Survey
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project
1 Journal of Alzheimer’s Disease 34 (2013) 1–114 DOI 10.3233/JAD-121729 IOS Press 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 Thomson 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 Corneliu 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 2 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes (and unconscious) mental activities, including preattentional sensory gating, attention, learning and memory, problem solving, planning, reasoning and judgment, understanding, knowing and representing, creativity, intuition and insight, spontaneous thought, introspection, as well as mental time travel, self-awareness and meta cognition (thinking and knowledge about cognition)”. Since a first review in 1989 on “Families of Cognition Enhancers” by Froestl and Maı̂tre [4], substantial progress has been made in the understanding of the mechanism(s) of cognitive enhancers. Therefore, we propose a new classification to assign cognition enhancing drugs to 19 categories: In Part 1, drugs interacting with receptors were described [5]. In Part 2, drugs interacting with enzymes were described [1696]. Here, in Part 3, we give an overview on drugs interacting with targets 3 to 10 and compounds and preparations of categories 11 to 19. Disease modifying drugs are aimed to counteract the progression of a disease. In particular for AD, many excellent reviews discuss this subject (in chronological order) [6–27]. ␣-secretase activators and - and ␥-secretase inhibitors (and modulators) were presented in Part 2 of this review. Drugs interacting with amyloid (A) and tau including immunotherapy are described in Part 3. DRUGS INTERACTING WITH CYTOKINES 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Drugs interacting with Receptors (Part 1) Drugs interacting with Enzymes (Part 2) Drugs interacting with Cytokines (Part 3) Drugs interacting with Gene Expression (Part 3) Drugs interacting with Heat Shock Proteins (Part 3) Drugs interacting with Hormones (Part 3) Drugs interacting with Ion Channels (nonReceptors) (Part 3) Drugs interacting with Nerve Growth Factors (Part 3) Drugs interacting with Re-uptake Transporters (Part 3) Drugs interacting with Transcription Factors (Part 3) Antioxidants (Part 3) Metal Chelators (Part 3) Natural Products (Part 3) Nootropics (“Drugs without mechanism”) (Part 3) Peptides (Part 3) Drugs preventing amyloid- aggregation (Part 3) 16.1. Ligands interacting with amyloid- (Part 3) 16.2. Inhibitors of serum amyloid P component binding (Part 3) 16.3. Vaccines against amyloid- (Part 3) 16.4. Antibodies against amyloid- (Part 3) Drugs interacting with tau (Part 3) 17.1. Small molecules preventing tau aggregation (Part 3) 17.2. Ligands interacting with tau (Part 3) 17.3. Vaccines against tau (Part 3) 17.4. Antibodies against tau (Part 3) Stem Cells (Part 3) Miscellaneous (Part 3) There is abundant evidence that inflammatory mechanisms within the central nervous system (CNS) contribute to cognitive impairment via cytokinemediated interactions between neurons and glial cells. A current hypothesis is that an extracellular insult to neurons could trigger the production of inflammatory cytokines by astrocytes and microglia [28]. Conversely, A has been shown to induce the expression of interleukin (IL)-1, tumor necrosis factor-␣ (TNF-␣) and IL-6 in astrocytes and microglia in culture [29]. One genome-wide analysis in 691 subjects of mean age of 72.6 years showed that raised chemokine (C-C motif) receptor 2 (CCR2) expression was the most strongly associated transcript with lower MiniMental Status Exam scores and accelerated decline in score over a period of 9 years [30]. Decline in score over a period of 9 years [30]. The expression profiles of cytokines in the brains of Alzheimer’s disease patients were compared to the brains of non-demented patients [1697]. Also see 11. Antioxidants, Also see 11. Antioxidants, because inflammation is tightly connected with the oxidative cascade. TT-301 (MW01-6-189WH; MW-189; Transition Therapeutics, Toronto following its acquisition of NeuroMedix under license from Northwestern University) is an inhibitor of microglial cell activation and proinflammatory cytokine production for the potential i.v. treatment of CNS diseases including AD. Preclinical characterization of the suppression of brain proinflammatory cytokine upregulation was carried out [31]. A double-blind, randomized, placebo-controlled Phase I clinical trial in healthy male subjects (n = 16) started in the US in May 2011 (Thomson Reuters Pharma, update of September 14, 2012). The structure was not communicated. 