DevRes 70, 111-124.

[967] Haydar SN, Yun H, Staal RGW, Hirst WD (2009)Small-molecule protein-protein interaction inhibitors astherapeutic agents for neurodegenerative diseases: Recentprogress and future directions. Ann Rep Med Chem 44,51-69.

[968] Yadav A, Sonker M (2009) Perspectives in designing antiaggregation agents as Alzheimer disease drugs. Eur J MedChem 44, 3866-3873.

[969] Kim HY, Kim Y, Han G, Kim DJ (2010) Regulation of invitro Abeta1-40 aggregation mediated by small molecules.J Alzheimers Dis 22, 73-85.

[970] Butterfield SM, Lashuel HA (2010) Amyloidogenicprotein-membrane interactions: Mechanistic insight frommodel systems. Angew Chem Int Ed Engl 49, 5628-5654.

[971] Scherzer-Attali R, Pellarin R, Convertino M, Frydman-Marom A, Egoz-Matia N, Peled S, Levy-Sakin M, ShalevDE, Caflisch A, Gazit E, Segal D (2010) Complete phe-notypic recovery of an Alzheimer’s disease model by aquinone-tryptophan hybrid aggregation inhibitor. PLoSOne 5, e11101.

[972] Dorgeret B, Khemtemourian L, Correia I, Soulier JL,Lequin O, Ongeri S (2011) Sugar-based peptidomimet-ics inhibit amyloid beta-peptide aggregation. Eur J MedChem 46, 5959-5969.

[973] Richman M, Wilk S, Skirtenko N, Perelman A,Rahimipour S (2011) Surface-modified protein micro-spheres capture amyloid-beta and inhibit its aggregationand toxicity. Chemistry 17, 11171-11177.

[974] Butterfield S, Hejjaoui M, Fauvet B, Awad L, Lashuel HA(2012) Chemical strategies for controlling protein fold-ing and elucidating the molecular mechanisms of amyloidformation and toxicity. J Mol Biol 421, 204-236.

[975] Scherzer-Attali R, Shaltiel-Karyo R, Adalist YH, SegalD, Gazit E (2012) Generic inhibition of amyloido-genic proteins by two naphthoquinone-tryptophan hybridmolecules. Proteins 80, 1962-1973.

[976] Scherzer-Attali R, Farfara D, Cooper I, Levin A, Ben-Romano T, Trudler D, Vientrov M, Shaltiel-Karyo R,Shalev DE, Segev-Amzaleg N, Gazit E, Segal D, FrenkelD (2012) Naphthoquinone-tyrptophan reduces neurotoxicAbeta*56 levels and improves cognition in Alzheimer’sdisease animal model. Neurobiol Dis 46, 663-672.

[977] Ratner M (2009) Spotlight focuses on protein-misfoldingtherapies. Nat Biotechnol 27, 874.

[978] Plante-Bordeneuve V, Said G (2011) Familial amyloidpolyneuropathy. Lancet Neurol 10, 1086-1097.

[979] Yamamoto T, Muto K, Komiyama M, Canivet J, Yam-aguchi J, Itami K (2011) Nickel-catalyzed C-H arylationof azoles with haloarenes: Scope, mechanism, and appli-cations to the synthesis of bioactive molecules. Chemistry17, 10113-10122.

[980] Bulawa CE, Connelly S, Devit M, Wang L, WeigelC, Fleming JA, Packman J, Powers ET, Wiseman RL,Foss TR, Wilson IA, Kelly JW, Labaudiniere R (2012)Tafamidis, a potent and selective transthyretin kinetic sta-bilizer that inhibits the amyloid cascade. Proc Natl AcadSci U S A 109, 9629-9634.

[981] Du J, Cho PY, Yang DT, Murphy RM (2012) Identifica-tion of beta-amyloid-binding sites on transthyretin. ProteinEng Des Sel 25, 337-345.

[982] Johnson SM, Connelly S, Fearns C, Powers ET, Kelly JW(2012) The transthyretin amyloidoses: From delineatingthe molecular mechanism of aggregation linked to pathol-ogy to a regulatory-agency-approved drug. J Mol Biol 421,185-203.

[983] Buxbaum JN, Linke RP (2012) A molecular history of theamyloidoses. J Mol Biol 421, 142-159.

[984] Dember LM, Hawkins PN, Hazenberg BP, Gorevic PD,Merlini G, Butrimiene I, Livneh A, Lesnyak O, PuechalX, Lachmann HJ, Obici L, Balshaw R, Garceau D, HauckW, Skinner M (2007) Eprodisate for the treatment of renaldisease in AA amyloidosis. N Engl J Med 356, 2349-2360.

[985] Manenti L, Tansinda P, Vaglio A (2007) Eprodisate in AAamyloidosis. N Engl J Med 357, 1153-1154.

Page 88: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

88 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[986] Manenti L, Tansinda P, Vaglio A (2008) Eprodisate in amy-loid A amyloidosis: A novel therapeutic approach? ExpertOpin Pharmacother 9, 2175-2180.

[987] Rule AD, Leung N (2007) Eprodisate slows the progres-sion of renal disease in patients with AA amyloidosis. NatClin Pract Nephrol 3, 592-593.

[988] Loeb MB, Molloy DW, Smieja M, Standish T, GoldsmithCH, Mahony J, Smith S, Borrie M, Decoteau E, DavidsonW, McDougall A, Gnarpe J, O’DONNell M, CherneskyM (2004) A randomized, controlled trial of doxycyclineand rifampin for patients with Alzheimer’s disease. J AmGeriatr Soc 52, 381-387.

[989] Salloway S, Sperling R, Keren R, Porsteinsson AP, vanDyck CH, Tariot PN, Gilman S, Arnold D, Abushakra S,Hernandez C, Crans G, Liang E, Quinn G, Bairu M, PastrakA, Cedarbaum JM (2011) A phase 2 randomized trial ofELND005, scyllo-inositol, in mild to moderate Alzheimerdisease. Neurology 77, 1253-1262.

[990] McLaurin J, Golomb R, Jurewicz A, Antel JP, FraserPE (2000) Inositol stereoisomers stabilize an oligomericaggregate of Alzheimer amyloid beta peptide and inhibitabeta -induced toxicity. J Biol Chem 275, 18495-18502.

[991] McLaurin J, Kierstead ME, Brown ME, Hawkes CA,Lambermon MH, Phinney AL, Darabie AA, Cousins JE,French JE, Lan MF, Chen F, Wong SS, Mount HT, FraserPE, Westaway D, St George-Hyslop P (2006) Cyclo-hexanehexol inhibitors of Abeta aggregation prevent andreverse Alzheimer phenotype in a mouse model. Nat Med12, 801-808.

[992] Fenili D, Brown M, Rappaport R, McLaurin J (2007)Properties of scyllo-inositol as a therapeutic treatment ofAD-like pathology. J Mol Med (Berl) 85, 603-611.

[993] Hawkes CA, Deng LH, Shaw JE, Nitz M, McLaurin J(2010) Small molecule beta-amyloid inhibitors that stabi-lize protofibrillar structures in vitro improve cognition andpathology in a mouse model of Alzheimer’s disease. EurJ Neurosci 31, 203-213.

[994] Townsend M, Cleary JP, Mehta T, Hofmeister J, Lesne S,O’Hare E, Walsh DM, Selkoe DJ (2006) Orally availablecompound prevents deficits in memory caused by the Alz-heimer amyloid-beta oligomers. Ann Neurol 60, 668-676.

[995] Griffith HR, Den Hollander JA, Stewart CC, EvanochkoWT, Buchthal SD, Harrell LE, Zamrini EY, Brocking-ton JC, Marson DC (2007) Elevated brain scyllo-inositolconcentrations in patients with Alzheimer’s disease. NMRBiomed 20, 709-716.

[996] Sun Y, Zhang G, Hawkes CA, Shaw JE, McLaurin J, NitzM (2008) Synthesis of scyllo-inositol derivatives and theireffects on amyloid beta peptide aggregation. Bioorg MedChem 16, 7177-7184.

[997] Vasdev N, Chio J, van Oosten EM, Nitz M, McLaurinJ, Vines DC, Houle S, Reilly RM, Wilson AA (2009)Synthesis and preliminary biological evaluations of [18F]-1-deoxy-1-fluoro-scyllo-inositol. Chem Commun (Camb)5527-5529.

[998] Sinha S, Du Z, Maiti P, Klarner FG, Schrader T, Wang C,Bitan G (2012) Comparison of Three Amyloid AssemblyInhibitors: The Sugar scyllo-Inositol, the Polyphenol Epi-gallocatechin Gallate, and the Molecular Tweezer CLR01.ACS Chem Neurosci 3, 451-458.

[999] Gunther EC, Strittmatter SM (2010) Beta-amyloidoligomers and cellular prion protein in Alzheimer’s dis-ease. J Mol Med (Berl) 88, 331-338.

[1000] Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmat-ter SM (2009) Cellular prion protein mediates impairment

of synaptic plasticity by amyloid-beta oligomers. Nature457, 1128-1132.

[1001] Nygaard HB, Strittmatter SM (2009) Cellular prion proteinmediates the toxicity of beta-amyloid oligomers: Impli-cations for Alzheimer disease. Arch Neurol 66, 1325-1328.

[1002] Chung E, Ji Y, Sun Y, Kascsak RJ, Kascsak RB, Mehta PD,Strittmatter SM, Wisniewski T (2010) Anti-PrPC mono-clonal antibody infusion as a novel treatment for cognitivedeficits in an Alzheimer’s disease model mouse. BMCNeurosci 11, 130.

[1003] Gimbel DA, Nygaard HB, Coffey EE, Gunther EC, LaurenJ, Gimbel ZA, Strittmatter SM (2010) Memory impair-ment in transgenic Alzheimer mice requires cellular prionprotein. J Neurosci 30, 6367-6374.

[1004] Kroth H, Ansaloni A, Varisco Y, Jan A, SreenivasacharyN, Rezaei-Ghaleh N, Giriens V, Lohmann S, Lopez-DeberMP, Adolfsson O, Pihlgren M, Paganetti P, Froestl W,Nagel-Steger L, Willbold D, Schrader T, Zweckstetter M,Pfeifer A, Lashuel HA, Muhs A (2012) Discovery andstructure activity relationship of small molecule inhibitorsof toxic beta-amyloid-42 fibril formation. J Biol Chem 287,34786-34800.

[1005] Rzepecki P, Wehner M, Molt O, Zadmard R, Harms K,Schrader T (2003) Aminopyrazole oligomers for beta-sheet stabilization of peptides. Synthesis, 1815-1826.

[1006] Rzepecki P, Nagel-Steger L, Feuerstein S, Linne U, MoltO, Zadmard R, Aschermann K, Wehner M, Schrader T,Riesner D (2004) Prevention of Alzheimer’s disease-associated Abeta aggregation by rationally designednonpeptidic beta-sheet ligands. J Biol Chem 279, 47497-47505.

[1007] Nagel-Steger L, Demeler B, Meyer-Zaika W, HochdorfferK, Schrader T, Willbold D (2010) Modulation of aggregatesize- and shape-distributions of the amyloid-beta peptideby a designed beta-sheet breaker. Eur Biophys J 39, 415-422.

[1008] Rzepecki P, Hochdorffer K, Schaller T, Zienau J, HarmsK, Ochsenfeld C, Xie X, Schrader T (2008) Hierarchicalself-assembly of aminopyrazole peptides into nanorosettesin water. J Am Chem Soc 130, 586-591.

[1009] Hochdorffer K, Marz-Berberich J, Nagel-Steger L, EppleM, Meyer-Zaika W, Horn AH, Sticht H, Sinha S, Bitan G,Schrader T (2011) Rational design of beta-sheet ligandsagainst Abeta42-induced toxicity. J Am Chem Soc 133,4348-4358.

[1010] Rzepecki P, Geib N, Peifer M, Biesemeier F, Schrader T(2007) Synthesis and binding studies of Alzheimer ligandson solid support. J Org Chem 72, 3614-3624.

[1011] Cernovska K, Kemter M, Gallmeier HC, Rzepecki P,Schrader T, Konig B (2004) PEG-supported synthesis ofpyrazole oligoamides with peptide beta-sheet affinity. OrgBiomol Chem 2, 1603-1611.

[1012] Rzepecki P, Schrader T (2005) beta-Sheet ligands inaction: KLVFF recognition by aminopyrazole hybridreceptors in water. J Am Chem Soc 127, 3016-3025.

[1013] Fricke H, Gerlach A, Unterberg C, Wehner M, Schrader T,Gerhards M (2009) Interactions of small protected pep-tides with aminopyrazole derivatives: The efficiency ofblocking a beta-sheet model in the gas phase. Angew ChemInt Ed Engl 48, 900-904.

[1014] Lecanu L, Tillement L, Rammouz G, Tillement JP, Gree-son J, Papadopoulos V (2009) Caprospinol: Moving froma neuroactive steroid to a neurotropic drug. Expert OpinInvestig Drugs 18, 265-276.

Page 89: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 89

[1015] Lecanu L, Rammouz G, McCourty A, Sidahmed EK,Greeson J, Papadopoulos V (2010) Caprospinol reducesamyloid deposits and improves cognitive function in a ratmodel of Alzheimer’s disease. Neuroscience 165, 427-435.

[1016] Tillement L, Lecanu L, Papadopoulos V (2011) Furtherevidence on mitochondrial targeting of beta-amyloid andspecificity of beta-amyloid-induced mitotoxicity in neu-rons. Neurodegener Dis 8, 331-344.

[1017] Papadopoulos V, Lecanu L (2012) Caprospinol: Discoveryof a steroid drug candidate to treat Alzheimer’s diseasebased on 22R-hydroxycholesterol structure and properties.J Neuroendocrinol 24, 93-101.

[1018] Howlett DR, George AR, Owen DE, Ward RV, Mark-well RE (1999) Common structural features determinethe effectiveness of carvedilol, daunomycin and rolite-tracycline as inhibitors of Alzheimer beta-amyloid fibrilformation. Biochem J 343 Pt 2, 419-423.

[1019] Fokkens M, Schrader T, Klarner FG (2005) A molecu-lar tweezer for lysine and arginine. J Am Chem Soc 127,14415-14421.

[1020] Sinha S, Lopes DH, Du Z, Pang ES, Shanmugam A,Lomakin A, Talbiersky P, Tennstaedt A, McDaniel K,Bakshi R, Kuo PY, Ehrmann M, Benedek GB, Loo JA,Klarner FG, Schrader T, Wang C, Bitan G (2011) Lysine-specific molecular tweezers are broad-spectrum inhibitorsof assembly and toxicity of amyloid proteins. J Am ChemSoc 133, 16958-16969.

[1021] Prabhudesai S, Sinha S, Attar A, Kotagiri A, Fitzmau-rice AG, Lakshmanan R, Ivanova MI, Loo JA, KlarnerFG, Schrader T, Stahl M, Bitan G, Bronstein JM (2012)A novel “molecular tweezer” inhibitor of alpha-synucleinneurotoxicity in vitro and in vivo. Neurotherapeutics 9,464-476.

[1022] Klarner FG, Schrader T (2012) Aromatic interactions bymolecular tweezers and clips in chemical and biologicalsystems. Acc Chem Res, doi: 10.1021/ar300061c [Epubahead of print].

[1023] Talbiersky P, Bastkowski F, Klarner FG, Schrader T (2008)Molecular clip and tweezer introduce new mechanisms ofenzyme inhibition. J Am Chem Soc 130, 9824-9828.

[1024] Klarner FG, Kahlert B, Nellesen A, Zienau J, Ochsen-feld C, Schrader T (2006) Molecular tweezer and clipin aqueous solution: Unexpected self-assembly, power-ful host-guest complex formation, quantum chemical 1HNMR shift calculation. J Am Chem Soc 128, 4831-4841.

[1025] Echeverria V, Zeitlin R (2012) Cotinine: A potential newtherapeutic agent against Alzheimer’s disease. CNS Neu-rosci Ther 18, 517-523.

[1026] Bergamaschini L, Rossi E, Storini C, Pizzimenti S, Dis-taso M, Perego C, De LA, Vergani C, De Simoni MG(2004) Peripheral treatment with enoxaparin, a low molec-ular weight heparin, reduces plaques and beta-amyloidaccumulation in a mouse model of Alzheimer’s disease.J Neurosci 24, 4181-4186.

[1027] Timmer NM, Herbert MK, Kleinovink JW, Kiliaan AJ,de Waal RM, Verbeek MM (2010) Limited expressionof heparan sulphate proteoglycans associated with Abetadeposits in the APPswe/PS1dE9 mouse model for Alzhe-imer’s disease. Neuropathol Appl Neurobiol 36, 478-486.

[1028] Timmer NM, van DL, van der Zee CE, Kiliaan A, deWaal RM, Verbeek MM (2010) Enoxaparin treatmentadministered at both early and late stages of amyloid betadeposition improves cognition of APPswe/PS1dE9 micewith differential effects on brain Abeta levels. NeurobiolDis 40, 340-347.

[1029] 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.

[1030] Petrlova J, Kalai T, Maezawa I, Altman R, HarishchandraG, Hong HS, Bricarello DA, Parikh AN, Lorigan GA, JinLW, Hideg K, Voss JC (2012) The influence of spin-labeledfluorene compounds on the assembly and toxicity of theabeta Peptide. PLoS One 7, e35443.

[1031] Chavant F, Deguil J, Pain S, Ingrand I, Milin S, Faucon-neau B, Perault-Pochat MC, Lafay-Chebassier C (2010)Imipramine, in part through tumor necrosis factor alphainhibition, prevents cognitive decline and beta-amyloidaccumulation in a mouse model of Alzheimer’s disease.J Pharmacol Exp Ther 332, 505-514.

[1032] Byun JH, Kim H, Kim Y, Mook-Jung I, Kim DJ, LeeWK, Yoo KH (2008) Aminostyrylbenzofuran derivativesas potent inhibitors for Abeta fibril formation. Bioorg MedChem Lett 18, 5591-5593.

[1033] Lee YS, Kim HY, Kim Y, Seo JH, Roh EJ, Han H,Shin KJ (2012) Small molecules that protect against beta-amyloid-induced cytotoxicity by inhibiting aggregation ofbeta-amyloid. Bioorg Med Chem 20, 4921-4935.

[1034] Choi Y, Kim HS, Shin KY, Kim EM, Kim M, Kim HS,Park CH, Jeong YH, Yoo J, Lee JP, Chang KA, Kim S,Suh YH (2007) Minocycline attenuates neuronal cell deathand improves cognitive impairment in Alzheimer’s diseasemodels. Neuropsychopharmacology 32, 2393-2404.

[1035] 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.

[1036] 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.

[1037] 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.

[1038] 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.J Neurosci 27, 3057-3063.

[1039] 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.

[1040] Kreutzmann P, Wolf G, Kupsch K (2010) Minocyclinerecovers MTT-formazan exocytosis impaired by amyloidbeta peptide. Cell Mol Neurobiol 30, 979-984.

[1041] 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.

[1042] Noble W, Garwood CJ, Hanger DP (2009) Minocycline asa potential therapeutic agent in neurodegenerative disor-ders characterised by protein misfolding. Prion 3, 78-83.

[1043] Garwood CJ, Cooper JD, Hanger DP, Noble W (2010)Anti-inflammatory impact of minocycline in a mousemodel of tauopathy. Front Psychiatr 1, 136.

[1044] Song Y, Wei EQ, Zhang WP, Ge QF, Liu JR, Wang ML,Huang XJ, Hu X, Chen Z (2006) Minocycline protects

Page 90: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

90 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

PC12 cells against NMDA-induced injury via inhibiting5-lipoxygenase activation. Brain Res 1085, 57-67.

[1045] Hashimoto K (2011) Can minocycline prevent the onset ofAlzheimer’s disease? Ann Neurol 69, 739-740.

[1046] Chaudhry IB, Hallak J, Husain N, Minhas F, Stir-ling J, Richardson P, Dursun S, Dunn G, DeakinB (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.

[1047] Tomiyama T, Asano S, Suwa Y, Morita T, Kataoka K, MoriH, Endo N (1994) Rifampicin prevents the aggregation andneurotoxicity of amyloid beta protein in vitro. BiochemBiophys Res Commun 204, 76-83.

[1048] Tomiyama T, Shoji A, Kataoka K, Suwa Y, Asano S,Kaneko H, Endo N (1996) Inhibition of amyloid betaprotein aggregation and neurotoxicity by rifampicin. Itspossible function as a hydroxyl radical scavenger. J BiolChem 271, 6839-6844.

[1049] Tomiyama T, Kaneko H, Kataoka K, Asano S, Endo N(1997) Rifampicin inhibits the toxicity of pre-aggregatedamyloid peptides by binding to peptide fibrils and pre-venting amyloid-cell interaction. Biochem J 322(Pt 3),859-865.

[1050] Qosa H, Abuznait AH, Hill RA, Kaddoumi A (2012)Enhanced brain amyloid-beta clearance by rifampicinand caffeine as a possible protective mechanism againstAlzheimer’s disease. J Alzheimers Dis 31, 151-165.

[1051] Forloni G, Colombo L, Girola L, Tagliavini F, SalmonaM (2001) Anti-amyloidogenic activity of tetracyclines:Studies in vitro. FEBS Lett 487, 404-407.

[1052] Diomede L, Cassata G, Fiordaliso F, Salio M, Ami D,Natalello A, Doglia SM, De LA, Salmona M (2010)Tetracycline and its analogues protect Caenorhabditis ele-gans from beta amyloid-induced toxicity by targetingoligomers. Neurobiol Dis 40, 424-431.

[1053] Averback P (1998) Spherons as a drug target inAlzheimer’s disease. Drug News Perspect 11, 469-479.

[1054] Averback P, Morse D, Ghanbari H (1998) Bursting densemicrospheres (spherons) in Alzheimer’s disease: A reviewof studies (1980–1997) on spherons and the pathogenesisof Alzheimer’s disease. J Alzheimers Dis 1, 1-34.

[1055] Garofalo AW, Wone DW, Phuc A, Audia JE, Bales CA,Dovey HF, Dressen DB, Folmer B, Goldbach EG, GuinnAC, Latimer LH, Mabry TE, Nissen JS, Pleiss MA, Sohn S,Thorsett ED, Tung JS, Wu J (2002) A series of C-terminalamino alcohol dipeptide A beta inhibitors. Bioorg MedChem Lett 12, 3051-3053.

[1056] Friedman OM, Matsudaira P, Reis AH Jr, Simister N,Correia I, Kew D, Wei JY, Pochapsky T (2007) Substitutedorganosiloxanes as potential therapeutics for treatment andprevention of neurodegenerative diseases. J AlzheimersDis 11, 291-300.

[1057] Wood SJ, MacKenzie L, Maleeff B, Hurle MR, Wetzel R(1996) Selective inhibition of Abeta fibril formation. J BiolChem 271, 4086-4092.

[1058] Howlett DR, Perry AE, Godfrey F, Swatton JE, JenningsKH, Spitzfaden C, Wadsworth H, Wood SJ, Markwell RE(1999) Inhibition of fibril formation in beta-amyloid pep-tide by a novel series of benzofurans. Biochem J 340(Pt1), 283-289.

[1059] Parker MH, Chen R, Conway KA, Lee DH, Luo C,Boyd RE, Nortey SO, Ross TM, Scott MK, ReitzAB (2002) Synthesis of (-)-5,8-dihydroxy-3R-methyl-2R-(dipropylamino)-1,2,3,4-tetrahydronaphthale ne: An

inhibitor of beta-amyloid(1-42) aggregation. Bioorg MedChem 10, 3565-3569.

[1060] Simons LJ, Caprathe BW, Callahan M, Graham JM,Kimura T, Lai Y, LeVine H III, Lipinski W, Sakkab AT,Tasaki Y, Walker LC, Yasunaga T, Ye Y, Zhuang N,ugelli-Szafran CE (2009) The synthesis and structure-activity relationship of substituted N-phenyl anthranilicacid analogs as amyloid aggregation inhibitors. BioorgMed Chem Lett 19, 654-657.

[1061] Merlini G, Ascari E, Amboldi N, Bellotti V, Arbustini E,Perfetti V, Ferrari M, Zorzoli I, Marinone MG, GariniP, et al. (1995) Interaction of the anthracycline 4′-iodo-4′-deoxydoxorubicin with amyloid fibrils: Inhibition ofamyloidogenesis. Proc Natl Acad Sci U S A 92, 2959-2963.

[1062] Higaki JN, Chakravarty S, Bryant CM, Cowart LR, HardenP, Scardina JM, Mavunkel B, Luedtke GR, Cordell B(1999) A combinatorial approach to the identification ofdipeptide aldehyde inhibitors of beta-amyloid production.J Med Chem 42, 3889-3898.

[1063] Kisilevsky R, Lemieux LJ, Fraser PE, Kong X, Hultin PG,Szarek WA (1995) Arresting amyloidosis in vivo usingsmall-molecule anionic sulphonates or sulphates: Impli-cations for Alzheimer’s disease. Nat Med 1, 143-148.

[1064] Kisilevsky R (1996) Anti-amyloid drugs: Potential in thetreatment of diseases associated with aging. Drugs Aging8, 75-83.

[1065] Kisilevsky R (1997) Can deposition of amyloid be pre-vented in Alzheimer’s disease? Ann N Y Acad Sci 826,117-127.

[1066] Tjernberg LO, Lilliehook C, Callaway DJ, Naslund J,Hahne S, Thyberg J, Terenius L, Nordstedt C (1997)Controlling amyloid beta-peptide fibril formation withprotease-stable ligands. J Biol Chem 272, 12601-12605.

[1067] Banks WA, Farr SA, Butt W, Kumar VB, Franko MW, Mor-ley JE (2001) Delivery across the blood-brain barrier ofantisense directed against amyloid beta: Reversal of learn-ing and memory deficits in mice overexpressing amyloidprecursor protein. J Pharmacol Exp Ther 297, 1113-1121.

[1068] Heal JR, Roberts GW, Christie G, Miller AD (2002) Inhi-bition of beta-amyloid aggregation and neurotoxicity bycomplementary (antisense) peptides. Chembiochem 3, 86-92.

[1069] Gestwicki JE, Crabtree GR, Graef IA (2004) Harness-ing chaperones to generate small-molecule inhibitors ofamyloid beta aggregation. Science 306, 865-869.

[1070] Lin SJ, Shiao YJ, Chi CW, Yang LM (2004) Abeta aggrega-tion inhibitors. Part 1: Synthesis and biological activity ofphenylazo benzenesulfonamides. Bioorg Med Chem Lett14, 1173-1176.

[1071] Bose M, Gestwicki JE, Devasthali V, Crabtree GR, GraefIA (2005) ‘Nature-inspired’ drug-protein complexes asinhibitors of Abeta aggregation. Biochem Soc Trans 33,543-547.

[1072] Lee KH, Shin BH, Shin KJ, Kim DJ, Yu J (2005) A hybridmolecule that prohibits amyloid fibrils and alleviates neu-ronal toxicity induced by beta-amyloid (1-42). BiochemBiophys Res Commun 328, 816-823.

[1073] Schwarzman AL, Tsiper M, Gregori L, Goldgaber D,Frakowiak J, Mazur-Kolecka B, Taraskina A, PchelinaS, Vitek MP (2005) Selection of peptides binding to theamyloid �-protein reveals potential inhibitors of amyloidformation. Amyloid 12, 199-209.

[1074] Cohen T, Frydman-Marom A, Rechter M, Gazit E (2006)Inhibition of amyloid fibril formation and cytotoxicity byhydroxyindole derivatives. Biochemistry 45, 4727-4735.

Page 91: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 91

[1075] Dolphin GT, Dumy P, Garcia J (2006) Control of amyloidbeta-peptide protofibril formation by a designed templateassembly. Angew Chem Int Ed Engl 45, 2699-2702.

[1076] Etienne MA, Aucoin JP, Fu Y, McCarley RL, HammerRP (2006) Stoichiometric inhibition of amyloid beta-protein aggregation with peptides containing alternatingalpha,alpha-disubstituted amino acids. J Am Chem Soc128, 3522-3523.

[1077] Inbar P, Yang J (2006) Inhibiting protein-amyloid interac-tions with small molecules: A surface chemistry approach.Bioorg Med Chem Lett 16, 1076-1079.

[1078] Inbar P, Li CQ, Takayama SA, Bautista MR, Yang J(2006) Oligo(ethylene glycol) derivatives of thioflavin T asinhibitors of protein-amyloid interactions. Chembiochem7, 1563-1566.

[1079] Kwak JW, Kim HK, Chae CB (2006) Potential lead foran Alzheimer drug: A peptide that blocks intermolecularinteraction and amyloid beta protein-induced cytotoxicity.J Med Chem 49, 4813-4817.

[1080] Orner BP, Liu L, Murphy RM, Kiessling LL (2006) Phagedisplay affords peptides that modulate beta-amyloid aggre-gation. J Am Chem Soc 128, 11882-11889.

[1081] Pai AS, Rubinstein I, Onyuksel H (2006) PEGylated phos-pholipid nanomicelles interact with beta-amyloid(1-42)and mitigate its beta-sheet formation, aggregation and neu-rotoxicity in vitro. Peptides 27, 2858-2866.

[1082] Porat Y, Abramowitz A, Gazit E (2006) Inhibition of amy-loid fibril formation by polyphenols: Structural similarityand aromatic interactions as a common inhibition mecha-nism. Chem Biol Drug Des 67, 27-37.

[1083] Byeon SR, Lee JH, Sohn JH, Kim DC, Shin KJ, Yoo KH,Mook-Jung I, Lee WK, Kim DJ (2007) Bis-styrylpyridineand bis-styrylbenzene derivatives as inhibitors for Abetafibril formation. Bioorg Med Chem Lett 17, 1466-1470.

[1084] Byeon SR, Jin YJ, Lim SJ, Lee JH, Yoo KH, Shin KJ, OhSJ, Kim DJ (2007) Ferulic acid and benzothiazole dimerderivatives with high binding affinity to beta-amyloid fib-rils. Bioorg Med Chem Lett 17, 4022-4025.

[1085] Dolphin GT, Ouberai M, Dumy P, Garcia J (2007)Designed amyloid beta peptide fibril –a tool for high-throughput screening of fibril inhibitors. ChemMedChem2, 1613-1623.

[1086] Chafekar SM, Malda H, Merkx M, Meijer EW, Viertl D,Lashuel HA, Baas F, Scheper W (2007) Branched KLVFFtetramers strongly potentiate inhibition of beta-amyloidaggregation. Chembiochem 8, 1857-1864.

[1087] Etienne MA, Edwin NJ, Aucoin JP, Russo PS, McCarleyRL, Hammer RP (2007) Beta-amyloid protein aggrega-tion. Methods Mol Biol 386, 203-225.

[1088] Flaherty DP, Walsh SM, Kiyota T, Dong Y, Ikezu T,Vennerstrom JL (2007) Polyfluorinated bis-styrylbenzenebeta-amyloid plaque binding ligands. J Med Chem 50,4986-4992.

[1089] Honson NS, Johnson RL, Huang W, Inglese J, AustinCP, Kuret J (2007) Differentiating Alzheimer disease-associated aggregates with small molecules. Neurobiol Dis28, 251-260.

[1090] Necula M, Kayed R, Milton S, Glabe CG (2007) Smallmolecule inhibitors of aggregation indicate that amyloidbeta oligomerization and fibrillization pathways are inde-pendent and distinct. J Biol Chem 282, 10311-10324.

[1091] Okuno H, Mori K, Okada T, Yokoyama Y, Suzuki H (2007)Development of aggregation inhibitors for amyloid-betapeptides and their evaluation by quartz-crystal microbal-ance. Chem Biol Drug Des 69, 356-361.

[1092] Riviere C, Richard T, Quentin L, Krisa S, MerillonJM, Monti JP (2007) Inhibitory activity of stilbenes onAlzheimer’s beta-amyloid fibrils in vitro. Bioorg MedChem 15, 1160-1167.

[1093] Sato J, Takahashi T, Oshima H, Matsumura S, Mihara H(2007) Design of peptides that form amyloid-like fibrilscapturing amyloid beta1-42 peptides. Chemistry 13, 7745-7752.

[1094] Takahashi T, Ohta K, Mihara H (2007) Embedding theamyloid beta-peptide sequence in green fluorescent proteininhibits Abeta oligomerization. Chembiochem 8, 985-988.

[1095] Austen BM, Paleologou KE, Ali SA, Qureshi MM, AllsopD, El-Agnaf OM (2008) Designing peptide inhibitors foroligomerization and toxicity of Alzheimer’s beta-amyloidpeptide. Biochemistry 47, 1984-1992.

[1096] Bravo R, Arimon M, Valle-Delgado JJ, Garcia R, DuranyN, Castel S, Cruz M, Ventura S, Fernandez-Busquets X(2008) Sulfated polysaccharides promote the assembly ofamyloid beta(1-42) peptide into stable fibrils of reducedcytotoxicity. J Biol Chem 283, 32471-32483.

[1097] Cabaleiro-Lago C, Quinlan-Pluck F, Lynch I, Lindman S,Minogue AM, Thulin E, Walsh DM, Dawson KA, Linse S(2008) Inhibition of amyloid beta protein fibrillation bypolymeric nanoparticles. J Am Chem Soc 130, 15437-15443.

[1098] Cellamare S, Stefanachi A, Stolfa DA, Basile T, CattoM, Campagna F, Sotelo E, Acquafredda P, Carotti A(2008) Design, synthesis, and biological evaluation ofglycine-based molecular tongs as inhibitors of Abeta1-40aggregation in vitro. Bioorg Med Chem 16, 4810-4822.

[1099] Dolphin GT, Chierici S, Ouberai M, Dumy P, Garcia J(2008) A multimeric quinacrine conjugate as a poten-tial inhibitor of Alzheimer’s beta-amyloid fibril formation.Chembiochem 9, 952-963.

[1100] Esteras-Chopo A, Pastor MT, Serrano L, Lopez de laPM (2008) New strategy for the generation of specificD-peptide amyloid inhibitors. J Mol Biol 377, 1372-1381.

[1101] Feng BY, Toyama BH, Wille H, Colby DW, Collins SR,May BC, Prusiner SB, Weissman J, Shoichet BK (2008)Small-molecule aggregates inhibit amyloid polymeriza-tion. Nat Chem Biol 4, 197-199.

[1102] Lee JH, Byeon SR, Lim SJ, Oh SJ, Moon DH, Yoo KH,Chung BY, Kim DJ (2008) Synthesis and evaluation of stil-benylbenzoxazole and stilbenylbenzothiazole derivativesfor detecting beta-amyloid fibrils. Bioorg Med Chem Lett18, 1534-1537.

[1103] Velkova A, Tatarek-Nossol M, Andreetto E, KapurniotuA (2008) Exploiting cross-amyloid interactions to inhibitprotein aggregation but not function: Nanomolar affinityinhibition of insulin aggregation by an IAPP mimic. AngewChem Int Ed Engl 47, 7114-7118.

[1104] Wang H, Duennwald ML, Roberts BE, Rozeboom LM,Zhang YL, Steele AD, Krishnan R, Su LJ, Griffin D,Mukhopadhyay S, Hennessy EJ, Weigele P, Blanchard BJ,King J, Deniz AA, Buchwald SL, Ingram VM, LindquistS, Shorter J (2008) Direct and selective elimination of spe-cific prions and amyloids by 4,5-dianilinophthalimide andanalogs. Proc Natl Acad Sci U S A 105, 7159-7164.

[1105] Wisniewski T, Sadowski M (2008) Preventing beta-amyloid fibrillization and deposition: Beta-sheet breakersand pathological chaperone inhibitors. BMC Neurosci9(Suppl 2), S5.

[1106] Barthel A, Trieschmann L, Strohl D, Kluge R, Bohm G,Csuk R (2009) Synthesis of dimeric quinazolin-2-one,1,4-benzodiazepin-2-one, and isoalloxazine compounds as

Page 92: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

92 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

inhibitors of amyloid peptides association. Arch Pharm(Weinheim) 342, 445-452.

[1107] Campiglia P, Scrima M, Grimaldi M, Cioffi G, BertaminoA, Sala M, Aquino C, Gomez-Monterrey I, GriecoP, Novellino E, D’Ursi AM (2009) A new series of1,3-dihidro-imidazo[1,5-c]thiazole-5,7-dione derivatives:Synthesis and interaction with Abeta(25-35) amyloid pep-tide. Chem Biol Drug Des 74, 224-233.

[1108] Csuk R, Barthel A, Raschke C, Kluge R, Strohl D, Tri-eschmann L, Bohm G (2009) Synthesis of monomeric anddimeric acridine compounds as potential therapeutics inAlzheimer and prion diseases. Arch Pharm (Weinheim)342, 699-709.

[1109] Frydman-Marom A, Rechter M, Shefler I, Bram Y, ShalevDE, Gazit E (2009) Cognitive-performance recovery ofAlzheimer’s disease model mice by modulation of earlysoluble amyloidal assemblies. Angew Chem Int Ed Engl48, 1981-1986.

[1110] Giordano C, Masi A, Pizzini A, Sansone A, Consalvi V,Chiaraluce R, Lucente G (2009) Synthesis and activityof fibrillogenesis peptide inhibitors related to the 17-21beta-amyloid sequence. Eur J Med Chem 44, 179-189.

[1111] Grillo-Bosch D, Carulla N, Cruz M, Sanchez L, Pujol-Pina R, Madurga S, Rabanal F, Giralt E (2009)Retro-enantio N-methylated peptides as beta-amyloidaggregation inhibitors. ChemMedChem 4, 1488-1494.

[1112] Madine J, Wang X, Brown DR, Middleton DA (2009)Evaluation of beta-alanine- and GABA-substituted pep-tides as inhibitors of disease-linked protein aggregation.Chembiochem 10, 1982-1987.

[1113] Dolphin GT, Renaudet O, Ouberai M, Dumy P, GarciaJ, Reymond JL (2009) Phenolic oxime oligomers inhibitAlzheimer’s amyloid fibril formation and disaggregate fib-rils in vitro. Chembiochem 10, 1325-1329.

[1114] Catto M, Aliano R, Carotti A, Cellamare S, Pallu-otto F, Purgatorio R, De SA, Campagna F (2010)Design, synthesis and biological evaluation ofindane-2-arylhydrazinylmethylene-1,3-diones andindol-2-aryldiazenylmethylene-3-ones as beta-amyloidaggregation inhibitors. Eur J Med Chem 45, 1359-1366.

[1115] Matharu B, El-Agnaf O, Razvi A, Austen BM (2010)Development of retro-inverso peptides as anti-aggregationdrugs for beta-amyloid in Alzheimer’s disease. Peptides31, 1866-1872.

[1116] Suzuki M, Takahashi T, Sato J, Mie M, Kobatake E, MiharaH (2010) Designed short peptides that form amyloid-likefibrils in coassembly with amyloid beta-peptide (Abeta)decrease the toxicity of Abeta to neuronal PC12 cells.Chembiochem 11, 1525-1530.

[1117] Zhou Y, Jiang C, Zhang Y, Liang Z, Liu W, Wang L, LuoC, Zhong T, Sun Y, Zhao L, Xie X, Jiang H, Zhou N, LiuD, Liu H (2010) Structural optimization and biologicalevaluation of substituted bisphenol A derivatives as beta-amyloid peptide aggregation inhibitors. J Med Chem 53,5449-5466.

[1118] Ladiwala AR, Dordick JS, Tessier PM (2011) Aromaticsmall molecules remodel toxic soluble oligomers of amy-loid beta through three independent pathways. J Biol Chem286, 3209-3218.

[1119] Ortega A, Rincon A, Jimenez-Aliaga KL, Bermejo-BescosP, Martin-Aragon S, Molina MT, Csaky AG (2011) Syn-thesis and evaluation of arylquinones as BACE1 inhibitors,beta-amyloid peptide aggregation inhibitors, and destabi-lizers of preformed beta-amyloid fibrils. Bioorg Med ChemLett 21, 2183-2187.

[1120] Ryan TM, Griffin MD, Teoh CL, Ooi J, Howlett GJ (2011)High-affinity amphipathic modulators of amyloid fibrilnucleation and elongation. J Mol Biol 406, 416-429.

[1121] Ryan TM, Griffin MD, Bailey MF, Schuck P, Howlett GJ(2011) NBD-labeled phospholipid accelerates apolipopro-tein C-II amyloid fibril formation but is not incorporatedinto mature fibrils. Biochemistry 50, 9579-9586.

[1122] Frydman-Marom A, Shaltiel-Karyo R, Moshe S, Gazit E(2011) The generic amyloid formation inhibition effect ofa designed small aromatic beta-breaking peptide. Amyloid18, 119-127.

[1123] Lukiw WJ (2012) Amyloid beta (Abeta) peptide modula-tors and other current treatment strategies for Alzheimer’sdisease (AD). Expert Opin Emerg Drugs 17, 43-60.

[1124] Di Santo R, Costi R, Cuzzucoli CG, Pescatori L, Rosi F,Scipione L, Celona D, Vertechy M, Ghirardi O, PiovesanP, Marzi M, Caccia S, Guiso G, Giorgi F, Minetti P (2012)Design, synthesis and structure-activity relationship of N-Aryl-naphthylamine derivatives as amyloid aggregationinhibitors. J Med Chem 55, 8538-8548.

[1125] Cheng PN, Liu C, Zhao M, Eisenberg D, Nowick JS (2012)Amyloid beta-sheet mimics that antagonize protein aggre-gation and reduce amyloid toxicity. Nat Chem 4, 927-933.

[1126] Huang L, Lu C, Sun Y, Mao F, Luo Z, Su T,Jiang H, Shan W, Li X (2012) Multi-target-directedbenzylidene-indanone derivatives: Anti-beta amyloid(Abeta) aggregation, antioxidant, metal chelation andmonoamine oxidase B (MAO-B) inhibition propertiesagainst Alzheimer’s disease. J Med Chem 55, 8483-8492.

[1127] Liu R, Su R, Liang M, Huang R, Wang M, Qi W, He Z(2012) Physicochemical strategies for inhibition of amy-loid fibril formation: An overview of recent advances. CurrMed Chem 19, 4157-4174.

[1128] Ryan TM, Friedhuber A, Lind M, Howlett GJ, Mas-ters C, Roberts BR (2012) Small amphipathic moleculesmodulate secondary structure and amyloid fibril-formingkinetics of Alzheimer disease peptide Abeta(1-42). J BiolChem 287, 16947-16954.

[1129] Lopez LC, Dos-Reis S, Espargaro A, Carrodeguas JA,Maddelein ML, Ventura S, Sancho J (2012) Discovery ofnovel inhibitors of amyloid beta-peptide 1-42 aggregation.J Med Chem 55, 9521-9530.

[1130] Guo JT, Xu Y (2006) Amyloid fibril structure modelingusing protein threading and molecular dynamics simula-tions. Methods Enzymol 412, 300-314.

[1131] Guo JT, Xu Y (2008) Towards modeling of amyloid fibrilstructures. Front Biosci 13, 4039-4050.

[1132] Esteras-Chopo A, Morra G, Moroni E, Serrano L, Lopezde la PM, Colombo G (2008) A molecular dynamics studyof the interaction of D-peptide amyloid inhibitors withtheir target sequence reveals a potential inhibitory phar-macophore conformation. J Mol Biol 383, 266-280.

[1133] Wu C, Wang Z, Lei H, Duan Y, Bowers MT, Shea JE (2008)The binding of thioflavin T and its neutral analog BTA-1 to protofibrils of the Alzheimer’s disease Abeta(16-22)peptide probed by molecular dynamics simulations. J MolBiol 384, 718-729.

[1134] Wu C, Murray MM, Bernstein SL, Condron MM, BitanG, Shea JE, Bowers MT (2009) The structure of Abeta42C-terminal fragments probed by a combined experimentaland theoretical study. J Mol Biol 387, 492-501.

[1135] Chebaro Y, Mousseau N, Derreumaux P (2009) Struc-tures and thermodynamics of Alzheimer’s amyloid-betaAbeta(16-35) monomer and dimer by replica exchangemolecular dynamics simulations: Implication for full-

Page 93: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 93

length Abeta fibrillation. J Phys Chem B 113, 7668-7675.

[1136] Chebaro Y, Derreumaux P (2009) Targeting the early stepsof Abeta16-22 protofibril disassembly by N-methylatedinhibitors: A numerical study. Proteins 75, 442-452.

[1137] Convertino M, Pellarin R, Catto M, Carotti A, CaflischA (2009) 9,10-Anthraquinone hinders beta-aggregation:How does a small molecule interfere with Abeta-peptideamyloid fibrillation? Protein Sci 18, 792-800.

[1138] Frydman-Marom A, Convertino M, Pellarin R, LampelA, Shaltiel-Karyo R, Segal D, Caflisch A, Shalev DE,Gazit E (2011) Structural basis for inhibiting beta-amyloidoligomerization by a non-coded beta-breaker-substitutedendomorphin analogue. ACS Chem Biol 6, 1265-1276.

[1139] Stempler S, Levy-Sakin M, Frydman-Marom A, Amir Y,Scherzer-Attali R, Buzhansky L, Gazit E, Senderowitz H(2011) Quantitative structure-activity relationship analysisof beta-amyloid aggregation inhibitors. J Comput AidedMol Des 25, 135-144.

[1140] Wise-Scira O, Xu L, Kitahara T, Perry G, Coskuner O(2011) Amyloid-beta peptide structure in aqueous solutionvaries with fragment size. J Chem Phys 135, 205101.

[1141] Convertino M, Vitalis A, Caflisch A (2011) Disorderedbinding of small molecules to Abeta(12-28). J Biol Chem286, 41578-41588.

[1142] Chebaro Y, Jiang P, Zang T, Mu Y, Nguyen PH, MousseauN, Derreumaux P (2012) Structures of abeta17-42 trimersin isolation and with five small-molecule drugs using ahierarchical computational procedure. J Phys Chem B 116,8412-8422.

[1143] Novick PA, Lopes DH, Branson KM, Esteras-Chopo A,Graef IA, Bitan G, Pande VS (2012) Design of beta-amyloid aggregation inhibitors from a predicted structuralmotif. J Med Chem 55, 3002-3010.

[1144] Srivastava A, Balaji PV (2012) Size, orientation and orga-nization of oligomers that nucleate amyloid fibrils: Cluesfrom MD simulations of pre-formed aggregates. BiochimBiophys Acta 1824, 963-973.

[1145] Greenberg SM, Rosand J, Schneider AT, Creed PL, GandySE, Rovner B, Fitzsimmons BF, Smith EE, Edip GM,Schwab K, Laurin J, Garceau D (2006) A phase 2 study oftramiprosate for cerebral amyloid angiopathy. AlzheimerDis Assoc Disord 20, 269-274.

[1146] Wright TM (2006) Tramiprosate. Drugs Today (Barc) 42,291-298.

[1147] Gervais F, Paquette J, Morissette C, Krzywkowski P, Yu M,Azzi M, Lacombe D, Kong X, Aman A, Laurin J, SzarekWA, Tremblay P (2007) Targeting soluble Abeta peptidewith Tramiprosate for the treatment of brain amyloidosis.Neurobiol Aging 28, 537-547.

[1148] Gauthier S, Aisen PS, Ferris SH, Saumier D, Duong A,Haine D, Garceau D, Suhy J, Oh J, Lau W, SampalisJ (2009) Effect of tramiprosate in patients with mild-to-moderate Alzheimer’s disease: Exploratory analyses of theMRI sub-group of the Alphase study. J Nutr Health Aging13, 550-557.

[1149] Saumier D, Aisen PS, Gauthier S, Vellas B, Ferris SH,Duong A, Suhy J, Oh J, Lau W, Garceau D, Haine D,Sampalis J (2009) Lessons learned in the use of volumet-ric MRI in therapeutic trials in Alzheimer’s disease: TheALZHEMED (Tramiprosate) experience. J Nutr HealthAging 13, 370-372.

[1150] Saumier D, Duong A, Haine D, Garceau D, Sampalis J(2009) Domain-specific cognitive effects of tramiprosate

in patients with mild to moderate Alzheimer’s disease:ADAS-cog subscale results from the Alphase Study. J NutrHealth Aging 13, 808-812.

[1151] Aisen PS, Gauthier S, Ferris SH, Saumier D, Haine D,Garceau D, Duong A, Suhy J, Oh J, Lau WC, SampalisJ (2011) Tramiprosate in mild-to-moderate Alzheimer’sdisease - a randomized, double-blind, placebo-controlled,multi-centre study (the Alphase Study). Arch Med Sci 7,102-111.

[1152] Swanoski MT (2009) Homotaurine: A failed drug forAlzheimer’s disease and now a nutraceutical for mem-ory protection. Am J Health Syst Pharm 66, 1950-1953.

[1153] Adessi C, Frossard MJ, Boissard C, Fraga S, Bieler S,Ruckle T, Vilbois F, Robinson SM, Mutter M, Banks WA,Soto C (2003) Pharmacological profiles of peptide drugcandidates for the treatment of Alzheimer’s disease. J BiolChem 278, 13905-13911.

[1154] Uryu S, Tokuhiro S, Murasugi T, Oda T (2002) Anovel compound, RS-1178, specifically inhibits neuronalcell death mediated by beta-amyloid-induced macrophageactivation in vitro. Brain Res 946, 298-306.

[1155] Nakagami Y, Nishimura S, Murasugi T, Kaneko I, MeguroM, Marumoto S, Kogen H, Koyama K, Oda T (2002)A novel beta-sheet breaker, RS-0406, reverses amyloidbeta-induced cytotoxicity and impairment of long-termpotentiation in vitro. Br J Pharmacol 137, 676-682.

[1156] Nakagami Y, Nishimura S, Murasugi T, Kubo T, KanekoI, Meguro M, Marumoto S, Kogen H, Koyama K, OdaT (2002) A novel compound RS-0466 reverses beta-amyloid-induced cytotoxicity through the Akt signalingpathway in vitro. Eur J Pharmacol 457, 11-17.

[1157] Walsh DM, Townsend M, Podlisny MB, Shankar GM,Fadeeva JV, El AO, Hartley DM, Selkoe DJ (2005) Cer-tain inhibitors of synthetic amyloid beta-peptide (Abeta)fibrillogenesis block oligomerization of natural Abeta andthereby rescue long-term potentiation. J Neurosci 25,2455-2462.

[1158] O’Hare E, Scopes DI, Treherne JM, Norwood K, Span-swick D, Kim EM (2010) RS-0406 arrests amyloid-betaoligomer-induced behavioural deterioration in vivo. BehavBrain Res 210, 32-37.

[1159] Rodrigue KM, Kennedy KM, Park DC (2009) Beta-amyloid deposition and the aging brain. Neuropsychol Rev19, 436-450.

[1160] Rodrigue KM, Kennedy KM, Devous MD, Rieck JR,Hebrank AC, Diaz-Arrastia R, Mathews D, Park DC(2012) beta-Amyloid burden in healthy aging: Regionaldistribution and cognitive consequences. Neurology 78,387-395.

[1161] Cohen RM (2007) The application of positron-emittingmolecular imaging tracers in Alzheimer’s disease. MolImaging Biol 9, 204-216.

[1162] Mathis CA, Wang Y, Holt DP, Huang GF, Debnath ML,Klunk WE (2003) Synthesis and evaluation of 11C-labeled6-substituted 2-arylbenzothiazoles as amyloid imagingagents. J Med Chem 46, 2740-2754.

[1163] Blennow K, de Leon MJ, Zetterberg H (2006) Alzheimer’sdisease. Lancet 368, 387-403.

[1164] Fagan AM, Mintun MA, Mach RH, Lee SY, DenceCS, Shah AR, LaRossa GN, Spinner ML, Klunk WE,Mathis CA, Dekosky ST, Morris JC, Holtzman DM (2006)Inverse relation between in vivo amyloid imaging load andcerebrospinal fluid Abeta42 in humans. Ann Neurol 59,512-519.

Page 94: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

94 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1165] Quigley H, Colloby SJ, O’Brien JT (2011) PET imag-ing of brain amyloid in dementia: A review. Int J GeriatrPsychiatry 26, 991-999.

[1166] Tarawneh R, Holtzman DM (2010) Biomarkers intranslational research of Alzheimer’s disease. Neurophar-macology 59, 310-322.

[1167] Forsberg A, Almkvist O, Engler H, Wall A, Langstrom B,Nordberg A (2010) High PIB retention in Alzheimer’s dis-ease is an early event with complex relationship with CSFbiomarkers and functional parameters. Curr Alzheimer Res7, 56-66.

[1168] Grimmer T, Henriksen G, Wester HJ, Forstl H, Klunk WE,Mathis CA, Kurz A, Drzezga A (2009) Clinical severity ofAlzheimer’s disease is associated with PIB uptake in PET.Neurobiol Aging 30, 1902-1909.

[1169] Ances BM, Benzinger TL, Christensen JJ, Thomas J,Venkat R, Teshome M, Aldea P, Fagan AM, Holtzman DM,Morris JC, Clifford DB (2012) 11C-PiB imaging of humanimmunodeficiency virus-associated neurocognitive disor-der. Arch Neurol 69, 72-77.

[1170] Kung HF (2012) The beta-amyloid hypotheis inAlzheimer’s disease: Seeing is believing. ACS Med ChemLett 3, 265-267.

[1171] Ossenkoppele R, van Berckel BN, Prins ND (2011)Amyloid imaging in prodromal Alzheimer’s disease.Alzheimers Res Ther 3, 26.

[1172] Ossenkoppele R, Tolboom N, Foster-Dingley JC, Adri-aanse SF, Boellaard R, Yaqub M, Windhorst AD,Barkhof F, Lammertsma AA, Scheltens P, van der FlierWM, van Berckel BN (2012) Longitudinal imaging ofAlzheimer pathology using [(11)C]PIB, [(18)F]FDDNPand [(18)F]FDG PET. Eur J Nucl Med Mol Imaging 39,990-1000.