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; Transition Therapeutics, Toronto; Fig. 1) is an inhibitor of proinflammatory cytokine production by activated glia for the potential oral treatment of CNS disorders including AD, traumatic brain injury and inflammatory diseases such as arthritis [32]. Phase I clinical trials are expected for 2012 (Thomson Reuters Pharma, update of September 29, 2012). Minozac (MW01-2-151SRM, MW-151, Transition Therapeutics, Toronto following its acquisition of NeuroMedix, under license from Northwestern University) (Fig. 1) is an inhibitor of pro-inflammatory cytokine production by activated glia for the potential treatment of CNS diseases including AD, Parkinson’s disease (PD), multiple sclerosis, and traumatic brain injury [33–36] (Thomson Reuters Pharma, update of August 13, 2012). AD-16 (Guangzhou Institutes of Biomedicine and Health) is the thiophene analogue of Minozac. It reduced amyloid- induced spatial learning and memory impairment as potently as donepezil in an Alzheimer’s mouse model [1698]. (Thomson Reuters Pharma, update of October 19, 2012). SEN-1176 (Senexis, Cambridge, UK) (Fig. 1) is a pyrrolo[3,2-e][1,2,4]triazolo[1,5-a]pyrimidine, which suppresses A42 -induced macrophage production of nitric oxide, TNF-␣, IL-1, and IL-6 in a dosedependent fashion, an activity profile consistent with a neuroinflammation inhibitor [1711] (Thomson Reuters Pharma, update of June 24, 2011). Infliximab (Janssen Biotech, Horsham, PA), a launched monoclonal antibody against TNF-␣, improved cognition after intrathecal administration in a woman with AD [38]. The University of Zurich is investigating an antibody against the interleukin-12 subunit p40 for the potential treatment of Alzheimer’s disease [1699] (Thomson Reuters Pharma, update of November 26, 2012). The development of REN-1189 (formerly CPI1189; Centaur Pharmaceuticals), a TNF-␣ release inhibitor, was terminated. Also the development of Semapimod (CNI-1493; Cytokine PharmaSciences, CPSI, formerly Cytokine Networks), a cytokine inhibitor, which inhibited A production, plaque formation, and cognitive deterioration in an animal model of AD was suspended [39, 40]. 4 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes DRUGS INTERACTING WITH GENE EXPRESSION Gene therapy in AD, a potential for disease modification, was discussed [1715]. An interesting review on genes and the parsing of cognitive processes was communicated [42]. Bi-phasic change in brain-derived neurotropic factor (BDNF) gene expression following antidepressant drug treatment was described [43, 44]. The modulation of Nur77 and Nor-1 expression by dopaminergic drugs was elucidated [45]. The regulation of GABAA receptor subunit expression by pharmacological agents was investigated [46]. Gene therapy in mouse models of Huntington’s disease (HD) was reviewed [47]. Beperminogene perplasmid (AMG-0001; DS992; Collategene; AnGes MG, formerly MedGene Bioscience, Osaka, in collaboration with Daiichi Sankyo) is a hepatocyte growth factor (HGF) plasmid-based gene therapy for i.m. injection. It showed nootropic properties with a potential for the treatment of PD. In November 2011, a Phase III trial for peripheral arterial disease was expected to be initiated in 2012. In April 2012, the Phase III studies were still in preparation [48]. A long-term follow-up evaluation of results from the clinical trial TREAT-HGF was reported [49] (Thomson Reuters Pharma, update of September 19, 2012). RVX-208 (Resverlogix, Calgary) (Fig. 1) is an apolipoprotein A1 (ApoA1) gene expression stimulator for the potential prevention of A plaque accumulation in AD. In January 2011, data from an analysis of AD biomarkers in the Phase II ASSERT trial in 299 patients showed that after 12 weeks of treatment with 150 mg/day, a positive effect on A40 was seen. A compilation of data on RVX-208 was published in Drugs in R & D [50]. The rationale for the SUSTAIN study (172 patients) and the ASSURE study (310 patients) was presented [51]. Preclinical evaluations were reported [52, 53] and clinical data were provided [54–56]. RVX-208 