[1173] Ossenkoppele R, Zwan MD, Tolboom N, van AssemaDM, Adriaanse SF, Kloet RW, Boellaard R, WindhorstAD, Barkhof F, Lammertsma AA, Scheltens P, van derFlier WM, van Berckel BN (2012) Amyloid burden andmetabolic function in early-onset Alzheimer’s disease:Parietal lobe involvement. Brain 135, 2115-2125.

[1174] Small GW, Siddarth P, Kepe V, Ercoli LM, Burggren AC,Bookheimer SY, Miller KJ, Kim J, Lavretsky H, HuangSC, Barrio JR (2012) Prediction of cognitive decline bypositron emission tomography of brain amyloid and tau.Arch Neurol 69, 215-222.

[1175] Herholz K (2012) Imaging cerebral amyloid plaques: Clin-ical perspective. Lancet Neurol 11, 652-653.

[1176] Driscoll I, Troncoso JC, Rudow G, Sojkova J, PletnikovaO, Zhou Y, Kraut MA, Ferrucci L, Mathis CA, Klunk WE,O’Brien RJ, Davatzikos C, Wong DF, Resnick SM (2012)Correspondence between in vivo (11)C-PiB-PET amy-loid imaging and postmortem, region-matched assessmentof plaques. Acta Neuropathol, doi: 10.1007/s00401-012-1025-1

[1177] Serdons K, Verduyckt T, Vanderghinste D, Borghgraef P,Cleynhens J, Van Leuven F, Kung H, Bormans G, Ver-bruggen A (2009) 11C-labelled PIB analogues as potentialtracer agents for in vivo imaging of amyloid beta inAlzheimer’s disease. Eur J Med Chem 44, 1415-1426.

[1178] Serdons K, Verbruggen A, Bormans GM (2009) Develop-ing new molecular imaging probes for PET. Methods 48,104-111.

[1179] Choi SR, Golding G, Zhuang Z, Zhang W, Lim N, HeftiF, Benedum TE, Kilbourn MR, Skovronsky D, Kung HF(2009) Preclinical properties of 18F-AV-45: A PET agentfor Abeta plaques in the brain. J Nucl Med 50, 1887-1894.

[1180] Choi SR, Schneider JA, Bennett DA, Beach TG, BedellBJ, Zehntner SP, Krautkramer MJ, Kung HF, SkovronskyDM, Hefti F, Clark CM (2012) Correlation of amyloidPET ligand florbetapir F 18 binding with abeta aggregationand neuritic plaque deposition in postmortem brain tissue.Alzheimer Dis Assoc Disord 26, 8-16.

[1181] Kung HF, Choi SR, Qu W, Zhang W, Skovronsky D (2010)18F stilbenes and styrylpyridines for PET imaging of Abeta plaques in Alzheimer’s disease: A miniperspective.J Med Chem 53, 933-941.

[1182] Lin KJ, Hsu WC, Hsiao IT, Wey SP, Jin LW, SkovronskyD, Wai YY, Chang HP, Lo CW, Yao CH, Yen TC, Kung MP(2010) Whole-body biodistribution and brain PET imagingwith [18F]AV-45, a novel amyloid imaging agent–a pilotstudy. Nucl Med Biol 37, 497-508.

[1183] Liu Y, Zhu L, Plossl K, Choi SR, Qiao H, Sun X, LiS, Zha Z, Kung HF (2010) Optimization of automatedradiosynthesis of [18F]AV-45: A new PET imaging agentfor Alzheimer’s disease. Nucl Med Biol 37, 917-925.

[1184] Okamura N, Yanai K (2010) Florbetapir (18F), a PETimaging agent that binds to amyloid plaques for the poten-tial detection of Alzheimer’s disease. IDrugs 13, 890-899.

[1185] Wong DF, Rosenberg PB, Zhou Y, Kumar A, RaymontV, Ravert HT, Dannals RF, Nandi A, Brasic JR, Ye W,Hilton J, Lyketsos C, Kung HF, Joshi AD, SkovronskyDM, Pontecorvo MJ (2010) In vivo imaging of amyloiddeposition in Alzheimer disease using the radioligand 18F-AV-45 (florbetapir [corrected] F 18). J Nucl Med 51, 913-920.

[1186] Yao CH, Lin KJ, Weng CC, Hsiao IT, Ting YS, YenTC, Jan TR, Skovronsky D, Kung MP, Wey SP (2010)GMP-compliant automated synthesis of [(18)F]AV-45(Florbetapir F 18) for imaging beta-amyloid plaques inhuman brain. Appl Radiat Isot 68, 2293-2297.

[1187] Lister-James J, Pontecorvo MJ, Clark C, Joshi AD, MintunMA, Zhang W, Lim N, Zhuang Z, Golding G, ChoiSR, Benedum TE, Kennedy P, Hefti F, Carpenter AP,Kung HF, Skovronsky DM (2011) Florbetapir f-18: Ahistopathologically validated Beta-amyloid positron emis-sion tomography imaging agent. Semin Nucl Med 41,300-304.

[1188] Carome M, Wolfe S (2011) Florbetapir-PET imaging andpostmortem beta-amyloid pathology. JAMA 305, 1857-1858.

[1189] Clark CM, Schneider JA, Bedell BJ, Beach TG, BilkerWB, Mintun MA, Pontecorvo MJ, Hefti F, Carpenter AP,Flitter ML, Krautkramer MJ, Kung HF, Coleman RE,Doraiswamy PM, Fleisher AS, Sabbagh MN, SadowskyCH, Reiman EP, Zehntner SP, Skovronsky DM (2011) Useof florbetapir-PET for imaging beta-amyloid pathology.JAMA 305, 275-283.

[1190] Fleisher AS, Chen K, Liu X, Roontiva A, Thiyyagura P,Ayutyanont N, Joshi AD, Clark CM, Mintun MA, Pon-tecorvo MJ, Doraiswamy PM, Johnson KA, SkovronskyDM, Reiman EM (2011) Using positron emission tomog-raphy and florbetapir F18 to image cortical amyloid inpatients with mild cognitive impairment or dementia dueto Alzheimer disease. Arch Neurol 68, 1404-1411.

[1191] Frisoni GB (2011) PET and 18F ligands in the diagnosisof Alzheimer’s disease. Lancet Neurol 10, 397-399.

[1192] Joshi AD, Pontecorvo MJ, Clark CM, Carpenter AP, Jen-nings DL, Sadowsky CH, Adler LP, Kovnat KD, SeibylJP, Arora A, Saha K, Burns JD, Lowrey MJ, MintunMA, Skovronsky DM (2012) Performance characteristicsof amyloid PET with florbetapir F 18 in patients with

Page 95: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 95

Alzheimer’s disease and cognitively normal subjects. JNucl Med 53, 378-384.

[1193] Saint-Aubert L, Planton M, Hannequin D, Albucher JF,Delisle MB, Payoux P, Hitzel A, Viallard G, Peran P, Cam-pion D, Laquerriere A, Barbeau EJ, Puel M, Raposo N,Chollet F, Pariente J (2012) Amyloid imaging with AV45(18F-florbetapir) in a cognitively normal AbetaPP dupli-cation carrier. J Alzheimers Dis 28, 877-883.

[1194] Sabbagh MN, Fleisher A, Chen K, Rogers J, Berk C,Reiman E, Pontecorvo M, Mintun M, Skovronsky D,Jacobson SA, Sue LI, Liebsack C, Charney AS, Cole L,Belden C, Beach TG (2011) Positron emission tomographyand neuropathologic estimates of fibrillar amyloid-beta ina patient with Down syndrome and Alzheimer disease.Arch Neurol 68, 1461-1466.

[1195] Huang KL, Lin KJ, Ho MY, Chang YJ, Chang CH, WeySP, Hsieh CJ, Yen TC, Hsiao IT, Lee TH (2012) Amy-loid deposition after cerebral hypoperfusion: Evidencedon [(18)F]AV-45 positron emission tomography. J NeurolSci 319, 124-129.

[1196] Hsiao IT, Huang CC, Hsieh CJ, Hsu WC, Wey SP, Yen TC,Kung MP, Lin KJ (2012) Correlation of early-phase 18F-florbetapir (AV-45/Amyvid) PET images to FDG images:Preliminary studies. Eur J Nucl Med Mol Imaging 39, 613-620.

[1197] Camus V, Payoux P, Barre L, Desgranges B, Voisin T,Tauber C, La JR, Tafani M, Hommet C, Chetelat G, Mon-don K, de LS, V, Cottier JP, Beaufils E, Ribeiro MJ, GissotV, Vierron E, Vercouillie J, Vellas B, Eustache F, Guil-loteau D (2012) Using PET with 18F-AV-45 (florbetapir)to quantify brain amyloid load in a clinical environment.Eur J Nucl Med Mol Imaging 39, 621-631.

[1198] Newberg AB, Arnold SE, Wintering N, Rovner BW, AlaviA (2012) Initial clinical comparison of 18F-Florbetapirand 18F-FDG PET in patients with Alzheimer disease andcontrols. J Nucl Med 53, 902-907.

[1199] Clark CM, Pontecorvo MJ, Beach TG, Bedell BJ, Cole-man RE, Doraiswamy PM, Fleisher AS, Reiman EM,Sabbagh MN, Sadowsky CH, Schneider JA, Arora A, Car-penter AP, Flitter ML, Joshi AD, Krautkramer MJ, Lu M,Mintun MA, Skovronsky DM (2012) Cerebral PET withflorbetapir compared with neuropathology at autopsy fordetection of neuritic amyloid-beta plaques: A prospectivecohort study. Lancet Neurol 11, 669-678.

[1200] Kingwell K (2012) Alzheimer disease: Florbetapir-a use-ful tool to image amyloid load and predict cognitive declinein Alzheimer disease. Nat Rev Neurol 8, 471.

[1201] Rowe CC, Ackerman U, Browne W, Mulligan R, Pike KL,O’Keefe G, Tochon-Danguy H, Chan G, Berlangieri SU,Jones G, Ckinson-Rowe KL, Kung HP, Zhang W, KungMP, Skovronsky D, Dyrks T, Holl G, Krause S, FriebeM, Lehman L, Lindemann S, Dinkelborg LM, MastersCL, Villemagne VL (2008) Imaging of amyloid beta inAlzheimer’s disease with 18F-BAY94-9172, a novel PETtracer: Proof of mechanism. Lancet Neurol 7, 129-135.

[1202] Barthel H, Gertz HJ, Dresel S, Peters O, Bartenstein P,Buerger K, Hiemeyer F, Wittemer-Rump SM, Seibyl J,Reininger C, Sabri O (2011) Cerebral amyloid-beta PETwith florbetaben (18F) in patients with Alzheimer’s dis-ease and healthy controls: A multicentre phase 2 diagnosticstudy. Lancet Neurol 10, 424-435.

[1203] Barthel H, Luthardt J, Becker G, Patt M, HammersteinE, Hartwig K, Eggers B, Sattler B, Schildan A, Hesse S,Meyer PM, Wolf H, Zimmermann T, Reischl J, RohdeB, Gertz HJ, Reininger C, Sabri O (2011) Individualized

quantification of brain beta-amyloid burden: Results ofa proof of mechanism phase 0 florbetaben PET trial inpatients with Alzheimer’s disease and healthy controls.Eur J Nucl Med Mol Imaging 38, 1702-1714.

[1204] Barthel H, Sabri O (2011) Florbetaben to trace amyloid-beta in the Alzheimer brain by means of PET. J AlzheimersDis 26(Suppl 3), 117-121.

[1205] Herholz K, Ebmeier K (2011) Clinical amyloid imagingin Alzheimer’s disease. Lancet Neurol 10, 667-670.

[1206] Vallabhajosula S (2011) Positron emission tomographyradiopharmaceuticals for imaging brain Beta-amyloid.Semin Nucl Med 41, 283-299.

[1207] Villemagne VL, Ong K, Mulligan RS, Holl G, PejoskaS, Jones G, O’Keefe G, Ackerman U, Tochon-DanguyH, Chan JG, Reininger CB, Fels L, Putz B, Rohde B,Masters CL, Rowe CC (2011) Amyloid imaging with(18)F-florbetaben in Alzheimer disease and other demen-tias. J Nucl Med 52, 1210-1217.

[1208] Villemagne VL, Okamura N, Pejoska S, Drago J, MulliganRS, Chetelat G, O’Keefe G, Jones G, Kung HF, PontecorvoM, Masters CL, Skovronsky DM, Rowe CC (2012) Differ-ential diagnosis in Alzheimer’s disease and dementia withLewy bodies via VMAT2 and amyloid imaging. Neurode-gener Dis 10, 161-165.

[1209] Wang H, Shi H, Yu H, Jiang S, Tang G (2011) Facile andrapid one-step radiosynthesis of [(18)F]BAY94-9172 witha new precursor. Nucl Med Biol 38, 121-127.

[1210] O’Keefe GJ, Saunder TH, Ng S, Ackerman U, Tochon-Danguy HJ, Chan JG, Gong S, Dyrks T, Lindemann S,Holl G, Dinkelborg L, Villemagne V, Rowe CC (2009)Radiation dosimetry of beta-amyloid tracers 11C-PiB and18F-BAY94-9172. J Nucl Med 50, 309-315.

[1211] Villemagne VL, Mulligan RS, Pejoska S, Ong K, JonesG, O’Keefe G, Chan JG, Young K, Tochon-Danguy H,Masters CL, Rowe CC (2012) Comparison of (11)C-PiBand (18)F-florbetaben for Abeta imaging in ageing andAlzheimer’s disease. Eur J Nucl Med Mol Imaging 39,983-989.

[1212] Schipke CG, Peters O, Heuser I, Grimmer T, SabbaghMN, Sabri O, Hock C, Kunz M, Kuhlmann J, ReiningerC, Blankenburg M (2012) Impact of beta-amyloid-specificflorbetaben PET imaging on confidence in early diagnosisof Alzheimer’s disease. Dement Geriatr Cogn Disord 33,416-422.

[1213] Nelissen N, Van LK, Thurfjell L, Owenius R, Vandenbul-cke M, Koole M, Bormans G, Brooks DJ, Vandenberghe R(2009) Phase 1 study of the Pittsburgh compound B deriva-tive 18F-flutemetamol in healthy volunteers and patientswith probable Alzheimer disease. J Nucl Med 50, 1251-1259.

[1214] Vandenberghe R, Van LK, Ivanoiu A, Salmon E, BastinC, Triau E, Hasselbalch S, Law I, Andersen A, Korner A,Minthon L, Garraux G, Nelissen N, Bormans G, BuckleyC, Owenius R, Thurfjell L, Farrar G, Brooks DJ (2010)18F-flutemetamol amyloid imaging in Alzheimer diseaseand mild cognitive impairment: A phase 2 trial. Ann Neurol68, 319-329.

[1215] Thomas BA, Erlandsson K, Modat M, Thurfjell L, Vanden-berghe R, Ourselin S, Hutton BF (2011) The importance ofappropriate partial volume correction for PET quantifica-tion in Alzheimer’s disease. Eur J Nucl Med Mol Imaging38, 1104-1119.

[1216] Wolk DA, Grachev ID, Buckley C, Kazi H, Grady MS,Trojanowski JQ, Hamilton RH, Sherwin P, McLainR, Arnold SE (2011) Association between in vivo

Page 96: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

96 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

fluorine 18-labeled flutemetamol amyloid positron emis-sion tomography imaging and in vivo cerebral corticalhistopathology. Arch Neurol 68, 1398-1403.

[1217] Thurfjell L, Lotjonen J, Lundqvist R, KoikkalainenJ, Soininen H, Waldemar G, Brooks DJ, Vanden-berghe R (2012) Combination of biomarkers: PET[18F]flutemetamol imaging and structural MRI in demen-tia and mild cognitive impairment. Neurodegener Dis 10,246-249.

[1218] Vandenberghe R, Nelissen N, Salmon E, Ivanoiu A, Has-selbalch S, Andersen A, Korner A, Minthon L, Brooks DJ,Van LK, Dupont P (2012) Binary classification of (18)F-flutemetamol PET using machine learning: Comparisonwith visual reads and structural MRI. Neuroimage 64C,517-525.

[1219] Snellman A, Rokka J, Lopez-Picon FR, Eskola O, WilsonI, Farrar G, Scheinin M, Solin O, Rinne JO, Haaparanta-Solin M (2012) Pharmacokinetics of [(18)F]flutemetamolin wild-type rodents and its binding to beta amyloiddeposits in a mouse model of Alzheimer’s disease. EurJ Nucl Med Mol Imaging 39, 1784-1795.

[1220] Jureus A, Swahn BM, Sandell J, Jeppsson F, JohnsonAE, Johnstrom P, Neelissen JA, Sunnemark D, Farde L,Svensson SP (2010) Characterization of AZD4694, a novelfluorinated Abeta plaque neuroimaging PET radioligand.J Neurochem 114, 784-794.

[1221] Brockschnieder D, Schmitt-Willich H, Heinrich T, VarroneA, Gulyas B, Toth M, Andersson J, Boemer U, Krause S,Friebe M, Dinkelborg L, Halldin C, Dyrks T (2012) Pre-clinical characterization of a novel class of 18F-LabeledPET tracers for amyloid-beta. J Nucl Med 53, 1794-1801.

[1222] Furukawa K, Okamura N, Tashiro M, Waragai M, Furu-moto S, Iwata R, Yanai K, Kudo Y, Arai H (2010) AmyloidPET in mild cognitive impairment and Alzheimer’s dis-ease with BF-227: Comparison to FDG-PET. J Neurol 257,721-727.

[1223] Shao H, Okamura N, Sugi K, Furumoto S, Furukawa K,Tashiro M, Iwata R, Matsuda H, Kudo Y, Arai H, Fukuda H,Yanai K (2010) Voxel-based analysis of amyloid positronemission tomography probe [C]BF-227 uptake in mildcognitive impairment and alzheimer’s disease. DementGeriatr Cogn Disord 30, 101-111.

[1224] Kaneta T, Okamura N, Minoshima S, Furukawa K, TashiroM, Furumoto S, Iwata R, Fukuda H, Takahashi S, YanaiK, Kudo Y, Arai H (2011) A modified method of 3D-SSPanalysis for amyloid PET imaging using [(11)C]BF-227.Ann Nucl Med 25, 732-739.

[1225] Tomita N, Furukawa K, Okamura N, Tashiro M, Une K,Furumoto S, Iwata R, Yanai K, Kudo Y, Arai H (2012)Brain accumulation of amyloid beta protein visualized bypositron emission tomography and BF-227 in Alzheimer’sdisease patients with or without diabetes mellitus. Geri-atr Gerontol Int, doi: 10.1111/j.1447-0594.2012.00880.x[Epub ahead of print].

[1226] Fodero-Tavoletti MT, Mulligan RS, Okamura N, FurumotoS, Rowe CC, Kudo Y, Masters CL, Cappai R, Yanai K,Villemagne VL (2009) In vitro characterisation of BF227binding to alpha-synuclein/Lewy bodies. Eur J Pharmacol617, 54-58.

[1227] Kikuchi A, Takeda A, Okamura N, Tashiro M, HasegawaT, Furumoto S, Kobayashi M, Sugeno N, Baba T, Miki Y,Mori F, Wakabayashi K, Funaki Y, Iwata R, TakahashiS, Fukuda H, Arai H, Kudo Y, Yanai K, Itoyama Y(2010) In vivo visualization of alpha-synuclein depositionby carbon-11-labelled 2-[2-(2-dimethylaminothiazol-5-

yl)ethenyl]-6-[2-(fluoro)ethoxy]benzoxazole positronemission tomography in multiple system atrophy. Brain133, 1772-1778.

[1228] Okamura N, Shiga Y, Furumoto S, Tashiro M, Tsuboi Y,Furukawa K, Yanai K, Iwata R, Arai H, Kudo Y, ItoyamaY, Doh-ura K (2010) In vivo detection of prion amyloidplaques using [(11)C]BF-227 PET. Eur J Nucl Med MolImaging 37, 934-941.

[1229] Kung MP, Hou C, Zhuang ZP, Skovronsky D, Kung HF(2004) Binding of two potential imaging agents targetingamyloid plaques in postmortem brain tissues of patientswith Alzheimer’s disease. Brain Res 1025, 98-105.

[1230] Verhoeff NP, Wilson AA, Takeshita S, Trop L, HusseyD, Singh K, Kung HF, Kung MP, Houle S (2004) In-vivo imaging of Alzheimer disease beta-amyloid with[11C]SB-13 PET. Am J Geriatr Psychiatry 12, 584-595.

[1231] Zha Z, Choi SR, Ploessl K, Lieberman BP, Qu W, Hefti F,Mintun M, Skovronsky D, Kung HF (2011) Multidentate(18)F-polypegylated styrylpyridines as imaging agents forAbeta plaques in cerebral amyloid angiopathy (CAA). JMed Chem 54, 8085-8098.

[1232] Petric A, Johnson SA, Pham HV, Li Y, Ceh S, Golobic A,Agdeppa ED, Timbol G, Liu J, Keum G, Satyamurthy N,Kepe V, Houk KN, Barrio JR (2012) Dicyanovinylnaph-thalenes for neuroimaging of amyloids and relationships ofelectronic structures and geometries to binding affinities.Proc Natl Acad Sci U S A 109, 16492-16497.

[1233] Cui M, Ono M, Kimura H, Ueda M, Nakamoto Y, TogashiK, Okamoto Y, Ihara M, Takahashi R, Liu B, Saji H (2012)Novel (18)F-labeled benzoxazole derivatives as potentialpositron emission tomography probes for imaging of cere-bral beta-amyloid plaques in Alzheimer’s disease. J MedChem 55, 9136-9145.

[1234] Cui M, Wang X, Yu P, Zhang J, Li Z, Zhang X, YangY, Ono M, Jia H, Saji H, Liu B (2012) Synthesis andevaluation of novel (18)F labeled 2-pyridinylbenzoxazoleand 2-pyridinylbenzothiazole derivatives as ligands forpositron emission tomography (PET) imaging of beta-amyloid plaques. J Med Chem 55, 9283-9296.

[1235] Johnson AE, Jeppsson F, Sandell J, Wensbo D, NeelissenJA, Jureus A, Strom P, Norman H, Farde L, Svensson SP(2009) AZD2184: A radioligand for sensitive detection ofbeta-amyloid deposits. J Neurochem 108, 1177-1186.

[1236] Nyberg S, Jonhagen ME, Cselenyi Z, Halldin C, JulinP, Olsson H, Freund-Levi Y, Andersson J, Varnas K,Svensson S, Farde L (2009) Detection of amyloid inAlzheimer’s disease with positron emission tomographyusing [11C]AZD2184. Eur J Nucl Med Mol Imaging 36,1859-1863.

[1237] Andersson JD, Varnas K, Cselenyi Z, Gulyas B, WensboD, Finnema SJ, Swahn BM, Svensson S, Nyberg S,Farde L, Halldin C (2010) Radiosynthesis of the candi-date beta-amyloid radioligand [(11)C]AZD2184: Positronemission tomography examination and metabolite analysisin cynomolgus monkeys. Synapse 64, 733-741.

[1238] Shoghi-Jadid K, Small GW, Agdeppa ED, Kepe V, ErcoliLM, Siddarth P, Read S, Satyamurthy N, Petric A, HuangSC, Barrio JR (2002) Localization of neurofibrillary tan-gles and beta-amyloid plaques in the brains of livingpatients with Alzheimer disease. Am J Geriatr Psychiatry10, 24-35.

[1239] Ossenkoppele R, Tolboom N, Foster-Dingley JC, Adri-aanse SF, Boellaard R, Yaqub M, Windhorst AD,Barkhof F, Lammertsma AA, Scheltens P, van der FlierWM, van Berckel BN (2012) Longitudinal imaging of

Page 97: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 97

Alzheimer pathology using [(11)C]PIB, [(18)F]FDDNPand [(18)F]FDG PET. Eur J Nucl Med Mol Imaging 39,990-1000.

[1240] Protas HD, Kepe V, Hayashi KM, Klunder AD, BraskieMN, Ercoli L, Siddarth P, Bookheimer SY, Thompson PM,Small GW, Barrio JR, Huang SC (2012) Prediction of cog-nitive decline based on hemispheric cortical surface mapsof FDDNP PET. Neuroimage 61, 749-760.

[1241] Cai L, Cuevas J, Temme S, Herman MM, Dagostin C,Widdowson DA, Innis RB, Pike VW (2007) Synthesisand structure-affinity relationships of new 4-(6-iodo-H-imidazo[1,2-a]pyridin-2-yl)-N-dimethylbenzeneaminederivatives as ligands for human beta-amyloid plaques. JMed Chem 50, 4746-4758.

[1242] Ryu EK, Chen X (2008) Development of Alzheimer’s dis-ease imaging agents for clinical studies. Front Biosci 13,777-789.

[1243] Zhuang ZP, Kung MP, Wilson A, Lee CW, Plossl K,Hou C, Holtzman DM, Kung HF (2003) Structure-activityrelationship of imidazo[1,2-a]pyridines as ligands fordetecting beta-amyloid plaques in the brain. J Med Chem46, 237-243.

[1244] Newberg AB, Wintering NA, Plossl K, Hochold J, StabinMG, Watson M, Skovronsky D, Clark CM, Kung MP,Kung HF (2006) Safety, biodistribution, and dosimetry of123I-IMPY: A novel amyloid plaque-imaging agent for thediagnosis of Alzheimer’s disease. J Nucl Med 47, 748-754.

[1245] Pepys MB, Herbert J, Hutchinson WL, Tennent GA, Lach-mann HJ, Gallimore JR, Lovat LB, Bartfai T, Alanine A,Hertel C, Hoffmann T, Jakob-Roetne R, Norcross RD,Kemp JA, Yamamura K, Suzuki M, Taylor GW, MurrayS, Thompson D, Purvis A, Kolstoe S, Wood SP, HawkinsPN (2002) Targeted pharmacological depletion of serumamyloid P component for treatment of human amyloidosis.Nature 417, 254-259.

[1246] Kolstoe SE, Wood SP (2010) Drug targets for amyloidosis.Biochem Soc Trans 38, 466-470.

[1247] Millar DJ, Hutchinson WL, Pepys MB (2011) Immunora-diometric assay for human serum amyloid P component.J Immunol Methods 371, 18-24.

[1248] Kolstoe SE, Ridha BH, Bellotti V, Wang N, RobinsonCV, Crutch SJ, Keir G, Kukkastenvehmas R, GallimoreJR, Hutchinson WL, Hawkins PN, Wood SP, Rossor MN,Pepys MB (2009) Molecular dissection of Alzheimer’sdisease neuropathology by depletion of serum amyloid Pcomponent. Proc Natl Acad Sci U S A 106, 7619-7623.

[1249] Bodin K, Ellmerich S, Kahan MC, Tennent GA, LoeschA, Gilbertson JA, Hutchinson WL, Mangione PP, Gal-limore JR, Millar DJ, Minogue S, Dhillon AP, Taylor GW,Bradwell AR, Petrie A, Gillmore JD, Bellotti V, BottoM, Hawkins PN, Pepys MB (2010) Antibodies to humanserum amyloid P component eliminate visceral amyloiddeposits. Nature 468, 93-97.

[1250] Gillmore JD, Tennent GA, Hutchinson WL, GallimoreJR, Lachmann HJ, Goodman HJ, Offer M, Millar DJ,Petrie A, Hawkins PN, Pepys MB (2010) Sustainedpharmacological depletion of serum amyloid P componentin patients with systemic amyloidosis. Br J Haematol 148,760-767.

[1251] Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H,Guido T, Hu K, Huang J, Johnson-Wood K, Khan K,Kholodenko D, Lee M, Liao Z, Lieberburg I, Motter R,Mutter L, Soriano F, Shopp G, Vasquez N, VandevertC, Walker S, Wogulis M, Yednock T, Games D, Seu-bert P (1999) Immunization with amyloid-beta attenuates

Alzheimer-disease-like pathology in the PDAPP mouse.Nature 400, 173-177.

[1252] Schenk D (2002) Amyloid-beta immunotherapy forAlzheimer’s disease: The end of the beginning. Nat RevNeurosci 3, 824-828.

[1253] Orgogozo JM, Gilman S, Dartigues JF, Laurent B, PuelM, Kirby LC, Jouanny P, Dubois B, Eisner L, Flitman S,Michel BF, Boada M, Frank A, Hock C (2003) Subacutemeningoencephalitis in a subset of patients with AD afterAbeta42 immunization. Neurology 61, 46-54.

[1254] Hock C, Konietzko U, Papassotiropoulos A, Wollmer A,Streffer J, von Rotz RC, Davey G, Moritz E, Nitsch RM(2002) Generation of antibodies specific for beta-amyloidby vaccination of patients with Alzheimer disease. NatMed 8, 1270-1275.

[1255] Hock C, Konietzko U, Streffer JR, Tracy J, Signorell A,Muller-Tillmanns B, Lemke U, Henke K, Moritz E, GarciaE, Wollmer MA, Umbricht D, de Quervain DJ, HofmannM, Maddalena A, Papassotiropoulos A, Nitsch RM (2003)Antibodies against beta-amyloid slow cognitive decline inAlzheimer’s disease. Neuron 38, 547-554.

[1256] Imbimbo BP (2002) Toxicity of beta-amyloid vaccinationin patients with Alzheimer’s disease. Ann Neurol 51, 794.

[1257] Imbimbo BP (2002) �-Amyloid immunization approachesfor Alzheimer’s disease. Drug Dev Res 56, 150-162.

[1258] Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H,Weller RO (2003) Neuropathology of human Alzheimerdisease after immunization with amyloid-beta peptide: Acase report. Nat Med 9, 448-452.

[1259] Nicoll JA, Barton E, Boche D, Neal JW, Ferrer I, Thomp-son P, Vlachouli C, Wilkinson D, Bayer A, Games D,Seubert P, Schenk D, Holmes C (2006) Abeta speciesremoval after abeta42 immunization. J Neuropathol ExpNeurol 65, 1040-1048.

[1260] Robinson SR, Bishop GM, Lee HG, Munch G (2004)Lessons from the AN 1792 Alzheimer vaccine: Lest weforget. Neurobiol Aging 25, 609-615.

[1261] Bayer AJ, Bullock R, Jones RW, Wilkinson D, PatersonKR, Jenkins L, Millais SB, Donoghue S (2005) Evaluationof the safety and immunogenicity of synthetic Abeta42(AN1792) in patients with AD. Neurology 64, 94-101.

[1262] Boche D, Nicoll JA, Weller RO (2005) Immunotherapyfor Alzheimer’s disease and other dementias. Curr OpinNeurol 18, 720-725.

[1263] Boche D, Zotova E, Weller RO, Love S, Neal JW, Pick-ering RM, Wilkinson D, Holmes C, Nicoll JA (2008)Consequence of Abeta immunization on the vasculature ofhuman Alzheimer’s disease brain. Brain 131, 3299-3310.

[1264] Boche D, Donald J, Love S, Harris S, Neal JW, HolmesC, Nicoll JA (2010) Reduction of aggregated Tau in neu-ronal processes but not in the cell bodies after Abeta42immunisation in Alzheimer’s disease. Acta Neuropathol120, 13-20.

[1265] Boche D, Denham N, Holmes C, Nicoll JA (2010)Neuropathology after active Abeta42 immunotherapy:Implications for Alzheimer’s disease pathogenesis. ActaNeuropathol 120, 369-384.

[1266] Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, FoxNC, Eisner L, Kirby L, Rovira MB, Forette F, Orgogozo JM(2005) Clinical effects of Abeta immunization (AN1792)in patients with AD in an interrupted trial. Neurology 64,1553-1562.

[1267] Hock C, Nitsch RM (2005) Clinical observations with AN-1792 using TAPIR analyses. Neurodegener Dis 2, 273-276.

Page 98: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

98 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1268] Lee M, Bard F, Johnson-Wood K, Lee C, Hu K, Griffith SG,Black RS, Schenk D, Seubert P (2005) Abeta42 immuniza-tion in Alzheimer’s disease generates Abeta N-terminalantibodies. Ann Neurol 58, 430-435.

[1269] Masliah E, Hansen L, Adame A, Crews L, Bard F, Lee C,Seubert P, Games D, Kirby L, Schenk D (2005) Abetavaccination effects on plaque pathology in the absenceof encephalitis in Alzheimer disease. Neurology 64,129-131.

[1270] O’Toole M, Janszen DB, Slonim DK, Reddy PS, Ellis DK,Legault HM, Hill AA, Whitley MZ, Mounts WM, ZuberekK, Immermann FW, Black RS, Dorner AJ (2005) Risk fac-tors associated with beta-amyloid(1-42) immunotherapy inpreimmunization gene expression patterns of blood cells.Arch Neurol 62, 1531-1536.

[1271] Schenk DB, Seubert P, Grundman M, Black R (2005)A beta immunotherapy: Lessons learned for potentialtreatment of Alzheimer’s disease. Neurodegener Dis 2,255-260.

[1272] Patton RL, Kalback WM, Esh CL, Kokjohn TA, Van VickleGD, Luehrs DC, Kuo YM, Lopez J, Brune D, Ferrer I,Masliah E, Newel AJ, Beach TG, Castano EM, RoherAE (2006) Amyloid-beta peptide remnants in AN-1792-immunized Alzheimer’s disease patients: A biochemicalanalysis. Am J Pathol 169, 1048-1063.

[1273] Vasilevko V, Cribbs DH (2006) Novel approaches forimmunotherapeutic intervention in Alzheimer’s disease.Neurochem Int 49, 113-126.

[1274] Bombois S, Maurage CA, Gompel M, Deramecourt V,kowiak-Cordoliani MA, Black RS, Lavielle R, DelacourteA, Pasquier F (2007) Absence of beta-amyloid depositsafter immunization in Alzheimer disease with Lewy bodydementia. Arch Neurol 64, 583-587.

[1275] Koepsell TD, Chi YY, Zhou XH, Lee WW, Ramos EM,Kukull WA (2007) An alternative method for estimatingefficacy of the AN1792 vaccine for Alzheimer disease.Neurology 69, 1868-1872.

[1276] Hawkes CA, McLaurin J (2008) Clinical immunotherapytrials in Alzheimer’s disease. Drug Discovery Today: Ther-apeutic Strategies 5, 177-183.

[1277] Holmes C, Boche D, Wilkinson D, Yadegarfar G, HopkinsV, Bayer A, Jones RW, Bullock R, Love S, Neal JW, ZotovaE, Nicoll JA (2008) Long-term effects of Abeta42 immuni-sation in Alzheimer’s disease: Follow-up of a randomised,placebo-controlled phase I trial. Lancet 372, 216-223.

[1278] Holtzman DM (2008) Alzheimer’s disease: Movingtowards a vaccine. Nature 454, 418-420.

[1279] Pride M, Seubert P, Grundman M, Hagen M, Eldridge J,Black RS (2008) Progress in the active immunotherapeuticapproach to Alzheimer’s disease: Clinical investigationsinto AN1792-associated meningoencephalitis. Neurode-gener Dis 5, 194-196.

[1280] St George-Hyslop PH, Morris JC (2008) Will anti-amyloidtherapies work for Alzheimer’s disease? Lancet 372, 180-182.

[1281] Kokjohn TA, Roher AE (2009) Antibody responses,amyloid-beta peptide remnants and clinical effects of AN-1792 immunization in patients with AD in an interruptedtrial. CNS Neurol Disord Drug Targets 8, 88-97.

[1282] Town T (2009) Alternative Abeta immunotherapyapproaches for Alzheimer’s disease. CNS Neurol DisordDrug Targets 8, 114-127.

[1283] Vellas B, Black R, Thal LJ, Fox NC, Daniels M, McLen-nan G, Tompkins C, Leibman C, Pomfret M, Grundman M(2009) Long-term follow-up of patients immunized with

AN1792: Reduced functional decline in antibody respon-ders. Curr Alzheimer Res 6, 144-151.

[1284] Serrano-Pozo A, William CM, Ferrer I, Uro-Coste E,Delisle MB, Maurage CA, Hock C, Nitsch RM, MasliahE, Growdon JH, Frosch MP, Hyman BT (2010) Beneficialeffect of human anti-amyloid-beta active immunization onneurite morphology and tau pathology. Brain 133, 1312-1327.

[1285] Uro-Coste E, Russano de PG, Guilbeau-Frugier C, SastreN, Ousset PJ, da Silva NA, Lavialle-Guillotreau V, VellasB, Delisle MB (2010) Cerebral amyloid angiopathy andmicrohemorrhages after amyloid beta vaccination: Casereport and brief review. Clin Neuropathol 29, 209-216.

[1286] Zotova E, Holmes C, Johnston D, Neal JW, NicollJA, Boche D (2011) Microglial alterations in humanAlzheimer’s disease following Abeta42 immunization.Neuropathol Appl Neurobiol 37, 513-524.

[1287] Schneeberger A, Mandler M, Otawa O, Zauner W, Mat-tner F, Schmidt W (2009) Development of AFFITOPEvaccines for Alzheimer’s disease (AD)–from concept toclinical testing. J Nutr Health Aging 13, 264-267.

[1288] Schneeberger A, Mandler M, Mattner F, Schmidt W(2010) AFFITOME(R) technology in neurodegenerativediseases: The doubling advantage. Hum Vaccin 6, 948-952.

[1289] Winblad B, Andreasen N, Minthon L, Floesser A,Imbert G, Dumortier T, Maguire RP, Blennow K,Lundmark J, Staufenbiel M, Orgogozo JM, Graf A(2012) Safety, tolerability, and antibody response ofactive Abeta immunotherapy with CAD106 in patientswith Alzheimer’s disease: Randomised, double-blind,placebo-controlled, first-in-human study. Lancet Neurol11, 597-604.

[1290] Wisniewski T (2012) Active immunotherapy forAlzheimer’s disease. Lancet Neurol 11, 571-572.

[1291] Kingwell K (2012) Alzheimer disease: Amyloid-betaimmunotherapy CAD106 passes first safety test in patientswith Alzheimer disease. Nat Rev Neurol 8, 414.

[1292] Dyer MR, Renner WA, Bachmann MF (2006) A secondvaccine revolution for the new epidemics of the 21st cen-tury. Drug Discov Today 11, 1028-1033.

[1293] Jennings GT, Bachmann MF (2008) The coming of age ofvirus-like particle vaccines. Biol Chem 389, 521-536.

[1294] Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S,Jacobson LH, Jennings GT, Luond R, Ortmann R, Reich-wald J, Zurini M, Mir A, Bachmann MF, Staufenbiel M(2011) The second-generation active Abeta immunother-apy CAD106 reduces amyloid accumulation in APPtransgenic mice while minimizing potential side effects.J Neurosci 31, 9323-9331.

[1295] Ryan JM, Grundman M (2009) Anti-amyloid-betaimmunotherapy in Alzheimer’s disease: ACC-001 clinicaltrials are ongoing. J Alzheimers Dis 17, 243.

[1296] Muhs A, Hickman DT, Pihlgren M, Chuard N, GiriensV, Meerschman C, Van der Auwera I, Van Leuven F,Sugawara M, Weingertner MC, Bechinger B, GreferathR, Kolonko N, Nagel-Steger L, Riesner D, Brady RO,Pfeifer A, Nicolau C (2007) Liposomal vaccines withconformation-specific amyloid peptide antigens defineimmune response and efficacy in APP transgenic mice.Proc Natl Acad Sci U S A 104, 9810-9815.

[1297] Hickman DT, Lopez-Deber MP, Ndao DM, Silva AB,Nand D, Pihlgren M, Giriens V, Madani R, St-Pierre A,Karastaneva H, Nagel-Steger L, Willbold D, Riesner D,Nicolau C, Baldus M, Pfeifer A, Muhs A (2011) Sequence-independent control of peptide conformation in liposomal

Page 99: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 99

vaccines for targeting protein misfolding diseases. J BiolChem 286, 13966-13976.

[1298] Wang CY, Finstad CL, Walfield AM, Sia C, Sokoll KK,Chang TY, Fang XD, Hung CH, Hutter-Paier B, WindischM (2007) Site-specific UBITh amyloid-beta vaccine forimmunotherapy of Alzheimer’s disease. Vaccine 25, 3041-3052.

[1299] Savage MJ, Wu G, McCampbell A, Wessner KR, Cit-ron M, Liang X, Hsieh S, Wolfe AL, Kinney GG, RosenLB, Renger JJ (2010) A novel multivariant Abetapeptidevaccine with preclinical evidence of a central immuneresponse that generates antisera recognizing a wide rangeof Abeta peptide species. Alzheimers Dement 6, S142.

[1300] Davtyan H, Ghochikyan A, Movsesyan N, Ellefsen B,Petrushina I, Cribbs DH, Hannaman D, Evans CF, Agad-janyan MG (2012) Delivery of a DNA vaccine forAlzheimer’s disease by electroporation versus gene gungenerates potent and similar immune responses. Neurode-gener Dis 10, 261-264.

[1301] Xing XN, Zhang WG, Sha S, Li Y, Guo R, Wang C,Cao YP (2011) Amyloid beta 3-10 DNA vaccination sug-gests a potential new treatment for Alzheimer’s disease inBALB/c mice. Chin Med J (Engl) 124, 2636-2641.

[1302] Xing X, Sha S, Li Y, Zong L, Jiang T, Cao Y (2012) Immu-nization with a new DNA vaccine for Alzheimer’s diseaseelicited Th2 immune response in BALB/c mice by in vivoelectroporation. J Neurol Sci 313, 17-21.

[1303] Ma Y, Li Y, Zong LX, Xing XN, Zhang WG, CaoYP (2011) Improving memory and decreasing cognitiveimpairment in Tg-APPswe/PSEN1dE9 mice with Abeta3-10 repeat fragment plasmid by reducing Abeta depositionand inflammatory response. Brain Res 1400, 112-124.

[1304] Li Y, Ma Y, Zong LX, Xing XN, Sha S, Cao YP (2011)Intranasal inoculation with an adenovirus vaccine encod-ing ten repeats of Abeta3-10 induces Th2 immune responseagainst amyloid-beta in wild-type mouse. Neurosci Lett505, 128-133.

[1305] Li Y, Ma Y, Zong LX, Xing XN, Guo R, Jiang TZ, Sha S,Liu L, Cao YP (2012) Intranasal inoculation with an aden-ovirus vaccine encoding ten repeats of Abeta3-10 reducesAD-like pathology and cognitive impairment in Tg-APPswe/PSEN1dE9 mice. J Neuroimmunol 249, 16-26.

[1306] Santuccione AC, Merlini M, Shetty A, Tackenberg C, BaliJ, Ferretti MT, McAfoose J, Kulic L, Bernreuther C, WeltT, Grimm J, Glatzel M, Rajendran L, Hock C, NitschRM (2012) Active vaccination with ankyrin G reducesbeta-amyloid pathology in APP transgenic mice. MolPsychiatry, doi: 10.1038/mp.2012.70 [Epub ahead ofprint].

[1307] Butovsky O, Koronyo-Hamaoui M, Kunis G, Ophir E,Landa G, Cohen H, Schwartz M (2006) Glatiramer acetatefights against Alzheimer’s disease by inducing dendritic-like microglia expressing insulin-like growth factor 1. ProcNatl Acad Sci U S A 103, 11784-11789.

[1308] Hara H, Inoue M, Adachi K, Yonemitsu Y, Hasegawa M,Nabeshima T, Tabira T (2007) [Mucosal immunotherapyfor alzheimer’s disease with viral vectors]. Nihon ShinkeiSeishin Yakurigaku Zasshi 27, 53-56.

[1309] Janus C, Pearson J, McLaurin J, Mathews PM, Jiang Y,Schmidt SD, Chishti MA, Horne P, Heslin D, French J,Mount HT, Nixon RA, Mercken M, Bergeron C, FraserPE, St George-Hyslop P, Westaway D (2000) A beta pep-tide immunization reduces behavioural impairment andplaques in a model of Alzheimer’s disease. Nature 408,979-982.

[1310] Morgan D, Diamond DM, Gottschall PE, Ugen KE, DickeyC, Hardy J, Duff K, Jantzen P, DiCarlo G, Wilcock D,Connor K, Hatcher J, Hope C, Gordon M, Arendash GW(2000) A beta peptide vaccination prevents memory lossin an animal model of Alzheimer’s disease. Nature 408,982-985.

[1311] Sigurdsson EM, Scholtzova H, Mehta PD, Fran-gione B, Wisniewski T (2001) Immunization with anontoxic/nonfibrillar amyloid-beta homologous peptidereduces Alzheimer’s disease-associated pathology intransgenic mice. Am J Pathol 159, 439-447.

[1312] Qu B, Rosenberg RN, Li L, Boyer PJ, Johnston SA (2004)Gene vaccination to bias the immune response to amyloid-beta peptide as therapy for Alzheimer disease. Arch Neurol61, 1859-1864.

[1313] Agadjanyan MG, Ghochikyan A, Petrushina I, VasilevkoV, Movsesyan N, Mkrtichyan M, Saing T, Cribbs DH(2005) Prototype Alzheimer’s disease vaccine using theimmunodominant B cell epitope from beta-amyloid andpromiscuous T cell epitope pan HLA DR-binding peptide.J Immunol 174, 1580-1586.

[1314] Asuni AA, Boutajangout A, Scholtzova H, Knudsen E, LiYS, Quartermain D, Frangione B, Wisniewski T, Sigurds-son EM (2006) Vaccination of Alzheimer’s model micewith Abeta derivative in alum adjuvant reduces Abetaburden without microhemorrhages. Eur J Neurosci 24,2530-2542.

[1315] Lemere CA, Maier M, Jiang L, Peng Y, Seabrook TJ (2006)Amyloid-beta immunotherapy for the prevention and treat-ment of Alzheimer disease: Lessons from mice, monkeys,and humans. Rejuvenation Res 9, 77-84.

[1316] Maier M, Seabrook TJ, Lazo ND, Jiang L, Das P, Janus C,Lemere CA (2006) Short amyloid-beta (Abeta) immuno-gens reduce cerebral Abeta load and learning deficits inan Alzheimer’s disease mouse model in the absence of anAbeta-specific cellular immune response. J Neurosci 26,4717-4728.

[1317] Seabrook TJ, Jiang L, Thomas K, Lemere CA (2006)Boosting with intranasal dendrimeric Abeta1-15 but notAbeta1-15 peptide leads to an effective immune responsefollowing a single injection of Abeta1-40/42 in APP-tgmice. J Neuroinflammation 3, 14.

[1318] Seabrook TJ, Thomas K, Jiang L, Bloom J, SpoonerE, Maier M, Bitan G, Lemere CA (2007) DendrimericAbeta1-15 is an effective immunogen in wildtype andAPP-tg mice. Neurobiol Aging 28, 813-823.

[1319] Lemere CA, Maier M, Peng Y, Jiang L, Seabrook TJ(2007) Novel Abeta immunogens: Is shorter better? CurrAlzheimer Res 4, 427-436.

[1320] Mouri A, Noda Y, Hara H, Mizoguchi H, Tabira T,Nabeshima T (2007) Oral vaccination with a viral vec-tor containing Abeta cDNA attenuates age-related Abetaaccumulation and memory deficits without causing inflam-mation in a mouse Alzheimer model. FASEB J 21,2135-2148.

[1321] Nikolic WV, Bai Y, Obregon D, Hou H, Mori T, ZengJ, Ehrhart J, Shytle RD, Giunta B, Morgan D, Town T,Tan J (2007) Transcutaneous beta-amyloid immunizationreduces cerebral beta-amyloid deposits without T cell infil-tration and microhemorrhage. Proc Natl Acad Sci U S A104, 2507-2512.

[1322] Petrushina I, Ghochikyan A, Mktrichyan M, MamikonyanG, Movsesyan N, Davtyan H, Patel A, Head E, Cribbs DH,Agadjanyan MG (2007) Alzheimer’s disease peptide epi-tope vaccine reduces insoluble but not soluble/oligomeric

Page 100: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

100 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Abeta species in amyloid precursor protein transgenicmice. J Neurosci 27, 12721-12731.

[1323] Petrushina I, Ghochikyan A, Mkrtichyan M, MamikonyanG, Movsesyan N, Ajdari R, Vasilevko V, Karapetyan A,Lees A, Agadjanyan MG, Cribbs DH. (2008) Mannan-Abeta28 conjugate prevents Abeta-plaque deposition, butincreases microhemorrhages in the brains of vaccinatedTg2576 (APPsw) mice. J Neuroinflammation 5, 42

[1324] Movsesyan N, Mkrtichyan M, Petrushina I, Ross TM,Cribbs DH, Agadjanyan MG, Ghochikyan A (2008)DNA epitope vaccine containing complement componentC3d enhances anti-amyloid-beta antibody production andpolarizes the immune response towards a Th2 phenotype.J Neuroimmunol 205, 57-63.

[1325] Boutajangout A, Goni F, Knudsen E, Schreiber F, Asuni A,Quartermain D, Frangione B, Chabalgoity A, WisniewskiT, Sigurdsson EM (2009) Diminished amyloid-beta burdenin Tg2576 mice following a prophylactic oral immu-nization with a salmonella-based amyloid-beta derivativevaccine. J Alzheimers Dis 18, 961-972.

[1326] Kitazawa M, Vasilevko V, Cribbs DH, LaFerla FM (2009)Immunization with amyloid-beta attenuates inclusionbody myositis-like myopathology and motor impairmentin a transgenic mouse model. J Neurosci 29, 6132-6141.

[1327] Trouche SG, Asuni A, Rouland S, Wisniewski T, Fran-gione B, Verdier JM, Sigurdsson EM, Mestre-Frances N(2009) Antibody response and plasma Abeta1-40 levelsin young Microcebus murinus primates immunized withAbeta1-42 and its derivatives. Vaccine 27, 957-964.

[1328] Wilcock DM, Gharkholonarehe N, Van Nostrand WE,Davis J, Vitek MP, Colton CA (2009) Amyloid reduc-tion by amyloid-beta vaccination also reduces mouse taupathology and protects from neuron loss in two mousemodels of Alzheimer’s disease. J Neurosci 29, 7957-7965.

[1329] Ishii-Katsuno R, Nakajima A, Katsuno T, Nojima J,Futai E, Sasagawa N, Yoshida T, Watanabe Y, Ishiura S(2010) Reduction of amyloid beta-peptide accumulationin Tg2576 transgenic mice by oral vaccination. BiochemBiophys Res Commun 399, 593-599.

[1330] Vasilevko V, Pop V, Kim HJ, Saing T, Glabe CC, Milton S,Barrett EG, Cotman CW, Cribbs DH, Head E (2010) Linearand conformation specific antibodies in aged beagles afterprolonged vaccination with aggregated Abeta. NeurobiolDis 39, 301-310.

[1331] Vollmar P, Kullmann JS, Thilo B, Claussen MC, Rothham-mer V, Jacobi H, Sellner J, Nessler S, Korn T, HemmerB (2010) Active immunization with amyloid-beta 1-42impairs memory performance through TLR2/4-dependentactivation of the innate immune system. J Immunol 185,6338-6347.

[1332] Wang CM, deVries S, Camboni M, Glass M, Martin PT(2010) Immunization with the SDPM1 peptide lowersamyloid plaque burden and improves cognitive functionin the APPswePSEN1(A246E) transgenic mouse modelof Alzheimer’s disease. Neurobiol Dis 39, 409-422.

[1333] Mantile F, Basile C, Cicatiello V, De FD, Caivano A, DeBP, Prisco A (2011) A multimeric immunogen for theinduction of immune memory to beta-amyloid. ImmunolCell Biol 89, 604-609.

[1334] Axelsen TV, Holm A, Christiansen G, Birkelund S (2011)Identification of the shortest Abeta-peptide generatinghighly specific antibodies against the C-terminal end ofamyloid-beta42. Vaccine 29, 3260-3269.

[1335] Kofler J, Lopresti B, Janssen C, Trichel AM, Masliah E,Finn OJ, Salter RD, Murdoch GH, Mathis CA, Wiley CA(2012) Preventive immunization of aged and juvenile non-human primates to beta-amyloid. J Neuroinflammation 9,84.

[1336] Esiri MM (2001) Is an effective immune intervention forAlzheimer’s disease in prospect? Trends Pharmacol Sci22, 2-3.

[1337] Ingram DK (2001) Vaccine development for Alzheimer’sdisease: A shot of good news. Trends Neurosci 24, 305-307.

[1338] Sigurdsson EM, Wisniewski T, Frangione B (2002) Asafer vaccine for Alzheimer’s disease? Neurobiol Aging23, 1001-1008.

[1339] Sigurdsson EM, Frangione B, Wisniewski T (2002) Immu-nization for Alzheimer’s disease. Drug Develop Res 56,135-142.

[1340] Solomon B (2002) Towards Alzheimer’s disease vaccina-tion. Mini Rev Med Chem 2, 85-92.

[1341] Wisniewski T, Sigurdsson EM (2002) Immunization treat-ment approaches in Alzheimer’s and prion diseases. CurrNeurol Neurosci Rep 2, 400-404.

[1342] Dodart JC, Bales KR, Paul SM (2003) Immunotherapy forAlzheimer’s disease: Will vaccination work? Trends MolMed 9, 85-87.

[1343] Broytman O, Malter JS (2004) Anti-Abeta: The good, thebad, and the unforeseen. J Neurosci Res 75, 301-306.

[1344] Brown ME, Dasilva KA, McLaurin J (2005) Refining anAlzheimer’s vaccine to avoid an inflammatory response.Expert Opin Biol Ther 5, 809-816.

[1345] Goni F, Sigurdsson EM (2005) New directions towardssafer and effective vaccines for Alzheimer’s disease. CurrOpin Mol Ther 7, 17-23.

[1346] Rosenberg RN (2005) Immunotherapy for Alzheimer dis-ease: The promise and the problem. Arch Neurol 62,1506-1507.