significantly increased high-density lipoprotein (HDL)-C (p = 0.001), the primary endpoint of the SUSTAIN trial, a Phase 2b clinical study. SUSTAIN also successfully met secondary endpoints, showed increases in levels of Apo-AI (p = 0.002) and large HDL particles (p = 0.02), both believed to be important factors in enhancing reverse cholesterol transport activity. In July 2012, the company was planning to initiate a Phase II trial of RVX-208 in patients with mild cognitive impairment (MCI) in the second half of 2013 (Thomson Reuters Pharma, update of September 27, 2012). There are several drugs interacting with gene expression in preclinical evaluation (in alphabetical order): AAV-CYP46A1 (INSERM in collaboration with sanofi) is an adeno-associated virus (AAV) gene therapy encoding cholesterol 24-hydroxylase (CYP46A1 gene) for the potential injectable treatment of AD. Reduced A peptides, amyloid deposits, and trimeric oligomers were observed in APP23 mice. Significant improvements in cognitive function assessed by the Morris 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 with cholesterol 24-hydroxylase (CYP46A1). AZ-AAV9 (RegenX Biosciences, Washington DC) is an AAV vector-9 that carries neuroprotective genes for the potential injectable treatment of AD (Thomson Reuters Pharma, update of July 27, 2012). HSD17B10 is a gene, which encodes HSD10, a mitochondrial multifunctional enzyme that plays a significant part in the metabolism of neuroactive steroids and the degradation of isoleucine. Elevated levels of HSD10 were observed in the hippocampi of AD patients [58]. PRO-289 (Prosensa Therapeutics, Leiden, the Netherlands) is the lead from a series of single-stranded RNA-based antisense oligonucleotide-based therapeutics to inhibit the CTG trinucleotide repeat expansion of the huntingtin gene (HTT) by preventing the cellular production of aberrantly expanded HTT mRNA and mutant huntingtin protein (Thomson Reuters Pharma, update of February 10, 2012). SynCav (Raft Therapeutics, San Diego CA) is a gene therapy that upregulates synaptic driven Caveolin-1 to promote regeneration of neurons for the potential treatment of neurodegenerative diseases such as AD, PD, HD, and amyotrophic lateral sclerosis (ALS) (Thomson Reuters Pharma, update of August 10, 2012). Intrahippocampal gene transfer of F-spondin, an activator of the reelin pathway, improved spatial learning/memory in the Morris water maze and increased the exploration of the novel object in the Novel Object Recognition test in wild-type mice [59]. lnventiva (Daix, Bourgogne, France) is investigating compounds that increase expression of a target gene under epigenetic control resulting in increased levels of a secreted neuronal protein for the potential treatment of Alzheimer’s disease. (Thomson Reuters Pharma, update of November 23, 2012). Structures were not communicated. Neurologix under license from Keio University was investigating colivelin, a hybrid peptide composed of W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes activity-dependent neurotrophic factor [60–64] and a humanin derivative for the potential gene therapy of AD [65, 66]. The program was terminated. The development of LX-6171 (Lexicon Pharmaceuticals, formerly Lexicon Genetics), a small molecule inhibitor of the SLC6A gene and the LG617 receptor, was discontinued as well. DRUGS INTERACTING WITH HEAT SHOCK PROTEINS Binding of heat shock protein 90 (HsP90) to tau facilitates a conformational change that results in its phosphorylation by glycogen synthase kinase 3 (GSK-3) and its aggregation into filamentous structures [67]. HsP90 regulates tau pathology through co-chaperone complexes [68]. HsP90 inhibitors, such as PU-H71, PU-3, PU24FCl (Fig. 2) and PU-DZ8 of the purine class of HsP90 inhibitors from the Memorial Sloan-Kettering Institute of Cancer Research, New York caused an elimination of aggregated tau [69–75]. 