[1347] Dasilva KA, Aubert I, McLaurin J (2006) Vaccine devel-opment for Alzheimer’s disease. Curr Pharm Des 12,4283-4293.

[1348] Sigurdsson EM (2006) Immunotherapy for conformationaldiseases. Curr Pharm Des 12, 2569-2585.

[1349] Solomon B (2006) Alzheimer’s disease immunotherapy:From in vitro amyloid immunomodulation to in vivo vac-cination. J Alzheimers Dis 9, 433-438.

[1350] Weiner HL, Frenkel D (2006) Immunology andimmunotherapy of Alzheimer’s disease. Nat Rev Immunol6, 404-416.

[1351] Mohajeri MH (2007) The underestimated potential ofthe immune system in prevention of Alzheimer’s diseasepathology. Bioessays 29, 927-932.

[1352] Purcell AW, McCluskey J, Rossjohn J (2007) More thanone reason to rethink the use of peptides in vaccine design.Nat Rev Drug Discov 6, 404-414.

[1353] Solomon B (2007) Beta-amyloidbased immunotherapy asa treatment of Alzheimers disease. Drugs Today (Barc) 43,333-342.

[1354] Solomon B (2007) Clinical immunologic approaches forthe treatment of Alzheimer’s disease. Expert Opin InvestigDrugs 16, 819-828.

[1355] Solomon B (2007) Active immunization againstAlzheimer’s beta-amyloid peptide using phage displaytechnology. Vaccine 25, 3053-3056.

[1356] Brody DL, Holtzman DM (2008) Active and passiveimmunotherapy for neurodegenerative disorders. AnnuRev Neurosci 31, 175-193.

Page 101: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 101

[1357] Nitsch RM, Hock C (2008) Targeting beta-amyloid pathol-ogy in Alzheimer’s disease with Abeta immunotherapy.Neurotherapeutics 5, 415-420.

[1358] Popovich PG, Longbrake EE (2008) Can the immune sys-tem be harnessed to repair the CNS? Nat Rev Neurosci 9,481-493.

[1359] Solomon B (2008) Immunological approaches foramyloid-beta clearance toward treatment for Alzheimer’sdisease. Rejuvenation Res 11, 349-357.

[1360] Agadjanyan MG, Cribbs DH (2009) Active and passiveAbeta-immunotherapy: Preclinical and clinical studies andfuture directions: Part I. CNS Neurol Disord Drug Targets8, 1-6.

[1361] Cribbs DH, Agadjanyan MG (2009) Active and passiveAbeta-immunotherapy: Preclinical and clinical studies andfuture directions: Part II. CNS Neurol Disord Drug Targets8, 82-87.

[1362] Foster JK, Verdile G, Bates KA, Martins RN (2009) Immu-nization in Alzheimer’s disease: Naive hope or realisticclinical potential? Mol Psychiatry 14, 239-251.

[1363] Solomon B (2009) Immunotherapeutic strategies forAlzheimer’s disease treatment. ScientificWorldJournal 9,909-919.

[1364] Cribbs DH (2010) Abeta DNA vaccination for Alzheimer’sdisease: Focus on disease prevention. CNS Neurol DisordDrug Targets 9, 207-216.

[1365] Lemere CA, Masliah E (2010) Can Alzheimer diseasebe prevented by amyloid-beta immunotherapy? Nat RevNeurol 6, 108-119.

[1366] Maarouf CL, Daugs ID, Kokjohn TA, Kalback WM, PattonRL, Luehrs DC, Masliah E, Nicoll JA, Sabbagh MN, BeachTG, Castano EM, Roher AE (2010) The biochemical after-math of anti-amyloid immunotherapy. Mol Neurodegener5, 39.

[1367] Solomon B, Frenkel D (2010) Immunotherapy forAlzheimer’s disease. Neuropharmacology 59, 303-309.

[1368] von Bernhardi R (2010) Immunotherapy in Alzheimer’sdisease: Where do we stand? Where should we go? JAlzheimers Dis 19, 405-421.

[1369] Wang YJ, Zhou HD, Zhou XF (2010) Modifiedimmunotherapies against Alzheimer’s disease: Towardsafer and effective amyloid clearance. J Alzheimers Dis21, 1065-1075.

[1370] Wisniewski T, Boutajangout A (2010) Vaccination as atherapeutic approach to Alzheimer’s disease. Mt Sinai JMed 77, 17-31.

[1371] Menendez-Gonzalez M, Perez-Pinera P, Martinez-RiveraM, Muniz AL, Vega JA (2011) Immunotherapy forAlzheimer’s disease: Rational basis in ongoing clinicaltrials. Curr Pharm Des 17, 508-520.

[1372] Morgan D (2011) Immunotherapy for Alzheimer’s disease.J Internal Med 269, 54-63.

[1373] Schnabel J (2011) Vaccines: Chasing the dream. Nature475, S18-S19.

[1374] Shah S, Federoff HJ (2011) Therapeutic potential of vac-cines for Alzheimer’s disease. Immunotherapy 3, 287-298.

[1375] Delrieu J, Ousset PJ, Caillaud C, Vellas B (2012)‘Clinical trials in Alzheimer’s disease’: Immunotherapyapproaches. J Neurochem 120(Suppl 1), 186-193.

[1376] Lobello K, Ryan JM, Liu E, Rippon G, Black R(2012) Targeting Beta amyloid: A clinical review ofimmunotherapeutic approaches in Alzheimer’s disease. IntJ Alzheimers Dis 2012, 628070.

[1377] Bach P, Tschape JA, Kopietz F, Braun G, Baade JK,Wiederhold KH, Staufenbiel M, Prinz M, Deller T, Kalinke

U, Buchholz CJ, Muller UC (2009) Vaccination withAbeta-displaying virus-like particles reduces soluble andinsoluble cerebral Abeta and lowers plaque burden in APPtransgenic mice. J Immunol 182, 7613-7624.

[1378] Zurbriggen R, Amacker M, Kammer AR, Westerfeld N,Borghgraef P, Van LF, Van dA I, Wera S (2005) Virosome-based active immunization targets soluble amyloid speciesrather than plaques in a transgenic mouse model ofAlzheimer’s disease. J Mol Neurosci 27, 157-166.

[1379] Salloway S, Sperling R, Gilman S, Fox NC, BlennowK, Raskind M, Sabbagh M, Honig LS, Doody R, vanDyck CH, Mulnard R, Barakos J, Gregg KM, Liu E,Lieberburg I, Schenk D, Black R, Grundman M (2009) Aphase 2 multiple ascending dose trial of bapineuzumab inmild to moderate Alzheimer disease. Neurology 73, 2061-2070.

[1380] Black RS, Sperling RA, Safirstein B, Motter RN, PallayA, Nichols A, Grundman M (2010) A single ascendingdose study of bapineuzumab in patients with Alzheimerdisease. Alzheimer Dis Assoc Disord 24, 198-203.

[1381] Laskowitz DT, Kolls BJ (2010) A phase 2 multiple ascend-ing dose trial of bapineuzumab in mild to moderateAlzheimer disease. Neurology 74, 2026-2027.

[1382] Sperling R, Salloway S, Brooks DJ, Tampieri D, Barakos J,Fox NC, Raskind M, Sabbagh M, Honig LS, PorsteinssonAP, Lieberburg I, Arrighi HM, Morris KA, Lu Y, Liu E,Gregg KM, Brashear HR, Kinney GG, Black R, Grund-man M (2012) Amyloid-related imaging abnormalitiesin patients with Alzheimer’s disease treated with bap-ineuzumab: A retrospective analysis. Lancet Neurol 11,241-249.

[1383] Rinne JO, Brooks DJ, Rossor MN, Fox NC, Bullock R,Klunk WE, Mathis CA, Blennow K, Barakos J, OkelloAA, Rodriguez Martinez de LS, Liu E, Koller M, GreggKM, Schenk D, Black R, Grundman M (2010) 11C-PiBPET assessment of change in fibrillar amyloid-beta loadin patients with Alzheimer’s disease treated with bap-ineuzumab: A phase 2, double-blind, placebo-controlled,ascending-dose study. Lancet Neurol 9, 363-372.

[1384] Blennow K, Zetterberg H, Rinne JO, Salloway S, WeiJ, Black R, Grundman M, Liu E (2012) Effect ofimmunotherapy with bapineuzumab on cerebrospinal fluidbiomarker levels in patients with mild to moderateAlzheimer disease. Arch Neurol 69, 1002-1010.

[1385] Kerchner GA, Boxer AL (2010) Bapineuzumab. ExpertOpin Biol Ther 10, 1121-1130.

[1386] Wilcock GK (2010) Bapineuzumab in Alzheimer’s dis-ease: Where now? Lancet Neurol 9, 134-136.

[1387] Panza F, Frisardi V, Imbimbo BP, D’Onofrio G, PietrarossaG, Seripa D, Pilotto A, Solfrizzi V (2010) Bapineuzumab:Anti-beta-amyloid monoclonal antibodies for the treat-ment of Alzheimer’s disease. Immunotherapy 2, 767-782.

[1388] Panza F, Frisardi V, Imbimbo BP, Seripa D, SolfrizziV, Pilotto A (2011) Monoclonal antibodies against beta-amyloid (Abeta) for the treatment of Alzheimer’s disease:The Abeta target at a crossroads. Expert Opin Biol Ther11, 679-686.

[1389] Panza F, Frisardi V, Imbimbo BP, Seripa D, Paris F,Santamato A, D’Onofrio G, Logroscino G, Pilotto A,Solfrizzi V (2011) Anti-beta-amyloid immunotherapyfor Alzheimer’s disease: Focus on bapineuzumab. CurrAlzheimer Res 8, 808-817.

[1390] Prins ND, Visser PJ, Scheltens P (2010) Can novel ther-apeutics halt the amyloid cascade? Alzheimers Res Ther2, 5.

Page 102: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

102 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1391] Roher AE, Maarouf CL, Daugs ID, Kokjohn TA, HunterJM, Sabbagh MN, Beach TG (2011) Neuropathology andamyloid-beta spectrum in a bapineuzumab immunother-apy recipient. J Alzheimers Dis 24, 315-325.

[1392] Zago W, Buttini M, Comery TA, Nishioka C, GardaiSJ, Seubert P, Games D, Bard F, Schenk D, Kinney GG(2012) Neutralization of Soluble, synaptotoxic amyloidbeta species by antibodies is epitope specific. J Neurosci32, 2696-2702.

[1393] Bard F, Cannon C, Barbour R, Burke RL, Games D, Gra-jeda H, Guido T, Hu K, Huang J, Johnson-Wood K, KhanK, Kholodenko D, Lee M, Lieberburg I, Motter R, NguyenM, Soriano F, Vasquez N, Weiss K, Welch B, Seubert P,Schenk D, Yednock T (2000) Peripherally administeredantibodies against amyloid beta-peptide enter the centralnervous system and reduce pathology in a mouse modelof Alzheimer disease. Nat Med 6, 916-919.

[1394] Siemers ER, Friedrich S, Dean RA, Gonzales CR, FarlowMR, Paul SM, DeMattos RB (2010) Safety and changes inplasma and cerebrospinal fluid amyloid beta after a singleadministration of an amyloid beta monoclonal antibodyin subjects with Alzheimer disease. Clin Neuropharmacol33, 67-73.

[1395] Carlson C, Estergard W, Oh J, Suhy J, Jack CR Jr, SiemersE, Barakos J (2011) Prevalence of asymptomatic vasogenicedema in pretreatment Alzheimer’s disease study cohortsfrom phase 3 trials of semagacestat and solanezumab.Alzheimers Dement 7, 396-401.

[1396] Reichert JM (2011) Antibody-based therapeutics to watchin 2011. MAbs 3, 76-99.

[1397] Samadi H, Sultzer D (2011) Solanezumab for Alzheimer’sdisease. Expert Opin Biol Ther 11, 787-798.

[1398] Imbimbo BP, Ottonello S, Frisardi V, Solfrizzi V, GrecoA, Seripa D, Pilotto A, Panza F (2012) Solanezumab forthe treatment of mild-to-moderate Alzheimer’s disease.Expert Rev Clin Immunol 8, 135-149.

[1399] Farlow M, Arnold SE, van Dyck CH, Aisen PS, SniderBJ, Porsteinsson AP, Friedrich S, Dean RA, Gonzales C,Sethuraman G, DeMattos RB, Mohs R, Paul SM, SiemersER (2012) Safety and biomarker effects of solanezumabin patients with Alzheimer’s disease. Alzheimers Dement8, 261-271.

[1400] Ostrowitzki S, Deptula D, Thurfjell L, Barkhof F,Bohrmann B, Brooks DJ, Klunk WE, Ashford E, YooK, Xu ZX, Loetscher H, Santarelli L (2012) Mechanismof amyloid removal in patients with Alzheimer diseasetreated with gantenerumab. Arch Neurol 69, 198-207.

[1401] Bohrmann B, Baumann K, Benz J, Gerber F, Huber W,Knoflach F, Messer J, Oroszlan K, Rauchenberger R,Richter WF, Rothe C, Urban M, Bardroff M, WinterM, Nordstedt C, Loetscher H (2012) Gantenerumab: Anovel human anti-abeta antibody demonstrates sustainedcerebral amyloid-beta binding and elicits cell-mediatedremoval of human amyloid-beta. J Alzheimers Dis 28,49-69.

[1402] Delrieu J, Ousset PJ, Vellas B (2012) Gantenerumab for thetreatment of Alzheimer’s disease. Expert Opin Biol Ther12, 1077-1086.

[1403] Adolfsson O, Pihlgren M, Toni N, Varisco Y, Buc-carello AL, Antoniello K, Lohmann S, Piorkowska K,Gafner V, Atwal JK, Maloney J, Chen M, GogineniA, Weimer RM, Mortensen DL, Friesenhahn M, HoC, Paul R, Pfeifer A, Muhs A, Watts RJ (2012) Aneffector-reduced anti-beta-amyloid (Abeta) antibody withunique Abeta binding properties promotes neuroprotec-

tion and glial engulfment of Abeta. J Neurosci 32, 9677-9689.

[1404] Balakrishnan K, ndrei-Selmer LC, Selmer T, Bacher M,Dodel R (2010) Comparison of intravenous immunoglob-ulins for naturally occurring autoantibodies againstamyloid-beta. J Alzheimers Dis 20, 135-143.

[1405] Dodel R, Neff F, Noelker C, Pul R, Du Y, Bacher M,Oertel W (2010) Intravenous immunoglobulins as a treat-ment for Alzheimer’s disease: Rationale and currentevidence. Drugs 70, 513-528.

[1406] Relkin NR (2008) Current state of immunotherapy forAlzheimer’s disease. CNS Spectr 13, 39-41.

[1407] Relkin NR, Szabo P, Adamiak B, Burgut T, Monthe C,Lent RW, Younkin S, Younkin L, Schiff R, Weksler ME(2009) 18-Month study of intravenous immunoglobulinfor treatment of mild Alzheimer disease. Neurobiol Aging30, 1728-1736.

[1408] Hughes RA, Dalakas MC, Cornblath DR, Latov N,Weksler ME, Relkin N (2009) Clinical applications ofintravenous immunoglobulins in neurology. Clin ExpImmunol 158(Suppl 1), 34-42.

[1409] Poetsch V, Neuhaus W, Noe CR (2010) Serum-derivedimmunoglobulins neutralize adverse effects of amyloid-beta peptide on the integrity of a blood-brain barrier invitro model. J Alzheimers Dis 21, 303-314.

[1410] Poetsch V, Benani-Baiti B, Neuhaus W, Muchitsch EM,Noe CR (2010) Serum-derived immunoglobulins alteramyloid beta transport across a blood-brain barrier in vitromodel. Pharmazie 65, 267-273.

[1411] Lanz TA, Schachter JB (2008) Solid-phase extractionenhances detection of beta-amyloid peptides in plasmaand enables Abeta quantification following passive immu-nization with Abeta antibodies. J Neurosci Methods 169,16-22.

[1412] Ding JD, Johnson LV, Herrmann R, Farsiu S, Smith SG,Groelle M, Mace BE, Sullivan P, Jamison JA, Kelly U,Harrabi O, Bollini SS, Dilley J, Kobayashi D, Kuang B, LiW, Pons J, Lin JC, Bowes RC (2011) Anti-amyloid therapyprotects against retinal pigmented epithelium damage andvision loss in a model of age-related macular degeneration.Proc Natl Acad Sci U S A 108, E279-E287.

[1413] Venkataramani V, Wirths O, Budka H, Hartig W,Kovacs GG, Bayer TA (2012) Antibody 9D5 recog-nizes oligomeric pyroglutamate amyloid-beta in a fractionof amyloid-beta deposits in Alzheimer’s disease with-out cross-reactivity with other protein aggregates. JAlzheimers Dis 29, 361-371.

[1414] 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.

[1415] 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.

[1416] 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.

[1417] 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.

Page 103: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 103

[1418] 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.

[1419] 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.

[1420] 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.

[1421] Pardridge WM, Boado RJ (2012) Reengineering biophar-maceuticals for targeted delivery across the blood-brainbarrier. Methods Enzymol 503, 269-292.

[1422] Boado RJ, Pardridge WM (2011) The Trojan Horse lipo-some technology for nonviral gene transfer across theblood-brain barrier. J Drug Deliv 2011, 296151.

[1423] Goldshmit Y, Matteo R, Sztal T, Ellett F, Frisca F, MorenoK, Crombie D, Lieschke GJ, Currie PD, Sabbadini RA,Pebay A (2012) Blockage of lysophosphatidic Acid sig-naling improves spinal cord injury outcomes. Am J Pathol181, 978-992.

[1424] Solomon B, Koppel R, Hanan E, Katzav T (1996) Mon-oclonal antibodies inhibit in vitro fibrillar aggregation ofthe Alzheimer beta-amyloid peptide. Proc Natl Acad SciU S A 93, 452-455.

[1425] Solomon B, Koppel R, Frankel D, Hanan-Aharon E(1997) Disaggregation of Alzheimer beta-amyloid by site-directed mAb. Proc Natl Acad Sci U S A 94, 4109-4112.

[1426] Buttini M, Masliah E, Barbour R, Grajeda H, MotterR, Johnson-Wood K, Khan K, Seubert P, Freedman S,Schenk D, Games D (2005) Beta-amyloid immunother-apy prevents synaptic degeneration in a mouse model ofAlzheimer’s disease. J Neurosci 25, 9096-9101.

[1427] Morgan D (2005) Mechanisms of A beta plaque clearancefollowing passive A beta immunization. Neurodegener Dis2, 261-266.

[1428] Levites Y, Das P, Price RW, Rochette MJ, KosturaLA, McGowan EM, Murphy MP, Golde TE (2006)Anti-Abeta42- and anti-Abeta40-specific mAbs attenuateamyloid deposition in an Alzheimer disease mouse model.J Clin Invest 116, 193-201.

[1429] Levites Y, Smithson LA, Price RW, Dakin RS, Yuan B,Sierks MR, Kim J, McGowan E, Reed DK, RosenberryTL, Das P, Golde TE (2006) Insights into the mechanismsof action of anti-Abeta antibodies in Alzheimer’s diseasemouse models. FASEB J 20, 2576-2578.

[1430] Chang WP, Downs D, Huang XP, Da H, Fung KM, TangJ (2007) Amyloid-beta reduction by memapsin 2 (beta-secretase) immunization. FASEB J 21, 3184-3196.

[1431] Chauhan NB (2007) Intracerebroventricular pas-sive immunization with anti-oligoAbeta antibody inTgCRND8. J Neurosci Res 85, 451-463.

[1432] Gardberg AS, Dice LT, Ou S, Rich RL, Helmbrecht E,Ko J, Wetzel R, Myszka DG, Patterson PH, Dealwis C(2007) Molecular basis for passive immunotherapy ofAlzheimer’s disease. Proc Natl Acad Sci U S A 104, 15659-15664.

[1433] Habicht G, Haupt C, Friedrich RP, Hortschansky P,Sachse C, Meinhardt J, Wieligmann K, Gellermann GP,Brodhun M, Gotz J, Halbhuber KJ, Rocken C, Horn U,Fandrich M (2007) Directed selection of a conformationalantibody domain that prevents mature amyloid fibril for-

mation by stabilizing Abeta protofibrils. Proc Natl AcadSci U S A 104, 19232-19237.

[1434] Kayed R, Head E, Sarsoza F, Saing T, Cotman CW, Nec-ula M, Margol L, Wu J, Breydo L, Thompson JL, RasoolS, Gurlo T, Butler P, Glabe CG (2007) Fibril specific,conformation dependent antibodies recognize a genericepitope common to amyloid fibrils and fibrillar oligomersthat is absent in prefibrillar oligomers. Mol Neurodegener2, 18.

[1435] Kayed R, Canto I, Breydo L, Rasool S, Lukacsovich T,Wu J, Albay R III, Pensalfini A, Yeung S, Head E, MarshJL, Glabe C (2010) Conformation dependent monoclonalantibodies distinguish different replicating strains or con-formers of prefibrillar Abeta oligomers. Mol Neurodegener5, 57.

[1436] Mamikonyan G, Necula M, Mkrtichyan M, GhochikyanA, Petrushina I, Movsesyan N, Mina E, Kiyatkin A, GlabeCG, Cribbs DH, Agadjanyan MG (2007) Anti-A beta 1-11antibody binds to different beta-amyloid species, inhibitsfibril formation, and disaggregates preformed fibrils butnot the most toxic oligomers. J Biol Chem 282, 22376-22386.

[1437] Moretto N, Bolchi A, Rivetti C, Imbimbo BP, VillettiG, Pietrini V, Polonelli L, Del SS, Smith KM, FerranteRJ, Ottonello S (2007) Conformation-sensitive antibod-ies against alzheimer amyloid-beta by immunization witha thioredoxin-constrained B-cell epitope peptide. J BiolChem 282, 11436-11445.

[1438] Poduslo JF, Ramakrishnan M, Holasek SS, Ramirez-Alvarado M, Kandimalla KK, Gilles EJ, Curran GL,Wengenack TM (2007) In vivo targeting of antibodyfragments to the nervous system for Alzheimer’s dis-ease immunotherapy and molecular imaging of amyloidplaques. J Neurochem 102, 420-433.

[1439] Rakover I, Arbel M, Solomon B (2007) Immunotherapyagainst APP beta-secretase cleavage site improves cogni-tive function and reduces neuroinflammation in Tg2576mice without a significant effect on brain abeta levels.Neurodegener Dis 4, 392-402.

[1440] Song MS, Mook-Jung I, Lee HJ, Min JY, Park MH (2007)Serum anti-amyloid-beta antibodies and Alzheimer’s dis-ease in elderly Korean patients. J Int Med Res 35, 301-306.

[1441] Tampellini D, Magrane J, Takahashi RH, Li F, Lin MT,Almeida CG, Gouras GK (2007) Internalized antibod-ies to the Abeta domain of APP reduce neuronal Abetaand protect against synaptic alterations. J Biol Chem 282,18895-18906.

[1442] Bolukbasi Hatip FF, Matsunaga Y, Yamada T (2008) Spe-cific reactivity of mild/severe Alzheimer’s disease patient’ssera to antibody against Abeta1-40 epitope 17-21. ActaNeurol Scand 117, 404-408.

[1443] Koenigsknecht-Talboo J, Meyer-Luehmann M, Parsada-nian M, Garcia-Alloza M, Finn MB, Hyman BT, BacskaiBJ, Holtzman DM (2008) Rapid microglial responsearound amyloid pathology after systemic anti-Abeta anti-body administration in PDAPP mice. J Neurosci 28,14156-14164.

[1444] Manea M, Kalaszi A, Mezo G, Horvati K, Bodor A, Hor-vath A, Farkas O, Perczel A, Przybylski M, Hudecz F(2008) Antibody recognition and conformational flexibil-ity of a plaque-specific beta-amyloid epitope modulatedby non-native peptide flanking regions. J Med Chem 51,1150-1161.

[1445] Schroeter S, Khan K, Barbour R, Doan M, Chen M, GuidoT, Gill D, Basi G, Schenk D, Seubert P, Games D (2008)

Page 104: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

104 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

Immunotherapy reduces vascular amyloid-beta in PDAPPmice. J Neurosci 28, 6787-6793.

[1446] Tucker SM, Borchelt DR, Troncoso JC (2008) Limitedclearance of pre-existing amyloid plaques after intracere-bral injection of Abeta antibodies in two mouse models ofAlzheimer disease. J Neuropathol Exp Neurol 67, 30-40.

[1447] Xu W, Kawarabayashi T, Matsubara E, Deguchi K,Murakami T, Harigaya Y, Ikeda M, Amari M, Kuwano R,Abe K, Shoji M (2008) Plasma antibodies to Abeta40 andAbeta42 in patients with Alzheimer’s disease and normalcontrols. Brain Res 1219, 169-179.

[1448] Itoh T, Satou T, Nishida S, Tsubaki M, Hashimoto S, Ito H(2009) Improvement of cerebral function by anti-amyloidprecursor protein antibody infusion after traumatic braininjury in rats. Mol Cell Biochem 324, 191-199.

[1449] Lord A, Gumucio A, Englund H, Sehlin D, Sundquist VS,Soderberg L, Moller C, Gellerfors P, Lannfelt L, PetterssonFE, Nilsson LN (2009) An amyloid-beta protofibril-selective antibody prevents amyloid formation in a mousemodel of Alzheimer’s disease. Neurobiol Dis 36, 425-434.

[1450] Rozkalne A, Spires-Jones TL, Stern EA, Hyman BT(2009) A single dose of passive immunotherapy hasextended benefits on synapses and neurites in an Alzh-eimer’s disease mouse model. Brain Res 1280, 178-185.

[1451] Spires-Jones TL, Mielke ML, Rozkalne A, Meyer-Luehmann M, de CA, Bacskai BJ, Schenk D, Hyman BT(2009) Passive immunotherapy rapidly increases structuralplasticity in a mouse model of Alzheimer disease. Neuro-biol Dis 33, 213-220.

[1452] Xu YX, Wang HQ, Yan J, Sun XB, Guo JC, Zhu CQ (2009)Antibody binding to cell surface amyloid precursor proteininduces neuronal injury by deregulating the phosphoryla-tion of focal adhesion signaling related proteins. NeurosciLett 465, 276-281.

[1453] Ying Z, Xin W, Jin-Sheng H, Fu-Xiang B, Wei-Min S,Xin-Xian D, Xiao-Bo W, Yi-Qin L, Xian-Xian Z, Hong-Gang H, Xiang-Lei P, Yan-Peng Z, Ling-Ling H, Tao H(2009) Preparation and characterization of a monoclonalantibody with high affinity for soluble Abeta oligomers.Hybridoma (Larchmt) 28, 349-354.

[1454] Axelsen TV, Holm A, Birkelund S, Christiansen G, PlougM, Holm IE (2009) Specific recognition of the C-terminalend of A beta 42 by a high affinity monoclonal antibody.Mol Immunol 46, 2267-2273.

[1455] Adekar SP, Klyubin I, Macy S, Rowan MJ, SolomonA, Dessain SK, O’Nuallain B (2010) Inherent anti-amyloidogenic activity of human immunoglobulin gammaheavy chains. J Biol Chem 285, 1066-1074.

[1456] Basi GS, Feinberg H, Oshidari F, Anderson J, BarbourR, Baker J, Comery TA, Diep L, Gill D, Johnson-WoodK, Goel A, Grantcharova K, Lee M, Li J, Partridge A,Griswold-Prenner I, Piot N, Walker D, Widom A, Pan-galos MN, Seubert P, Jacobsen JS, Schenk D, Weis WI(2010) Structural correlates of antibodies associated withacute reversal of amyloid beta-related behavioral deficitsin a mouse model of Alzheimer disease. J Biol Chem 285,3417-3427.

[1457] Luo F, Rustay NR, Seifert T, Roesner B, Hradil V, HillenH, Ebert U, Severin JM, Cox BF, Llano DA, Day M,Fox GB (2010) Magnetic resonance imaging detectionand time course of cerebral microhemorrhages during pas-sive immunotherapy in living amyloid precursor proteintransgenic mice. J Pharmacol Exp Ther 335, 580-588.

[1458] Szabo P, Mujalli DM, Rotondi ML, Sharma R, WeberA, Schwarz HP, Weksler ME, Relkin N (2010) Measure-

ment of anti-beta amyloid antibodies in human blood.J Neuroimmunol 227, 167-174.

[1459] Winkler DT, Abramowski D, Danner S, Zurini M,Paganetti P, Tolnay M, Staufenbiel M (2010) Rapid cere-bral amyloid binding by Abeta antibodies infused intobeta-amyloid precursor protein transgenic mice. Biol Psy-chiatry 68, 971-974.

[1460] Lindhagen-Persson M, Brannstrom K, Vestling M, SteinitzM, Olofsson A (2010) Amyloid-beta oligomer speci-ficity mediated by the IgM isotype–implications for aspecific protective mechanism exerted by endogenousauto-antibodies. PLoS One 5, e13928.

[1461] Miller DL, Potempska A, Wegiel J, Mehta PD (2011)High-affinity rabbit monoclonal antibodies specific foramyloid peptides amyloid-beta40 and amyloid-beta42.J Alzheimers Dis 23, 293-305.

[1462] Zhang Y, He JS, Wang X, Wang J, Bao FX, Pang SY,Yin F, Hu HG, Peng XL, Sun WM, Zheng YP, Hou LL,Hong T (2011) Administration of amyloid-beta42oligomer-specific monoclonal antibody improved mem-ory performance in SAMP8 mice. J Alzheimers Dis 23,551-561.

[1463] O’Nuallain B, Klyubin I, Mc Donald JM, Foster JS, WelzelA, Barry A, Dykoski RK, Cleary JP, Gebbink MF, RowanMJ, Walsh DM (2011) A monoclonal antibody against syn-thetic Abeta dimer assemblies neutralizes brain-derivedsynaptic plasticity-disrupting Abeta. J Neurochem 119,189-201.

[1464] Dodel R, Balakrishnan K, Keyvani K, Deuster O, Neff F,ndrei-Selmer LC, Roskam S, Stuer C, Al-Abed Y, NoelkerC, Balzer-Geldsetzer M, Oertel W, Du Y, Bacher M (2011)Naturally occurring autoantibodies against beta-amyloid:Investigating their role in transgenic animal and in vitromodels of Alzheimer’s disease. J Neurosci 31, 5847-5854.

[1465] Tanaka K, Nishimura M, Yamaguchi Y, Hashiguchi S,Takiguchi S, Yamaguchi M, Tahara H, Gotanda T, AbeR, Ito Y, Sugimura K (2011) A mimotope peptide ofAbeta42 fibril-specific antibodies with Abeta42 fibrillationinhibitory activity induces anti-Abeta42 conformer anti-body response by a displayed form on an M13 phage inmice. J Neuroimmunol 236, 27-38.

[1466] Morgado I, Wieligmann K, Bereza M, Ronicke R,Meinhardt K, Annamalai K, Baumann M, Wacker J,Hortschansky P, Malesevic M, Parthier C, Mawrin C,Schiene-Fischer C, Reymann KG, Stubbs MT, Balbach J,Gorlach M, Horn U, Fandrich M (2012) Molecular basisof beta-amyloid oligomer recognition with a conforma-tional antibody fragment. Proc Natl Acad Sci U S A 109,12503-12508.

[1467] Dorothee G, Bottlaender M, Moukari E, de Souza LC,Maroy R, Corlier F, Colliot O, Chupin M, Lamari F,Lehericy S, Dubois B, Sarazin M, Aucouturier P (2012)Distinct patterns of antiamyloid-beta antibodies in typicaland atypical Alzheimer disease. Arch Neurol 69, 1181-1185.

[1468] Arbel M, Solomon B (2007) A novel immunotherapy forAlzheimer’s disease: Antibodies against the beta-secretasecleavage site of APP. Curr Alzheimer Res 4, 437-445.

[1469] Arbel M, Solomon B (2007) Immunotherapy forAlzheimer’s disease: Attacking amyloid-beta from theinside. Trends Immunol 28, 511-513.

[1470] Britschgi M, Wyss-Coray T (2007) Systemic and acquiredimmune responses in Alzheimer’s disease. Int Rev Neuro-biol 82, 205-233.

Page 105: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 105

[1471] Cardinale A, Biocca S (2008) The potential of intracellularantibodies for therapeutic targeting of protein-misfoldingdiseases. Trends Mol Med 14, 373-380.

[1472] Lichtlen P, Mohajeri MH (2008) Antibody-basedapproaches in Alzheimer’s research: Safety, pharmacoki-netics, metabolism, and analytical tools. J Neurochem 104,859-874.

[1473] Bednar MM (2009) Anti-amyloid antibody drugs in clin-ical testing for Alzheimer’s disease. IDrugs 12, 566-575.

[1474] Jicha GA (2009) Is passive immunization for Alzheimer’sdisease ‘alive and well’ or ‘dead and buried’? Expert OpinBiol Ther 9, 481-491.

[1475] Lambert MP, Velasco PT, Viola KL, Klein WL (2009) Tar-geting generation of antibodies specific to conformationalepitopes of amyloid beta-derived neurotoxins. CNS NeurolDisord Drug Targets 8, 65-81.

[1476] Pul R, Dodel R, Stangel M (2011) Antibody-based therapyin Alzheimer’s disease. Expert Opin Biol Ther 11, 343-357.

[1477] Hermann DM, Keyvani K, van de NJ, Weimar C, WiltfangJ, Nitsch RM, Szodorai A (2011) Brain-reactive beta-amyloid antibodies in primary CNS angiitis with cerebralamyloid angiopathy. Neurology 77, 503-505.

[1478] Merlini G (2011) Reducing the amyloid burden throughimmunotherapy: A major therapeutic advance. NephrolDial Transplant 26, 1471-1473.

[1479] Perchiacca JM, Ladiwala AR, Bhattacharya M, TessierPM (2012) Structure-based design of conformation- andsequence-specific antibodies against amyloid beta. ProcNatl Acad Sci U S A 109, 84-89.

[1480] Liu YH, Giunta B, Zhou HD, Tan J, Wang YJ (2012)Immunotherapy for Alzheimer disease-the challenge ofadverse effects. Nat Rev Neurol 8, 465-469.

[1481] Dodart JC, Bales KR, Gannon KS, Greene SJ, DeMattosRB, Mathis C, DeLong CA, Wu S, Wu X, Holtzman DM,Paul SM (2002) Immunization reverses memory deficitswithout reducing brain Abeta burden in Alzheimer’s dis-ease model. Nat Neurosci 5, 452-457.

[1482] Yamada K, Yabuki C, Seubert P, Schenk D, Hori Y, Oht-suki S, Terasaki T, Hashimoto T, Iwatsubo T (2009) Abetaimmunotherapy: Intracerebral sequestration of Abeta byan anti-Abeta monoclonal antibody 266 with high affinityto soluble Abeta. J Neurosci 29, 11393-11398.

[1483] Freeman GB, Lin JC, Pons J, Raha NM (2012) 39-week toxicity and toxicokinetic study of ponezumab(PF-04360365) in cynomolgus monkeys with 12-weekrecovery period. J Alzheimers Dis 28, 531-541.

[1484] Freeman GB, Brown TP, Wallace K, Bales KR (2012)Chronic administration of an aglycosylated murine anti-body of ponezumab does not worsen microhemorrhagesin aged Tg2576 mice. Curr Alzheimer Res, [Epub aheadof print].

[1485] La Porte SL, Bollini SS, Lanz TA, Abdiche YN, RusnakAS, Ho WH, Kobayashi D, Harrabi O, Pappas D, MinaEW, Milici AJ, Kawabe TT, Bales K, Lin JC, Pons J(2012) Structural basis of C-terminal beta-amyloid pep-tide binding by the antibody ponezumab for the treatmentof Alzheimer’s disease. J Mol Biol 421, 525-536.

[1486] Crespo-Biel N, Theunis C, Van Leuven F (2012) Proteintau: Prime cause of synaptic and neuronal degenera-tion in Alzheimer’s disease. Int J Alzheimers Dis 2012,251426.

[1487] Morris M, Maeda S, Vossel K, Mucke L (2011) The manyfaces of tau. Neuron 70, 410-426.

[1488] Kolarova M, Garcia-Sierra F, Bartos A, Ricny J, Ripova D(2012) Structure and pathology of tau protein in Alzheimerdisease. Int J Alzheimers Dis 2012, 731526.

[1489] Mandelkow EM, Mandelkow E (2012) Biochemistry andcell biology of tau protein in neurofibrillary degeneration.Cold Spring Harb Perspect Med 2, a006247.

[1490] Lee VM, Brunden KR, Hutton M, Trojanowski JQ(2011) Developing therapeutic approaches to tau, selectedkinases, and related neuronal protein targets. Cold SpringHarb Perspect Med 1, a006437.

[1491] Santacruz K, Lewis J, Spires T, Paulson J, KotilinekL, Ingelsson M, Guimaraes A, DeTure M, Ramsden M,McGowan E, Forster C, Yue M, Orne J, Janus C, Mari-ash A, Kuskowski M, Hyman B, Hutton M, Ashe KH(2005) Tau suppression in a neurodegenerative mousemodel improves memory function. Science 309, 476-481.

[1492] Berger Z, Roder H, Hanna A, Carlson A, Rangachari V,Yue M, Wszolek Z, Ashe K, Knight J, Dickson D, AndorferC, Rosenberry TL, Lewis J, Hutton M, Janus C (2007)Accumulation of pathological tau species and memory lossin a conditional model of tauopathy. J Neurosci 27, 3650-3662.

[1493] Wang JZ, Xia YY, Grundke-Iqbal I, Iqbal K (2012)Abnormal hyperphosphorylation of tau: Sites, regulation,and molecular mechanism of neurofibrillary degeneration.J Alzheimers Dis, doi: 10.3233/JAD-2012-129031 [Epubahead of print].

[1494] Martin L, Latypova X, Wilson CM, Magnaudeix A, Per-rin ML, Yardin C, Terro F (2012) Tau protein kinases:Involvement in Alzheimer’s disease. Ageing Res Rev 12,289-309.

[1495] Bekris LM, Millard S, Lutz F, Li G, Galasko DR, FarlowMR, Quinn JF, Kaye JA, Leverenz JB, Tsuang DW, Yu CE,Peskind ER (2012) Tau phosphorylation pathway genesand cerebrospinal fluid tau levels in Alzheimer’s disease.Am J Med Genet B Neuropsychiatr Genet 159B, 874-883.

[1496] Kester MI, van d V, Blankenstein MA, Pijnenburg YA,van Elk EJ, Scheltens P, van der Flier WM (2009) CSFbiomarkers predict rate of cognitive decline in Alzheimerdisease. Neurology 73, 1353-1358.

[1497] Ittner LM, Gotz J (2011) Amyloid-beta and tau–a toxicpas de deux in Alzheimer’s disease. Nat Rev Neurosci 12,65-72.

[1498] Ittner LM, Ke YD, Delerue F, Bi M, Gladbach A, van EJ,Wolfing H, Chieng BC, Christie MJ, Napier IA, Eckert A,Staufenbiel M, Hardeman E, Gotz J (2010) Dendritic func-tion of tau mediates amyloid-beta toxicity in Alzheimer’sdisease mouse models. Cell 142, 387-397.

[1499] Ittner A, Ke YD, van EJ, Gladbach A, Gotz J, Ittner LM(2011) Brief update on different roles of tau in neurode-generation. IUBMB Life 63, 495-502.

[1500] Roberson ED, Halabisky B, Yoo JW, Yao J, Chin J, YanF, Wu T, Hamto P, Devidze N, Yu GQ, Palop JJ, NoebelsJL, Mucke L (2011) Amyloid-beta/Fyn-induced synaptic,network, and cognitive impairments depend on tau levels inmultiple mouse models of Alzheimer’s disease. J Neurosci31, 700-711.

[1501] Gotz J, Ittner A, Ittner LM (2012) Tau-targeted treatmentstrategies in Alzheimer’s disease. Br J Pharmacol 165,1246-1259.

[1502] Mondragon-Rodriguez S, Perry G, Zhu X, Boehm J (2012)Amyloid Beta and tau proteins as therapeutic targets forAlzheimer’s disease treatment: Rethinking the currentstrategy. Int J Alzheimers Dis 2012, 630182.

Page 106: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

106 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1503] Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, Spires-Jones TL, Hyman BT (2012) The synaptic accumulation ofhyperphosphorylated tau oligomers in Alzheimer diseaseis associated with dysfunction of the ubiquitin-proteasomesystem. Am J Pathol 181, 1426-1435.

[1504] Jack CR Jr, Knopman DS, Jagust WJ, Shaw LM, Aisen PS,Weiner MW, Petersen RC, Trojanowski JQ (2010) Hypo-thetical model of dynamic biomarkers of the Alzheimer’spathological cascade. Lancet Neurol 9, 119-128.

[1505] Jack CR Jr, Vemuri P, Wiste HJ, Weigand SD, Aisen PS,Trojanowski JQ, Shaw LM, Bernstein MA, Petersen RC,Weiner MW, Knopman DS (2011) Evidence for orderingof Alzheimer disease biomarkers. Arch Neurol 68, 1526-1535.

[1506] Jack CR Jr, Vemuri P, Wiste HJ, Weigand SD, Lesnick TG,Lowe V, Kantarci K, Bernstein MA, Senjem ML, GunterJL, Boeve BF, Trojanowski JQ, Shaw LM, Aisen PS,Weiner MW, Petersen RC, Knopman DS (2012) Shapesof the trajectories of 5 major biomarkers of Alzheimerdisease. Arch Neurol 69, 856-867.

[1507] Han SD, Gruhl J, Beckett L, Dodge HH, Stricker NH,Farias S, Mungas D (2012) Beta amyloid, tau, neuroimag-ing, and cognition: Sequence modeling of biomarkersfor Alzheimer’s disease. Brain Imaging Behav, doi:10.1007/s11682-012-9177-0 [Epub ahead of print].

[1508] Wischik CM, Bentham P, Wishik DJ, SK (2008) Tauaggregation inhibitor (TAI) therapy with Rember™ arrestsdisease progression in mild to moderate Alzheimer’s dis-ease over 50 weeks. Alzheimers Dementia 4, T167.

[1509] Wischik C, Staff R (2009) Challenges in the conduct ofdisease-modifying trials in AD: Practical experience froma phase 2 trial of Tau-aggregation inhibitor therapy. J NutrHealth Aging 13, 367-369.

[1510] Wischik CM, Edwards PC, Lai RY, Roth M, HarringtonCR (1996) Selective inhibition of Alzheimer disease-liketau aggregation by phenothiazines. Proc Natl Acad SciU S A 93, 11213-11218.

[1511] Atamna H, Nguyen A, Schultz C, Boyle K, Newberry J,Kato H, Ames BN (2008) Methylene blue delays cellularsenescence and enhances key mitochondrial biochemicalpathways. FASEB J 22, 703-712.

[1512] Oz M, Lorke DE, Petroianu GA (2009) Methylene blue andAlzheimer’s disease. Biochem Pharmacol 78, 927-932.

[1513] Oz M, Lorke DE, Hasan M, Petroianu GA (2011) Cellularand molecular actions of Methylene Blue in the nervoussystem. Med Res Rev 31, 93-117.

[1514] Oz M, Isaev D, Lorke DE, Hasan M, Petroianu G, Ship-penberg TS (2012) Methylene blue inhibits function of the5-HT transporter. Br J Pharmacol 166, 168-176.

[1515] Atamna H, Kumar R (2010) Protective role of methy-lene blue in Alzheimer’s disease via mitochondria andcytochrome c oxidase. J Alzheimers Dis 20(Suppl 2),S439-S452.

[1516] Schirmer RH, Adler H, Pickhardt M, Mandelkow E (2011)“Lest we forget you–methylene blue...”. Neurobiol Aging32, 2325-2316.

[1517] O’Leary JC, Li Q III, Marinec P, Blair LJ, Congdon EE,Johnson AG, Jinwal UK, Koren J III, Jones JR, Kraft C,Peters M, Abisambra JF, Duff KE, Weeber EJ, GestwickiJE, Dickey CA (2010) Phenothiazine-mediated rescue ofcognition in tau transgenic mice requires neuroprotectionand reduced soluble tau burden. Mol Neurodegener 5, 45.

[1518] Medina DX, Caccamo A, Oddo S (2011) Methylene bluereduces abeta levels and rescues early cognitive deficit byincreasing proteasome activity. Brain Pathol 21, 140-149.

[1519] Necula M, Breydo L, Milton S, Kayed R, van d V, Tone P,Glabe CG (2007) Methylene blue inhibits amyloid Abetaoligomerization by promoting fibrillization. Biochemistry46, 8850-8860.

[1520] del Ser T, Steinwachs KC, Gertz HJ, Andres MV, Gomez-Carrillo B, Medina M, Vericat JA, Redondo P, FleetD, Leon T (2012) Treatment of Alzheimer’s diseasewith the GSK-3 inhibitor tideglusib: A pilot study. JAlzheimers Dis, doi: 10.3233/JAD-2012-120805 [Epubahead of print].

[1521] Rojo LE, Alzate-Morales J, Saavedra IN, Davies P, Mac-cioni RB (2010) Selective interaction of lansoprazole andastemizole with tau polymers: Potential new clinical usein diagnosis of Alzheimer’s disease. J Alzheimers Dis 19,573-589.

[1522] Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M,Starkova N, Calingasan NY, Yang L, Tampellini D, StarkovAA, Chan RB, Di PG, Pujol A, Beal MF (2012) Bezafi-brate administration improves behavioral deficits and taupathology in P301S mice. Hum Mol Genet 21, 5091-5105.

[1523] Brunden KR, Zhang B, Carroll J, Yao Y, Potuzak JS,Hogan AM, Iba M, James MJ, Xie SX, Ballatore C, SmithAB III, Lee VM, Trojanowski JQ (2010) Epothilone Dimproves microtubule density, axonal integrity, and cogni-tion in a transgenic mouse model of tauopathy. J Neurosci30, 13861-13866.

[1524] Yang Y, Ma D, Wang Y, Jiang T, Hu S, Zhang M, YuX, Gong CX (2012) Intranasal insulin ameliorates tauhyperphosphorylation in a rat model of type 2 diabetes.J Alzheimers Dis, doi: 10.3233/JAD-2012-121294-2012[Epub ahead of print].

[1525] Taghavi A, Nasir S, Pickhardt M, Heyny-von Haussen R,Mall G, Mandelkow E, Mandelkow EM, Schmidt B (2011)N′-benzylidene-benzohydrazides as novel and selectivetau-PHF ligands. J Alzheimers Dis 27, 835-843.

[1526] Martin L, Latypova X, Wilson CM, Magnaudeix A, Per-rin ML, Terro F (2012) Tau protein phosphatases inAlzheimer’s disease: The leading role of PP2A. AgeingRes Rev 12, 39-49.

[1527] Braithwaite SP, Stock JB, Lombroso PJ, Nairn AC (2012)Protein phosphatases and Alzheimer’s disease. Prog MolBiol Transl Sci 106, 343-379.

[1528] van Eersel J, Ke YD, Liu X, Delerue F, Kril JJ, Gotz J,Ittner LM (2010) Sodium selenate mitigates tau pathology,neurodegeneration, and functional deficits in Alzheimer’sdisease models. Proc Natl Acad Sci U S A 107, 13888-13893.

[1529] 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.

[1530] Yuzwa SA, Vocadlo DJ (2009) O-GlcNAc modificationand the tauopathies: Insights from chemical biology. CurrAlzheimer Res 6, 451-454.

[1531] Yuzwa SA, Yadav AK, Skorobogatko Y, Clark T, VossellerK, Vocadlo DJ (2011) Mapping O-GlcNAc modificationsites on tau and generation of a site-specific O-GlcNAc tauantibody. Amino Acids 40, 857-868.

[1532] 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.

[1533] Shen DL, Gloster TM, Yuzwa SA, Vocadlo DJ (2012)Insights into O-GlcNAc processing and dynamics

Page 107: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 107

through kinetic analysis of O-GlcNAc transferase andO-GlcNAcase activity on protein substrates. J Biol Chem287, 15395-15408.

[1534] Fischer PM (2008) Turning down tau phosphorylation. NatChem Biol 4, 448-449.

[1535] Lefebvre T (2012) Recall sugars, forget Alzheimer’s.Nature Chem Biol 8, 325-326.

[1536] Navarrete LP, Perez P, Morales I, Maccioni RB (2011)Novel drugs affecting tau behavior in the treatment ofAlzheimer’s disease and tauopathies. Curr Alzheimer Res8, 678-685.

[1537] Navarrete LP, Guzman L, San MA, studillo-Saavedra L,Maccioni RB (2012) Molecules of the quinoline familyblock tau self-aggregation: Implications toward a thera-peutic approach for Alzheimer’s disease. J Alzheimers Dis29, 79-88.

[1538] Calcul L, Zhang B, Jinwal UK, Dickey CA, Baker BJ(2012) Natural products as a rich source of tau-targetingdrugs for Alzheimer’s disease. Future Med Chem 4, 1751-1761.

[1539] Chirita C, Necula M, Kuret J (2004) Ligand-dependentinhibition and reversal of tau filament formation. Biochem-istry 43, 2879-2887.

[1540] Necula M, Chirita CN, Kuret J (2005) Cyanine dye N744inhibits tau fibrillization by blocking filament extension:Implications for the treatment of tauopathic neurodegen-erative diseases. Biochemistry 44, 10227-10237.

[1541] Pickhardt M, von BM, Gazova Z, Hascher A, BiernatJ, Mandelkow EM, Mandelkow E (2005) Screening forinhibitors of tau polymerization. Curr Alzheimer Res 2,219-226.

[1542] Pickhardt M, Gazova Z, von BM, Khlistunova I, WangY, Hascher A, Mandelkow EM, Biernat J, Mandelkow E(2005) Anthraquinones inhibit tau aggregation and dis-solve Alzheimer’s paired helical filaments in vitro and incells. J Biol Chem 280, 3628-3635.

[1543] Khlistunova I, Biernat J, Wang Y, Pickhardt M, vonBM, Gazova Z, Mandelkow E, Mandelkow EM (2006)Inducible expression of Tau repeat domain in cell mod-els of tauopathy: Aggregation is toxic to cells but can bereversed by inhibitor drugs. J Biol Chem 281, 1205-1214.

[1544] Bulic B, Pickhardt M, Khlistunova I, Biernat J, MandelkowEM, Mandelkow E, Waldmann H (2007) Rhodanine-basedtau aggregation inhibitors in cell models of tauopathy.Angew Chem Int Ed Engl 46, 9215-9219.

[1545] Khlistunova I, Pickhardt M, Biernat J, Wang Y, Man-delkow EM, Mandelkow E (2007) Inhibition of tauaggregation in cell models of tauopathy. Curr AlzheimerRes 4, 544-546.

[1546] Pickhardt M, Larbig G, Khlistunova I, Coksezen A, MeyerB, Mandelkow EM, Schmidt B, Mandelkow E (2007)Phenylthiazolyl-hydrazide and its derivatives are potentinhibitors of tau aggregation and toxicity in vitro and incells. Biochemistry 46, 10016-10023.

[1547] Pickhardt M, Biernat J, Khlistunova I, Wang YP, GazovaZ, Mandelkow EM, Mandelkow E (2007) N-phenylaminederivatives as aggregation inhibitors in cell models oftauopathy. Curr Alzheimer Res 4, 397-402.

[1548] Crowe A, Ballatore C, Hyde E, Trojanowski JQ, LeeVM (2007) High throughput screening for small moleculeinhibitors of heparin-induced tau fibril formation. BiochemBiophys Res Commun 358, 1-6.

[1549] Honson NS, Jensen JR, Darby MV, Kuret J (2007) Potentinhibition of tau fibrillization with a multivalent ligand.Biochem Biophys Res Commun 363, 229-234.

[1550] Larbig G, Pickhardt M, Lloyd DG, Schmidt B, MandelkowE (2007) Screening for inhibitors of tau protein aggre-gation into Alzheimer paired helical filaments: A ligandbased approach results in successful scaffold hopping.Curr Alzheimer Res 4, 315-323.

[1551] Bulic B, Pickhardt M, Schmidt B, Mandelkow EM,Waldmann H, Mandelkow E (2009) Development of tauaggregation inhibitors for Alzheimer’s disease. AngewChem Int Ed Engl 48, 1740-1752.

[1552] Chang E, Congdon EE, Honson NS, Duff KE, Kuret J(2009) Structure-activity relationship of cyanine tau aggre-gation inhibitors. J Med Chem 52, 3539-3547.

[1553] Chang E, Honson NS, Bandyopadhyay B, Funk KE, JensenJR, Kim S, Naphade S, Kuret J (2009) Modulation anddetection of tau aggregation with small-molecule ligands.Curr Alzheimer Res 6, 409-414.

[1554] Crowe A, Huang W, Ballatore C, Johnson RL, HoganAM, Huang R, Wichterman J, McCoy J, Huryn D,Auld DS, Smith AB III, Inglese J, Trojanowski JQ,Austin CP, Brunden KR, Lee VM (2009) Identification ofaminothienopyridazine inhibitors of tau assembly by quan-titative high-throughput screening. Biochemistry 48, 7732-7745.

[1555] Honson NS, Jensen JR, Abraha A, Hall GF, Kuret J (2009)Small-molecule mediated neuroprotection in an in situmodel of tauopathy. Neurotox Res 15, 274-283.

[1556] Ballatore C, Brunden KR, Piscitelli F, James MJ, CroweA, Yao Y, Hyde E, Trojanowski JQ, Lee VM, Smith ABIII (2010) Discovery of brain-penetrant, orally bioavail-able aminothienopyridazine inhibitors of tau aggregation.J Med Chem 53, 3739-3747.

[1557] Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E(2010) Tau protein and tau aggregation inhibitors. Neu-ropharmacology 59, 276-289.

[1558] Schafer KN, Murale DP, Kim K, Cisek K, Kuret J,Churchill DG (2011) Structure-activity relationship ofcyclic thiacarbocyanine tau aggregation inhibitors. BioorgMed Chem Lett 21, 3273-3276.

[1559] Martin L, Magnaudeix A, Wilson CM, Yardin C, Terro F(2011) The new indirubin derivative inhibitors of glycogensynthase kinase-3, 6-BIDECO and 6-BIMYEO, preventtau phosphorylation and apoptosis induced by the inhibi-tion of protein phosphatase-2A by okadaic acid in culturedneurons. J Neurosci Res 89, 1802-1811.

[1560] Ballatore C, Crowe A, Piscitelli F, James M, Lou K, Ros-sidivito G, Yao Y, Trojanowski JQ, Lee VM, Brunden KR,Smith AB III (2012) Aminothienopyridazine inhibitors oftau aggregation: Evaluation of structure-activity relation-ship leads to selection of candidates with desirable in vivoproperties. Bioorg Med Chem 20, 4451-4461.

[1561] Ballatore C, Brunden KR, Huryn DM, Trojanowski JQ,Lee VM, Smith AB III (2012) Microtubule stabilizingagents as potential treatment for Alzheimer’s disease andrelated neurodegenerative tauopathies. J Med Chem 55,8979-8996.

[1562] Castro A, Martinez A (2000) Inhibition of tau phospho-rylation: A new therapeutic strategy for the treatment ofAlzheimer’s disease and other neurodegenerative disor-ders. Expert Opin Ther Patents 10, 1519-1527.

[1563] Lau LF, Schachter JB, Seymour PA, Sanner MA (2002)Tau protein phosphorylation as a therapeutic target inAlzheimer’s disease. Curr Top Med Chem 2, 395-415.

[1564] Dickey CA, Petrucelli L (2006) Current strategies for thetreatment of Alzheimer’s disease and other tauopathies.Expert Opin Ther Targets 10, 665-676.

Page 108: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

108 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1565] Ballatore C, Lee VM, Trojanowski JQ (2007) Tau-mediated neurodegeneration in Alzheimer’s disease andrelated disorders. Nat Rev Neurosci 8, 663-672.

[1566] Gong CX, Iqbal K (2008) Hyperphosphorylation ofmicrotubule-associated protein tau: A promising thera-peutic target for Alzheimer disease. Curr Med Chem 15,2321-2328.

[1567] Honson NS, Kuret J (2008) Tau aggregation and toxicityin tauopathic neurodegenerative diseases. J Alzheimers Dis14, 417-422.

[1568] Iqbal K, Grundke-Iqbal I (2008) Alzheimer neurofibrillarydegeneration: Significance, etiopathogenesis, therapeuticsand prevention. J Cell Mol Med 12, 38-55.

[1569] Schneider A, Mandelkow E (2008) Tau-based treatmentstrategies in neurodegenerative diseases. Neurotherapeu-tics 5, 443-457.

[1570] Brunden KR, Trojanowski JQ, Lee VM (2009) Advancesin tau-focused drug discovery for Alzheimer’s disease andrelated tauopathies. Nat Rev Drug Discov 8, 783-793.

[1571] Hanger DP, Anderton BH, Noble W (2009) Tau phospho-rylation: The therapeutic challenge for neurodegenerativedisease. Trends Mol Med 15, 112-119.

[1572] Brunden KR, Ballatore C, Crowe A, Smith AB III, LeeVM, Trojanowski JQ (2010) Tau-directed drug discoveryfor Alzheimer’s disease and related tauopathies: A focuson tau assembly inhibitors. Exp Neurol 223, 304-310.

[1573] Gong CX, Grundke-Iqbal I, Iqbal K (2010) Targeting tauprotein in Alzheimer’s disease. Drugs Aging 27, 351-365.

[1574] Meraz-Rios MA, Lira-De Leon KI, Campos-Pena V,De Anda-Hernandez MA, Mena-Lopez R (2010) Tauoligomers and aggregation in Alzheimer’s disease. J Neu-rochem 112, 1353-1367.

[1575] Ballatore C, Brunden KR, Trojanowski JQ, Lee VM, SmithAB, Huryn DM (2011) Modulation of protein-proteininteractions as a therapeutic strategy for the treatment ofneurodegenerative tauopathies. Curr Top Med Chem 11,317-330.

[1576] Corbo CP, Alonso AC (2011) Therapeutic targets inAlzheimer’s disease and related tauopathies. Prog Mol BiolTransl Sci 98, 47-83.

[1577] Noble W, Pooler AM, Hanger DP (2011) Advances in tau-based drug discovery. Expert Opin Drug Discov 6, 797-810.

[1578] Fuentes P, Catalan J (2011) A clinical perspective: Antitau’s treatment in Alzheimer’s disease. Curr Alzheimer Res8, 686-688.

[1579] Himmelstein DS, Ward SM, Lancia JK, Patterson KR,Binder LI (2012) Tau as a therapeutic target in neurode-generative disease. Pharmacol Ther 136, 8-22.

[1580] Ward SM, Himmelstein DS, Lancia JK, Binder LI (2012)Tau oligomers and tau toxicity in neurodegenerative dis-ease. Biochem Soc Trans 40, 667-671.

[1581] Karakaya T, Fusser F, Prvulovic D, Hampel H (2012)Treatment options for tauopathies. Curr Treat OptionsNeurol 14, 126-136.

[1582] Miller Y, Ma B, Nussinov R (2011) Synergistic interactionsbetween repeats in tau protein and Abeta amyloids may beresponsible for accelerated aggregation via polymorphicstates. Biochemistry 50, 5172-5181.

[1583] Zhang W, Arteaga J, Cashion DK, Chen G, Gangadhar-math U, Gomez LF, Kasi D, Lam C, Liang Q, Liu C,Mocharla VP, Mu F, Sinha A, Szardenings AK, WangE, Walsh JC, Xia C, Yu C, Zhao T, Kolb HC (2012) Ahighly selective and specific PET tracer for imaging of taupathologies. J Alzheimers Dis 31, 601-612.

[1584] Fodero-Tavoletti MT, Okamura N, Furumoto S, Mulli-gan RS, Connor AR, McLean CA, Cao D, RigopoulosA, Cartwright GA, O’Keefe G, Gong S, Adlard PA, Barn-ham KJ, Rowe CC, Masters CL, Kudo Y, Cappai R, YanaiK, Villemagne VL (2011) 18F-THK523: A novel in vivotau imaging ligand for Alzheimer’s disease. Brain 134,1089-1100.

[1585] Okamura N, Furumoto S, Harada R, Furukawa K, Arai H,Yanai K, Kudo Y (2011) Phenylquinoline derivatives forin vivo imaging of tau pathology in Alzheimer’s disease.Alzheimers Dement 7 (Suppl), e38.

[1586] Jones C, Nairne J (2012) Use of cyanine dyes for the detec-tion of tau for diagnosis of early-stage tauopathies. WO2012/068072, Prior. Nov. 19, 2010.

[1587] Watanabe H, Ono M, Kimura H, Matsumura K, YoshimuraM, Okamoto Y, Ihara M, Takahashi R, Saji H (2012)Synthesis and biological evaluation of novel oxin-dole derivatives for imaging neurofibrillary tangles inAlzheimer’s disease. Bioorg Med Chem Lett 22, 5700-5703.

[1588] Bolander A, Kieser D, Voss C, Bauer S, Schon C, Bur-gold S, Bittner T, Holzer J, Heyny-von HR, Mall G,Goetschy V, Czech C, Knust H, Berger R, Herms J, HilgerI, Schmidt B (2012) Bis(arylvinyl)pyrazines, -pyrimidines,and -pyridazines as imaging agents for tau fibrils and beta-amyloid plaques in Alzheimer’s disease models. J MedChem 55, 9170-9180.

[1589] Jensen JR, Cisek K, Funk KE, Naphade S, Schafer KN,Kuret J (2011) Research towards tau imaging. J AlzheimersDis 26(Suppl 3), 147-157.

[1590] Jensen JR, Cisek K, Honson NS, Kuret J (2011) Lig-and polarizability contributes to tau fibril binding affinity.Bioorg Med Chem 19, 5147-5154.

[1591] Boimel M, Grigoriadis N, Lourbopoulos A, Haber E,Abramsky O, Rosenmann H (2010) Efficacy and safety ofimmunization with phosphorylated tau against neurofib-rillary tangles in mice. Exp Neurol 224, 472-485.

[1592] Vana L, Kanaan NM, Ugwu IC, Wuu J, Mufson EJ,Binder LI (2011) Progression of tau pathology in choliner-gic Basal forebrain neurons in mild cognitive impairmentand Alzheimer’s disease. Am J Pathol 179, 2533-2550.

[1593] Patterson KR, Remmers C, Fu Y, Brooker S, KanaanNM, Vana L, Ward S, Reyes JF, Philibert K, GlucksmanMJ, Binder LI (2011) Characterization of prefibrillar Tauoligomers in vitro and in Alzheimer disease. J Biol Chem286, 23063-23076.

[1594] Rosenmann H, Meiner Z, Geylis V, Abramsky O, SteinitzM (2006) Detection of circulating antibodies against tauprotein in its unphosphorylated and in its neurofibrillarytangles-related phosphorylated state in Alzheimer’s dis-ease and healthy subjects. Neurosci Lett 410, 90-93.

[1595] Yen SH, Crowe A, Dickson DW (1985) Monoclonalantibodies to Alzheimer neurofibrillary tangles. 1. Iden-tification of polypeptides. Am J Pathol 120, 282-291.

[1596] Taniguchi T, Sumida M, Hiraoka S, Tomoo K, Kakehi T,Minoura K, Sugiyama S, Inaka K, Ishida T, Saito N, TanakaC (2005) Effects of different anti-tau antibodies on tau fib-rillogenesis: RTA-1 and RTA-2 counteract tau aggregation.FEBS Lett 579, 1399-1404.

[1597] Asuni AA, Boutajangout A, Quartermain D, SigurdssonEM (2007) Immunotherapy targeting pathological tau con-formers in a tangle mouse model reduces brain pathologywith associated functional improvements. J Neurosci 27,9115-9129.

Page 109: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 109

[1598] Sigurdsson EM (2008) Immunotherapy targeting patho-logical tau protein in Alzheimer’s disease and relatedtauopathies. J Alzheimers Dis 15, 157-168.

[1599] Zilka N, Kontsekova E, Novak M (2008) Chaperone-likeantibodies targeting misfolded tau protein: New vistas inthe immunotherapy of neurodegenerative foldopathies. JAlzheimers Dis 15, 169-179.

[1600] Kayed R, Jackson GR (2009) Prefilament tau species aspotential targets for immunotherapy for Alzheimer diseaseand related disorders. Curr Opin Immunol 21, 359-363.

[1601] Sigurdsson EM (2009) Tau-focused immunotherapyfor Alzheimer’s disease and related tauopathies. CurrAlzheimer Res 6, 446-450.

[1602] Boutajangout A, Quartermain D, Sigurdsson EM (2010)Immunotherapy targeting pathological tau prevents cogni-tive decline in a new tangle mouse model. J Neurosci 30,16559-16566.

[1603] Kayed R (2010) Anti-tau oligomers passive vaccinationfor the treatment of Alzheimer disease. Hum Vaccin 6,931-935.

[1604] Boutajangout A, Ingadottir J, Davies P, SigurdssonEM (2011) Passive immunization targeting pathologicalphospho-tau protein in a mouse model reduces functionaldecline and clears tau aggregates from the brain. J Neu-rochem 118, 658-667.

[1605] Boutajangout A, Sigurdsson EM, Krishnamurthy PK(2011) Tau as a therapeutic target for Alzheimer’s disease.Curr Alzheimer Res 8, 666-677.

[1606] Gu J, Sigurdsson EM (2011) Immunotherapy fortauopathies. J Mol Neurosci 45, 690-695.

[1607] Krishnamurthy PK, Deng Y, Sigurdsson EM (2011) Mech-anistic studies of antibody-mediated clearance of tauaggregates using an ex vivo brain slice model. Front Psy-chiatry 2, 59.

[1608] Mattson MP (2000) Emerging neuroprotective strategiesfor Alzheimer’s disease: Dietary restriction, telomeraseactivation, and stem cell therapy. Exp Gerontol 35, 489-502.

[1609] Sugaya K, Brannen CL (2001) Stem cell strategies forneuroreplacement therapy in Alzheimer’s disease. MedHypotheses 57, 697-700.

[1610] Tanne JH (2005) Activating stem cells may treatAlzheimer’s. BMJ 330, 622.

[1611] Wang QH, Xu RX, Nagao S (2005) Transplantationof cholinergic neural stem cells in a mouse model ofAlzheimer’s disease. Chin Med J (Engl) 118, 508-511.

[1612] Heese K, Low JW, Inoue N (2006) Nerve growth factor,neural stem cells and Alzheimer’s disease. Neurosignals15, 1-12.

[1613] Wang Q, Matsumoto Y, Shindo T, Miyake K, Shindo A,Kawanishi M, Kawai N, Tamiya T, Nagao S (2006) Neuralstem cells transplantation in cortex in a mouse model ofAlzheimer’s disease. J Med Invest 53, 61-69.

[1614] Sugaya K, Alvarez A, Marutle A, Kwak YD, Choumk-ina E (2006) Stem cell strategies for Alzheimer’s diseasetherapy. Panminerva Med 48, 87-96.

[1615] Fullwood NJ (2007) Neural stem cells, acetylcholine andAlzheimer’s disease. Nat Chem Biol 3, 435.

[1616] Sugaya K, Kwak YD, Ohmitsu O, Marutle A, Greig NH,Choumrina E (2007) Practical issues in stem cell ther-apy for Alzheimer’s disease. Curr Alzheimer Res 4, 370-377.

[1617] Sugaya K, Merchant S (2008) How to approachAlzheimer’s disease therapy using stem cell technologies.J Alzheimers Dis 15, 241-254.

[1618] Waldau B, Shetty AK (2008) Behavior of neural stem cellsin the Alzheimer brain. Cell Mol Life Sci 65, 2372-2384.

[1619] Lee JK, Jin HK, Bae JS (2009) Bone marrow-derived mes-enchymal stem cells reduce brain amyloid-beta depositionand accelerate the activation of microglia in an acutelyinduced Alzheimer’s disease mouse model. Neurosci Lett450, 136-141.

[1620] Feng Z, Zhao G, Yu L (2009) Neural stem cellsand Alzheimer’s disease: Challenges and hope. Am JAlzheimer’s Dis Other Demen 24, 52-57.

[1621] Taupin P (2009) Adult neurogenesis and the pathogenesisof Alzheimer’s disease. Med Sci Monit 15, LE1.

[1622] Taupin P (2009) Adult neurogenesis, neural stem cells andAlzheimer’s disease: Developments, limitations, problemsand promises. Curr Alzheimer Res 6, 461-470.

[1623] Dantuma E, Merchant S, Sugaya K (2010) Stem cells forthe treatment of neurodegenerative diseases. Stem Cell ResTher 1, 37.

[1624] Habisch HJ, Schmid B, von Arnim CA, Ludolph AC, Bren-ner R, Storch A (2010) Efficient processing of Alzheimer’sdisease amyloid-beta peptides by neuroectodermally con-verted mesenchymal stem cells. Stem Cells Dev 19,629-633.

[1625] Lee HJ, Lee JK, Lee H, Shin JW, Carter JE, Sakamoto T,Jin HK, Bae JS (2010) The therapeutic potential of humanumbilical cord blood-derived mesenchymal stem cells inAlzheimer’s disease. Neurosci Lett 481, 30-35.

[1626] Lee JK, Jin HK, Endo S, Schuchman EH, Carter JE, BaeJS (2010) Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduces amyloid-betadeposition and rescues memory deficits in Alzheimer’sdisease mice by modulation of immune responses. StemCells 28, 329-343.

[1627] Stellos K, Panagiota V, Sachsenmaier S, Trunk T, StratenG, Leyhe T, Seizer P, Geisler T, Gawaz M, Laske C (2010)Increased circulating progenitor cells in Alzheimer’s dis-ease patients with moderate to severe dementia: Evidencefor vascular repair and tissue regeneration? J AlzheimersDis 19, 591-600.

[1628] Taupin P (2010) Adult neurogenesis and neural stem cellsas a model for the discovery and development of noveldrugs. Expert Opin Drug Discov 5, 921-925.

[1629] Taupin P (2010) Aging and neurogenesis, a lesion fromAlzheimer’s disease. Aging Dis 1, 158-168.

[1630] Schwarz SC, Schwarz J (2010) Translation of stem celltherapy for neurological diseases. Transl Res 156, 155-160.

[1631] Couillard-Despres S, Iglseder B, Aigner L (2011) Neu-rogenesis, cellular plasticity and cognition: The impactof stem cells in the adult and aging brain–a mini-review.Gerontology 57, 559-564.

[1632] Lunn JS, Sakowski SA, Hur J, Feldman EL (2011) Stemcell technology for neurodegenerative diseases. Ann Neu-rol 70, 353-361.

[1633] Taupin P (2011) Neurogenesis, NSCs, pathogenesis andtherapies for Alzheimer’s disease. Front Biosci (Schol Ed)3, 178-190.

[1634] Abdel-Salam OM (2011) Stem cell therapy forAlzheimer’s disease. CNS Neurol Disord Drug Targets10, 459-485.

[1635] Chen C, Xiao SF (2011) Induced pluripotent stem cells andneurodegenerative diseases. Neurosci Bull 27, 107-114.

[1636] Borlongan CV (2012) Recent preclinical evidence advanc-ing cell therapy for Alzheimer’s disease. Exp Neurol 237,142-146.

Page 110: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

110 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1637] Rachubinski AL, Maclean KN, Evans JR, Bjugstad KB(2012) Modulating cognitive deficits and tau accumulationin a mouse model of aging down syndrome through neona-tal implantation of neural progenitor cells. Exp Gerontol47, 723-733.

[1638] Rachubinski AL, Crowley SK, Sladek Jr, Maclean KN,Bjugstad KB (2012) Effects of neonatal neural progeni-tor cell implantation on adult neuroanatomy and cognitionin the Ts65Dn model of Down syndrome. PLoS One 7,e36082.

[1639] Njie e, Kantorovich S, Astary GW, Green C, Zheng T,Semple-Rowland SL, Steindler DA, Sarntinoranont M,Streit WJ, Borchelt DR (2012) A preclinical assessmentof neural stem cells as delivery vehicles for anti-amyloidtherapeutics. PLoS One 7, e34097.

[1640] Khandekar N, Lie KH, Sachdev PS, Sidhu KS (2012)Amyloid precursor proteins, neural differentiation ofpluripotent stem cells and its relevance to Alzheimer’sdisease. Stem Cells Dev 21, 997-1006.

[1641] Feng Z, Gao F (2012) Stem cell challenges in the treatmentof neurodegenerative disease. CNS Neurosci Ther 18, 142-148.

[1642] Kitiyanant N, Kitiyanant Y, Svendsen CN, Thangnipon W(2012) BDNF-, IGF-1- and GDNF-secreting human neuralprogenitor cells rescue amyloid beta-induced toxicity incultured rat septal neurons. Neurochem Res 37, 143-152.

[1643] Park D, Joo SS, Kim TK, Lee SH, Kang H, Lee HJ, Lim I,Matsuo A, Tooyama I, Kim YB, Kim SU (2012) Humanneural stem cells overexpressing choline acetyltransferaserestore cognitive function of kainic acid-induced learningand memory deficit animals. Cell Transplant 21, 365-371.

[1644] Park D, Lee HJ, Joo SS, Bae DK, Yang G, Yang YH, LimI, Matsuo A, Tooyama I, Kim YB, Kim SU (2012) Humanneural stem cells over-expressing choline acetyltransferaserestore cognition in rat model of cognitive dysfunction.Exp Neurol 234, 521-526.

[1645] Kern DS, Maclean KN, Jiang H, Synder EY, Sladek JR,Bjugstad KB (2011) Neural stem cells reduce hippocam-pal tau and reelin accumulation in aged Ts65Dn downsyndrome mice. Cell Transplant 20, 371-379.

[1646] Xuan AG, Long DH, Gu HG, Yang DD, Hong LP, Leng SL(2008) BDNF improves the effects of neural stem cells onthe rat model of Alzheimer’s disease with unilateral lesionof fimbria-fornix. Neurosci Lett 440, 331-335.

[1647] Xuan AG, Luo M, Ji WD, Long DH (2009) Effects ofengrafted neural stem cells in Alzheimer’s disease rats.Neurosci Lett 450, 167-171.

[1648] Blurton-Jones M, Kitazawa M, Martinez-Coria H, CastelloNA, Muller FJ, Loring JF, Yamasaki TR, Poon WW, GreenKN, LaFerla FM (2009) Neural stem cells improve cogni-tion via BDNF in a transgenic model of Alzheimer disease.Proc Natl Acad Sci U S A 106, 13594-13599.

[1649] Lee HJ, Lim IJ, Park SW, Ko YB, Kim SU (2012)Human neural stem cells genetically modified to expresshuman nerve growth factor (NGF) gene restore cognitionin mouse with ibotenic acid-induced cognitive dys-function. Cell Transplant, doi: http://dx.doi.org/10.3727/096368912X638964

[1650] Lee HJ, Lim IJ, Lee MC, Kim SU (2010) Human neuralstem cells genetically modified to overexpress brain-derived neurotrophic factor promote functional recoveryand neuroprotection in a mouse stroke model. J NeurosciRes 88, 3282-3294.

[1651] Wu S, Sasaki A, Yoshimoto R, Kawahara Y, Manabe T,Kataoka K, Asashima M, Yuge L (2008) Neural stem cells

improve learning and memory in rats with Alzheimer’sdisease. Pathobiology 75, 186-194.

[1652] Wu L, Sluiter AA, Guo HF, Balesar RA, Swaab DF, ZhouJN, Verwer RW (2008) Neural stem cells improve neu-ronal survival in cultured postmortem brain tissue fromaged and Alzheimer patients. J Cell Mol Med 12, 1611-1621.

[1653] Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T,Yamanaka S, Okano H, Suzuki N (2011) Modeling familialAlzheimer’s disease with induced pluripotent stem cells.Hum Mol Genet 20, 4530-4539.

[1654] Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, HerreraC, Hefferan MP, Van GS, Nazor KL, Boscolo FS, CarsonCT, Laurent LC, Marsala M, Gage FH, Remes AM, KooEH, Goldstein LS (2012) Probing sporadic and familialAlzheimer’s disease using induced pluripotent stem cells.Nature 482, 216-220.

[1655] Choi SH, Tanzi RE (2012) iPSCs to the rescue inAlzheimer’s research. Cell Stem Cell 10, 235-236.

[1656] Shi Y, Kirwan P, Smith J, MacLean G, Orkin SH, LiveseyFJ (2012) A human stem cell model of early Alzheimer’sdisease pathology in Down syndrome. Sci Transl Med 4,124ra29.

[1657] Camnasio S, Carri AD, Lombardo A, Grad I, MariottiC, Castucci A, Rozell B, Riso PL, Castiglioni V, Zuccato C,Rochon C, Takashima Y, Diaferia G, Biunno I, Gellera C,Jaconi M, Smith A, Hovatta O, Naldini L, Di DS, Feki A,Cattaneo E (2012) The first reported generation of severalinduced pluripotent stem cell lines from homozygous andheterozygous Huntington’s disease patients demonstratesmutation related enhanced lysosomal activity. NeurobiolDis 46, 41-51.

[1658] Sadan O, Bahat-Stromza M, Barhum Y, Levy YS, Pis-nevsky A, Peretz H, Ilan AB, Bulvik S, Shemesh N, KrepelD, Cohen Y, Melamed E, Offen D (2009) Protective effectsof neurotrophic factor-secreting cells in a 6-OHDA ratmodel of Parkinson disease. Stem Cells Dev 18, 1179-1190.

[1659] Sadan O, Shemesh N, Barzilay R, Bahat-Stromza M,Melamed E, Cohen Y, Offen D (2008) Migration ofneurotrophic factors-secreting mesenchymal stem cellstoward a quinolinic acid lesion as viewed by magneticresonance imaging. Stem Cells 26, 2542-2551.

[1660] McGill TJ, Cottam B, Lu B, Wang S, Girman S, Tian C,Huhn SL, Lund RD, Capela A (2012) Transplantation ofhuman central nervous system stem cells - neuroprotectionin retinal degeneration. Eur J Neurosci 35, 468-477.

[1661] Lu P, Wang Y, Graham L, McHale K, Gao M, Wu D, BrockJ, Blesch A, Rosenzweig ES, Havton LA, Zheng B, Con-ner JM, Marsala M, Tuszynski MH (2012) Long-distancegrowth and connectivity of neural stem cells after severespinal cord injury. Cell 150, 1264-1273.

[1662] Glass JD, Boulis NM, Johe K, Rutkove SB, Federici T,Polak M, Kelly C, Feldman EL (2012) Lumbar intraspinalinjection of neural stem cells in patients with amyotrophiclateral sclerosis: Results of a phase I trial in 12 patients.Stem Cells 30, 1144-1151.

[1663] Borlongan CV, Skinner SJ, Geaney M, Vasconcellos AV,Elliott RB, Emerich DF (2004) CNS grafts of rat choroidplexus protect against cerebral ischemia in adult rats. Neu-roreport 15, 1543-1547.

[1664] Borlongan CV, Skinner SJ, Geaney M, Vasconcellos AV,Elliott RB, Emerich DF (2004) Neuroprotection by encap-sulated choroid plexus in a rodent model of Huntington’sdisease. Neuroreport 15, 2521-2525.

Page 111: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 111

[1665] Brinton RD, Wang JM (2006) Therapeutic potential of neu-rogenesis for prevention and recovery from Alzheimer’sdisease: Allopregnanolone as a proof of concept neuro-genic agent. Curr Alzheimer Res 3, 185-190.

[1666] Wang JM, Singh C, Liu L, Irwin RW, Chen S, ChungEJ, Thompson RF, Brinton RD (2010) Allopregnanolonereverses neurogenic and cognitive deficits in mouse modelof Alzheimer’s disease. Proc Natl Acad Sci U S A 107,6498-6503.

[1667] Singh C, Liu L, Wang JM, Irwin RW, Yao J, Chen S, HenryS, Thompson RF, Brinton RD (2012) Allopregnanolonerestores hippocampal-dependent learning and memory andneural progenitor survival in aging 3xTgAD and nonTgmice. Neurobiol Aging 33, 1493-1506.

[1668] Tsai KJ, Tsai YC, Shen CK (2007) G-CSF rescues thememory impairment of animal models of Alzheimer’s dis-ease. J Exp Med 204, 1273-1280.

[1669] Zhang XZ, Li XJ, Zhang HY (2010) Valproic acid as apromising agent to combat Alzheimer’s disease. Brain ResBull 81, 3-6.

[1670] Ling S, Zhou J, Rudd JA, Hu Z, Fang M (2011) Therecent updates of therapeutic approaches against abeta forthe treatment of Alzheimer’s disease. Anat Rec (Hoboken)294, 1307-1318.

[1671] Ulloa CM, Towfigh A, Safdieh J (2009) Review oflevetiracetam, with a focus on the extended release for-mulation, as adjuvant therapy in controlling partial-onsetseizures. Neuropsychiatr Dis Treat 5, 467-476.

[1672] Lyseng-Williamson KA (2011) Levetiracetam: A reviewof its use in epilepsy. Drugs 71, 489-514.

[1673] Lyseng-Williamson KA (2011) Spotlight on levetiracetamin epilepsy. CNS Drugs 25, 901-905.

[1674] Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P,Bajjalieh SM, Matagne A, Fuks B (2004) The synapticvesicle protein SV2A is the binding site for the antiepilep-tic drug levetiracetam. Proc Natl Acad Sci U S A 101, 9861-9866.

[1675] Lambeng N, Gillard M, Vertongen P, Fuks B, ChatelainP (2005) Characterization of [(3)H]ucb 30889 binding tosynaptic vesicle protein 2A in the rat spinal cord. Eur JPharmacol 520, 70-76.

[1676] Gillard M, Chatelain P, Fuks B (2006) Binding characteris-tics of levetiracetam to synaptic vesicle protein 2A (SV2A)in human brain and in CHO cells expressing the humanrecombinant protein. Eur J Pharmacol 536, 102-108.

[1677] Dong M, Yeh F, Tepp WH, Dean C, Johnson EA, JanzR, Chapman ER (2006) SV2 is the protein receptor forbotulinum neurotoxin A. Science 312, 592-596.

[1678] Yeh FL, Dong M, Yao J, Tepp WH, Lin G, Johnson EA,Chapman ER (2010) SV2 mediates entry of tetanus neu-rotoxin into central neurons. PLoS Pathog 6, e1001207.

[1679] Vogl C, Mochida S, Wolff C, Whalley BJ, Stephens GJ(2012) The synaptic vesicle glycoprotein 2A ligand leve-tiracetam inhibits presynaptic Ca2+ channels through anintracellular pathway. Mol Pharmacol 82, 199-208.

[1680] Angehagen M, Margineanu DG, Ben-Menachem E,Ronnback L, Hansson E, Klitgaard H (2003) Leve-tiracetam reduces caffeine-induced Ca2+ transients andepileptiform potentials in hippocampal neurons. Neurore-port 14, 471-475.

[1681] Ueda Y, Doi T, Nagatomo K, Tokumaru J, Takaki M,Willmore LJ (2007) Effect of levetiracetam on molecularregulation of hippocampal glutamate and GABA trans-porters in rats with chronic seizures induced by amygdalarFeCl3 injection. Brain Res 1151, 55-61.

[1682] Noyer M, Gillard M, Matagne A, Henichart JP, WulfertE (1995) The novel antiepileptic drug levetiracetam (ucbL059) appears to act via a specific binding site in CNSmembranes. Eur J Pharmacol 286, 137-146.

[1683] Loscher W, Honack D, Bloms-Funke P (1996) The novelantiepileptic drug levetiracetam (ucb L059) induces alter-ations in GABA metabolism and turnover in discrete areasof rat brain and reduces neuronal activity in substantianigra pars reticulata. Brain Res 735, 208-216.

[1684] Sills GJ, Leach JP, Fraser CM, Forrest G, Patsalos PN,Brodie MJ (1997) Neurochemical studies with the novelanticonvulsant levetiracetam in mouse brain. Eur J Phar-macol 325, 35-40.

[1685] Celikyurt IK, Ulak G, Mutlu O, Akar FY, Mulayim S,Erden F, Komsuoglu SS (2012) Positive impact of lev-etiracetam on emotional learning and memory in naivemice. Life Sci 90, 185-189.

[1686] Sanchez PE, Zhu L, Verret L, Vossel KA, Orr AG, Cir-rito JR, Devidze N, Ho K, Yu GQ, Palop JJ, Mucke L(2012) Levetiracetam suppresses neuronal network dys-function and reverses synaptic and cognitive deficits in anAlzheimer’s disease model. Proc Natl Acad Sci U S A 109,E2895-E2903.

[1687] Cho JR, Koo DL, Joo EY, Yoon SM, Ju E, Lee J, KimDY, Hong SB (2012) Effect of levetiracetam monotherapyon background EEG activity and cognition in drug-naiveepilepsy patients. Clin Neurophysiol 123, 883-891.

[1688] Schiemann-Delgado J, Yang H, Loge CL, Stalvey TJ,Jones J, Legoff D, Mintz M (2012) A long-term open-labelextension study assessing cognition and behavior, tolera-bility, safety, and efficacy of adjunctive levetiracetam inchildren aged 4 to 16 years with partial-onset seizures.J Child Neurol 27, 80-89.

[1689] Schulze-Bonhage A (2011) Brivaracetam for the treatmentof epilepsy. Expert Opin Pharmacother 12, 1959-1966.

[1690] Stephen LJ, Brodie MJ (2011) Pharmacotherapy ofepilepsy: Newly approved and developmental agents. CNSDrugs 25, 89-107.

[1691] Gillard M, Fuks B, Leclercq K, Matagne A (2011) Bindingcharacteristics of brivaracetam, a selective, high affinitySV2A ligand in rat, mouse and human brain: Relationshipto anti-convulsant properties. Eur J Pharmacol 664, 36-44.

[1692] Zona C, Pieri M, Carunchio I, Curcio L, Klitgaard H,Margineanu DG (2010) Brivaracetam (ucb 34714) inhibitsNa(+) current in rat cortical neurons in culture. EpilepsyRes 88, 46-54.

[1693] Detrait ER, Leclercq K, Loscher W, Potschka H,Niespodziany I, Hanon E, Kaminski RM, Matagne A,Lamberty Y (2010) Brivaracetam does not alter spatiallearning and memory in both normal and amygdala-kindled rats. Epilepsy Res 91, 74-83.

[1694] Meador KJ, Gevins A, Leese PT, Otoul C, Loring DW(2011) Neurocognitive effects of brivaracetam, levetirac-etam, and lorazepam. Epilepsia 52, 264-272.

[1695] Selkoe DJ (2012) Preventing Alzheimer’s disease. Science337, 1488-1492.

[1696] Froestl W, Muhs A, Pfeifer A (2012) Cognitive Enhancers(Nootropics). Part 2: Drugs Interacting with Enzymes. JAlzheimers Dis 33, 547-658.

[1697] Morimoto K, Horio J, Satoh H, Sue L, Beach T, AritaS, Tooyama I, Konishi Y (2011) Expression profilesof cytokines in the brains of Alzheimer’s disease (AD)patients compared to the brains of non-demented patientswith and without increasing AD pathology. J AlzheimersDis 25, 59-76.

Page 112: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

112 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

[1698] Zhou W, Zhong GF, Roa Xr, Xie H, Zeng SG, ChiTY, Zou LB, Wu DH, Hu WH (2012) Identification ofaminopyridazine-derived antineuroinflammatory agentseffective in an Alzheimer’s mouse model. ACS Med ChemLett, dx.doi.org/10.1021/ml3001769-

[1699] Vom Berg J, Prokop S, Miller KR, Obst J, Kalin RE,Lopategui-Cabezas I, Wegner A, Mair F, Schipke CG,Peters O, Winter Y, Becher B, Heppner FL (2012)Inhibition of IL-12/IL-23 signaling reduces Alzheimer’sdisease-like pathology and cognitive decline. Nat Med,doi: 10.1038/nm.2965-

[1700] Chan D (2012) American Chemical Society - 33rdNational Medicinal Chemistry Symposium (NMCS).Drugs of the Future 37, 599-603.

[1701] Thompson AD, Scaglione KM, Prensner J, Gillies AT,Chinnaiyan A, Paulson HL, Jinwal UK, Dickey CA, Gest-wicki JE (2012) Analysis of the tau-associated proteomereveals that exchange of hsp70 for hsp90 is involved in taudegradation. ACS Chem Biol 7, 1677-1686.

[1702] Berridge MJ (2013) Dysregulation of neural calciumsignaling in Alzheimer disease, bipolar disorder andschizophrenia. Prion 7 [Epub ahead of print].

[1703] Corbett A, Pickett J, Burns A, Corcoran J, Dunnett SB,Edison P, Hagan JJ, Holmes C, Jones E, Katona C, KearnsI, Kehoe P, Mudher A, Passmore A, Shepherd N, WalshF, Ballard C (2012) Drug repositioning for Alzheimer’sdisease. Nat Rev Drug Discov 11, 833-846.

[1704] Gholamipour-Badie H, Naderi N, Khodagholi F,Shaerzadeh F, Motamedi F (2013) L-type calciumchannel blockade alleviates molecular and reversal spatiallearning and memory alterations induced by entorhinalamyloid pathology in rats. Behav Brain Res 237,190-199.

[1705] Mufson EJ, He B, Nadeem M, Perez SE, Counts SE,Leurgans S, Fritz J, Lah J, Ginsberg SD, Wuu J, ScheffSW (2012) Hippocampal ProNGF Signaling Pathways andbeta-Amyloid Levels in Mild Cognitive Impairment andAlzheimer Disease. J Neuropathol Exp Neurol 71, 1018-1029.

[1706] Bendheim PE, Poeggeler B, Neria E, Ziv V, Pappolla MA,Chain DG (2002) Development of indole-3-propionic acid(OXIGON) for Alzheimer’s disease. J Mol Neurosci 19,213-217.

[1707] Bush AI (2012) The Metal Theory of Alzheimer’s Disease.J Alzheimers Dis, doi: 10.3233/JAD-2012-129011-

[1708] Wei G, Yunbo C, Chen DF, Lai XP, Liu DH, DengRD, Zhou JH, Zhang SX, Li YW, Li H, Liu LF(2012) beta-Asarone Inhibits Neuronal Apoptosis via theCaMKII/CREB/Bcl-2 Signaling Pathway in an in vitroModel and AbetaPP/PS1 Mice. J Alzheimers Dis, doi:10.3233/JAD-2012-120865-

[1709] Brunhofer G, Fallarero A, Karlsson D, Batista-GonzalezA, Shinde P, Gopi MC, Vuorela P (2012) Explorationof natural compounds as sources of new bifunctionalscaffolds targeting cholinesterases and beta amyloid aggre-gation: The case of chelerythrine. Bioorg Med Chem 20,6669-6679.

[1710] Zhao X, Zou Y, Xu H, Fan L, Guo H, Li X, Li G, Zhang X,Dong M (2012) Gastrodin protect primary cultured rat hip-pocampal neurons against amyloid-beta peptide-inducedneurotoxicity via ERK1/2-Nrf2 pathway. Brain Res 1482,13-21.

[1711] Zhu X, Ye L, Ge H, Chen L, Jiang N, Qian L, Li L, Liu R,Ji S, Zhang S, Jin J, Guan D, Fang W, Tan R, Xu Y (2012)Hopeahainol A attenuates memory deficits by targeting

beta-amyloid in APP/PS1 transgenic mice. Aging Cell, doi:10.1111/acel.12022-

[1712] Zou Y, Hong B, Fan L, Zhou L, Liu Y, Wu Q,Zhang X, Dong M (2012) Protective effect of puerarinagainst beta-amyloid-induced oxidative stress in neu-ronal cultures from rat hippocampus: involvement ofthe GSK-3beta/Nrf2 signaling pathway. Free Radic Res,doi:10.3109/10715762.2012.742518-

[1713] Ho L, Ferruzzi MG, Janle EM, Wang J, Gong B, ChenTY, Lobo J, Cooper B, Wu QL, Talcott ST, Percival SS,Simon JE, Pasinetti GM (2012) Identification of brain-targeted bioactive dietary quercetin-3-O-glucuronide as anovel intervention for Alzheimer’s disease. FASEB J, doi:10.1096/fj.12-212118-

[1714] McCracken C, Hudson P, Ellis R, McCaddon A (2006)Methylmalonic acid and cognitive function in the MedicalResearch Council Cognitive Function and Ageing Study.Am J Clin Nutr 84, 1406-1411.

[1715] Moore E, Mander A, Ames D, Carne R, Sanders K, WattersD (2012) Cognitive impairment and vitamin B12: a review.Int Psychogeriatr, 1-16.

[1716] Llewellyn DJ, Langa KM, Lang IA (2009) Serum25-hydroxyvitamin D concentration and cognitive impair-ment. J Geriatr Psychiatry Neurol 22, 188-195.

[1717] Llewellyn DJ, Lang IA, Langa KM, Muniz-Terrera G,Phillips CL, Cherubini A, Ferrucci L, Melzer D (2010)Vitamin D and risk of cognitive decline in elderly persons.Arch Intern Med 170, 1135-1141.

[1718] Llewellyn DJ, Lang IA, Langa KM, Melzer D (2011)Vitamin D and cognitive impairment in the elderlyU.S. population. J Gerontol A Biol Sci Med Sci 66,59-65.

[1719] Wang L, Ankati H, Akubathini SK, Balderamos M, StoreyCA, Patel AV, Price V, Kretzschmar D, Biehl ER, D’MelloSR (2010) Identification of novel 1,4-benzoxazine com-pounds that are protective in tissue culture and in vivomodels of neurodegeneration. J Neurosci Res 88, 1970-1984.

[1720] Hampel H (2012) Amyloid-beta and Cognition inAging and Alzheimer’s Disease: Molecular and Neu-rophysiological Mechanisms. J Alzheimers Dis, doi:10.3233/JAD-2012-129003-

[1721] Lim YY, Pietrzak RH, Ellis KA, Jaeger J, Harrington K,Ashwood T, Szoeke C, Martins RN, Bush AI, MastersCL, Rowe CC, Villemagne VL, Ames D, Darby D, MaruffP (2012) Rapid Decline in Episodic Memory in HealthyOlder Adults with High Amyloid-beta. J Alzheimers Dis,doi: 10.3233/JAD-2012-121516-

[1722] Lim YY, Ellis KA, Pietrzak RH, Ames D, Darby D, Har-rington K, Martins RN, Masters CL, Rowe C, SavageG, Szoeke C, Villemagne VL, Maruff P (2012) Strongereffect of amyloid load than APOE genotype on cogni-tive decline in healthy older adults. Neurology 79, 1645-1652.

[1723] Lim YY, Ellis KA, Harrington K, Pietrzak RH, GaleJ, Ames D, Bush AI, Darby D, Martins RN, MastersCL, Rowe CC, Savage G, Szoeke C, Villemagne VL,Maruff P (2012) Cognitive Decline in Adults with Amnes-tic Mild Cognitive Impairment and High Amyloid-beta:Prodromal Alzheimer’s Disease? J Alzheimers Dis, doi:10.3233/JAD-121771-

[1724] Said G, Grippon S, Kirkpatrick P (2012) Tafamidis. NatRev Drug Discov 11, 185-186.

[1725] Echeverria V, Zeitlin R, Burgess S, Patel S, Barman A,Thakur G, Mamcarz M, Wang L, Sattelle DB, Kirschner

Page 113: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 113

DA, Mori T, Leblanc RM, Prabhakar R, ArendashGW (2011) Cotinine reduces amyloid-beta aggregationand improves memory in Alzheimer’s disease mice. JAlzheimers Dis 24, 817-835.

[1726] Ghosh S (2012) Alzheimer’s Association InternationalConference. Drugs of the Future 37, 663-670.

[1727] O’Hare E, Scopes DI, Kim EM, Palmer P, JonesM, Whyment AD, Spanswick D, Amijee H, NerouE, McMahon B, Treherne JM, Jeggo R (2012)Orally bioavailable small molecule drug protects mem-ory in Alzheimer’s disease models. Neurobiol Aging,doi:10.1016/j.neurobiolaging.2012.10.016-

[1728] McKoy AF, Chen J, Schupbach T, Hecht MH (2012) ANovel Inhibitor of Amyloid beta (Abeta) Peptide Aggre-gation: From high throughput screening to efficacy in ananimal model of Alzheimer disease 1. J Biol Chem 287,38992-39000.

[1729] Amijee H, Bate C, Williams A, Virdee J, Jeggo R, Span-swick D, Scopes DI, Treherne JM, Mazzitelli S, ChawnerR, Eyers CE, Doig AJ (2012) The N-Methylated Pep-tide SEN304 Powerfully Inhibits Abeta(1-42) Toxicityby Perturbing Oligomer Formation. Biochemistry, doi:10.1021/bi300415v-

[1730] Mathis CA, Mason NS, Lopresti BJ, Klunk WE (2012)Development of Positron Emission Tomography beta-Amyloid Plaque Imaging Agents. Semin Nucl Med 42,423-432.

[1731] Landau SM, Breault C, Joshi AD, Pontecorvo M, MathisCA, Jagust WJ, Mintun MA (2012) Amyloid-beta Imagingwith Pittsburgh Compound B and Florbetapir: ComparingRadiotracers and Quantification Methods. J Nucl Med, doi:10.2967/jnumed.112.109009-

[1732] Handen BL, Cohen AD, Channamalappa U, Bulova P,Cannon SA, Cohen WI, Mathis CA, Price JC, KlunkWE (2012) Imaging brain amyloid in nondemented youngadults with Down syndrome using Pittsburgh compoundB. Alzheimers Dement 8, 496-501.

[1733] Doraiswamy PM, Sperling RA, Coleman RE, Johnson KA,Reiman EM, Davis MD, Grundman M, Sabbagh MN, Sad-owsky CH, Fleisher AS, Carpenter A, Clark CM, JoshiAD, Mintun MA, Skovronsky DM, Pontecorvo MJ (2012)Amyloid-beta assessed by florbetapir F 18 PET and 18-month cognitive decline: A multicenter study. Neurology79, 1636-1644.

[1734] Furst AJ, Kerchner GA (2012) From Alois to Amyvid:Seeing Alzheimer disease. Neurology 79, 1628-1629.

[1735] Poisnel G, Dhilly M, Moustie O, Delamare J, Abbas A,Guilloteau D, Barre L (2012) PET imaging with [18F]AV-45 in an APP/PS1-21 murine model of amyloid plaquedeposition. Neurobiol Aging 33, 2561-2571.

[1736] Fodero-Tavoletti MT, Brockschnieder D, Villemagne VL,Martin L, Connor AR, Thiele A, Berndt M, McLean CA,Krause S, Rowe CC, Masters CL, Dinkelborg L, DyrksT, Cappai R (2012) In vitro characterization of [(18)F]-florbetaben, an Abeta imaging radiotracer. Nucl Med Biol39, 1042-1048.

[1737] Swahn BM, Sandell J, Pyring D, Bergh M, Jeppsson F,Jureus A, Neelissen J, Johnstrom P, Schou M, SvenssonS (2012) Synthesis and evaluation of pyridylbenzofuran,pyridylbenzothiazole and pyridylbenzoxazole derivativesas (1)(8)F-PET imaging agents for beta-amyloid plaques.Bioorg Med Chem Lett 22, 4332-4337.

[1738] Swahn BM, Wensbo D, Sandell J, Sohn D, Slivo C, PyringD, Malmstrom J, Arzel E, Vallin M, Bergh M, Jeppsson F,Johnson AE, Jureus A, Neelissen J, Svensson S (2010)

Synthesis and evaluation of 2-pyridylbenzothiazole, 2-pyridylbenzoxazole and 2-pyridylbenzofuran derivativesas 11C-PET imaging agents for beta-amyloid plaques.Bioorg Med Chem Lett 20, 1976-1980.

[1739] Nelson LD, Siddarth P, Kepe V, Scheibel KE, Huang SC,Barrio JR, Small GW (2011) Positron emission tomogra-phy of brain beta-amyloid and tau levels in adults withDown syndrome. Arch Neurol 68, 768-774.

[1740] Shin J, Kepe V, Barrio JR, Small GW (2011) The mer-its of FDDNP-PET imaging in Alzheimer’s disease. JAlzheimers Dis 26(Suppl 3), 135-145.

[1741] Montanes M, Casabona D, Sarasa L, Pesini P, Sarasa M(2012) Prevention of Amyloid-beta Fibril Formation UsingAntibodies Against the C-terminal Region of Amyloid-beta1-40 and Amyloid-beta1-42. J Alzheimers Dis, doi:10.3233/JAD-120850-

[1742] Bard F, Fox M, Friedrich S, Seubert P, Schenk D, Kin-ney GG, Yednock T (2012) Sustained levels of antibodiesagainst Abeta in amyloid-rich regions of the CNS follow-ing intravenous dosing in human APP transgenic mice.Exp Neurol 238, 38-43.

[1743] Solomon B (2007) Intravenous immunoglobulin andAlzheimer’s disease immunotherapy. Curr Opin Mol Ther9, 79-85.

[1744] Robert R, Wark KL (2012) Engineered antibodyapproaches for Alzheimer’s disease immunotherapy. ArchBiochem Biophys 526, 132-138.

[1745] Yu G, Li Y, Tian Q, Liu R, Wang Q, Wang JZ, Wang X(2011) Berberine attenuates calyculin A-induced cytotox-icity and Tau hyperphosphorylation in HEK293 cells. JAlzheimers Dis 24, 525-535.

[1746] Peng Y, Hu Y, Xu S, Li P, Li J, Lu L, Yang H, FengN, Wang L, Wang X (2012) L-3-n-butylphthalide reducestau phosphorylation and improves cognitive deficits inAbetaPP/PS1-Alzheimer’s transgenic mice. J AlzheimersDis 29, 379-391.

[1747] Tolstykh T, Lee J, Vafai S, Stock JB (2000) Carboxylmethylation regulates phosphoprotein phosphatase 2Aby controlling the association of regulatory B subunits.EMBO J 19, 5682-5691.

[1748] Vafai SB, Stock JB (2002) Protein phosphatase 2A methy-lation: A link between elevated plasma homocysteine andAlzheimer’s Disease. FEBS Lett 518, 1-4.

[1749] Xing Y, Li Z, Chen Y, Stock JB, Jeffrey PD, Shi Y (2008)Structural mechanism of demethylation and inactivationof protein phosphatase 2A. Cell 133, 154-163.

[1750] Butler KV, Kalin J, Brochier C, Vistoli G, Langley B,Kozikowski AP (2010) Rational design and simple chem-istry yield a superior, neuroprotective HDAC6 inhibitor,tubastatin A. J Am Chem Soc 132, 10842-10846.

[1751] Benner L, Kalin JH, Kozikowski A, Gordon MN, Mor-gan D, Selenica MB (2012) Selective HDAC6 inhibitiondecreases tau levels and improves memory deficits in thertg4510 mouse model. Society for Neuroscience, NewOrleans, Poster 150.04-Sunday, November 14, 2012.

[1752] Okamura N, Suemoto T, Furumoto S, Suzuki M, Shi-madzu H, Akatsu H, Yamamoto T, Fujiwara H, NemotoM, Maruyama M, Arai H, Yanai K, Sawada T, KudoY (2005) Quinoline and benzimidazole derivatives: Can-didate probes for in vivo imaging of tau pathology inAlzheimer’s disease. J Neurosci 25, 10857-10862.

[1753] Gu J, Anumala UR, Lo MF, Kramer T, Heyny vonHR, Holzer J, Goetschy-Meyer V, Mall G, HilgerI, Czech C, Schmidt B (2012) 2-Styrylindoliumbased fluorescent probes visualize neurofibrillary tan-

Page 114: Cognitive Enhancers (Nootropics). Part 3: Drugs ... · 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

114 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes

gles in Alzheimer’s disease. Bioorg Med Chem Lett,http://dx.doi.org/10.1016/j.bmcl.2012.09.109-

[1754] Chai X, Wu S, Murray TK, Kinley R, Cella CV, SimsH, Buckner N, Hanmer J, Davies P, O’Neill MJ, HuttonML, Citron M (2011) Passive immunization with anti-Tau antibodies in two transgenic models: reduction of Taupathology and delay of disease progression. J Biol Chem286, 34457-34467.

[1755] Shih HH, Tu C, Cao W, Klein A, Ramsay R, Fennell BJ,Lambert M, Ni SD, Autin B, Kouranova E, LaxmananS, Braithwaite S, Wu L, it-Zahra M, Milici AJ, DuminJA, Lavallie ER, Arai M, Corcoran C, Paulsen JE, Gill D,Cunningham O, Bard J, Mosyak L, Finlay WJ (2012) Anultra-specific avian antibody to phosphorylated tau revealsa unique mechanism for phosphoepitope recognition. JBiol Chem, doi: 10.1074/jbc.M112.415935-

[1756] Wang W, Fan L, Xu D, Wen Z, Yu R, Ma Q (2012)Immunotherapy for Alzheimer’s disease. Acta BiochimBiophys Sin (Shanghai) 44, 807-814.

[1757] Young JE, Goldstein LS (2012) Alzheimer’s disease in adish: promises and challenges of human stem cell models.Hum Mol Genet 21, R82-R89.

[1758] Grskovic M, Javaherian A, Strulovici B, Daley GQ (2011)Induced pluripotent stem cells–opportunities for diseasemodelling and drug discovery. Nat Rev Drug Discov 10,915-929.

[1759] Uchida N, Chen K, Dohse M, Hansen KD, Dean J, BuserJR, Riddle A, Beardsley DJ, Wan Y, Gong X, Nguyen T,Cummings BJ, Anderson AJ, Tamaki SJ, Tsukamoto A,Weissman IL, Matsumoto SG, Sherman LS, Kroenke CD,

Back SA (2012) Human neural stem cells induce func-tional myelination in mice with severe dysmyelination. SciTransl Med 4, 155ra136-

[1760] Gupta N, Henry RG, Strober J, Kang SM, Lim DA, BucciM, Caverzasi E, Gaetano L, Mandelli ML, Ryan T, PerryR, Farrell J, Jeremy RJ, Ulman M, Huhn SL, BarkovichAJ, Rowitch DH (2012) Neural stem cell engraftmentand myelination in the human brain. Sci Transl Med 4,155ra137-

[1761] Moreira PI, Santos RX, Zhu X, Lee HG, Smith MA,Casadesus G, Perry G (2010) Autophagy in Alzheimer’sdisease. Expert Rev Neurother 10, 1209-1218.

[1762] Barnett A, Brewer GJ (2011) Autophagy in agingand Alzheimer’s disease: Pathologic or protective? JAlzheimers Dis 25, 385-394.

[1763] Schaeffer V, Goedert M (2012) Stimulation of autophagyis neuroprotective in a mouse model of human tauopathy.Autophagy 8, http://dx.doi.org/10.4161/auto.21488-

[1764] Schaeffer V, Lavenir I, Ozcelik S, Tolnay M, Winkler DT,Goedert M (2012) Stimulation of autophagy reduces neu-rodegeneration in a mouse model of human tauopathy.Brain 135, 2169-2177.

[1765] Brown JR, Crawford BE, Esko JD (2007) Glycan antago-nists and inhibitors: A fount for drug discovery. Crit RevBiochem Mol Biol 42, 481-515.

[1766] Bakker A, Krauss GL, Albert MS, Speck CL, Jones LR,Stark CE, Yassa MA, Bassett SS, Shelton AL, Gallagher M(2012) Reduction of hippocampal hyperactivity improvescognition in amnestic mild cognitive impairment. Neuron74, 467-474.