5 PU-H71 is in Phase I clinical trials since July 2011 (n = 40) in the US. The study was expected to be complete by July 2013 (Thomson Reuters Pharma, update of April 24, 2012). ALS Biopharma (Doylestown, PA) is investigating small-molecule brain-penetrant modulators of HsP70 for the potential treatment of neurological disorders including AD (Thomson Reuters Pharma update of January 17, 2012). Conforma Therapeutics (San Diego, CA) presented a new class of HsP90 inhibitors [76–78] and showed that E102 (structure not disclosed) promoted selective decrease of the P-tau species in a mouse model of tauopathy. KU-32 (University of Kansas, Fig. 2) is a novobiocin derivative acting as an inhibitor of Hsp90 and an inducer of Hsp70 for the potential treatment of neurological diseases. In transgenic mice expressing human P301L-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 HsP90 inhibitors (Thomson Reuters Pharma update of March 31, 2011).The structures were not disclosed. Fig. 2. Heat shock protein 90 inhibitors and a thyrotropin-releasing hormone analogue. 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 heat shock-protein 70-interacting protein (CHIP), which functions as a tau ubiquitin ligase [79]. CHIP is a key molecular link between A and tau pathologies. Increasing CHIP levels may have beneficial effects to decrease tau pathology, because CHIP can polyubiquitinate tau and may play a crucial role in preventing accumulation of phospho-tau and neurofibrillary tangles. Heat shock protein 70 prevented both tau aggregation and the inhibitory effect of preexisting tau aggregates on fast axonal transport [80]. An exchange of HsP70 for HsP90 is involved in tau degradation [1701]. Excellent reviews on heat shock proteins were previously published [81, 82]. The development of AEG-33773 (Aegera Therapeutics), an allosteric modulator of HsP90 leading to HsP70 upregulation, was terminated. DRUGS INTERACTING WITH HORMONES Sustained efforts went into the exploration of the cognition enhancing effects of thyrotropin-releasing hormone and its analogues since it had been recognized as an activator of brain cholinergic systems [83]. Pilot studies of intravenous thyrotropin-releasing hormone in AD were undertaken [84, 85]. Improvement of cognitive deficits in depressed patients were reported [86]. Taltirelin (Ceredist, TA-0910; Mitsubishi Tanabe Pharma) (Fig. 2), an orally active synthetic thyrotropinreleasing hormone analogue, was launched in Japan in 2000 for the treatment of neurodegenerative disease, in particular spinocerebellar degeneration [87–89]. The sales in 2011 were USD 226.8 million (Thomson Reuters Pharma, update of August 30, 2012). KPS-0373 (Kissei under license from Shionogi) is a thyrotropin-releasing hormone derivative for the potential oral treatment of spinocerebellar ataxia in Phase II clinical trials in Japan (Thomson Reuters Pharma, update of July 31, 2012). The structure was not communicated. Leuprolide acetate implant (VP-4896, Memryte; Curaxis Pharmaceuticals, Raleigh NC, formerly Voyager Pharmaceutical and DURECT) is a biodegradable implant formulation of the gonadotropin-releasing hormone agonist 1-9-luteinizing hormone-releasing factor-6D-leucine-9-(N-ethyl-L-prolinamide), a nonapeptide, for the treatment of AD in women. A Phase IIb clinical study plan is currently evaluated. The effects of treatment with leuprolide acetate depot on working memory in women were described [90]. Luteinizing hormone modulated the processing of amyloid- protein precursor (APP) and A deposition [91] and of cognition in transgenic mice [92]. Several reviews were published [93–96] (Thomson Reuters Pharma, update of August 24, 2012). Tesamorelin (Egrifta; TH-9507; Theratechnologies, Quebec and US commercialization partner EMD Serono and licensee Sanofi) is a stabilized, truncated growth hormone releasing factor (GRF1-44; ThGRF1-44). In a controlled study of 152 adults (66 with MCI) participants self-administered daily subcutaneous injections of tesamorelin or placebo for 20 weeks, which had favorable effects on cognition in both adults with MCI and healthy older adults [97] (Thomson Reuters Pharma, update of August 15, 2012). The development of azetirelin (YM-14673; Yamanouchi, now Astellas [98, 99]), of calcitriol (Neurocalc; Apollo Biopharmaceuticals, now MitoKor), of JTP-2942 (Japan Tobacco [100–102]), of montirelin (CG-3703; CNK-602A; NS-3; Grünenthal [103]), of orotirelin (CG-3509; Grünenthal [104–106]), of posatirelin (RGH-2202; Gedeon Richter [107–109]), of protirelin (DN-1417; Takeda [110]), of thyrotropin-releasing hormone analogs (Roche), and of thymoliberin (RX-77368; Reckitt & Colman [111]) was terminated. DRUGS INTERACTING WITH ION CHANNELS (NON-RECEPTORS) Dysregulation of Ca2+ homeostasis plays a crucial role in the pathogenesis of AD. In the pathogenesis of AD [1702]. Oligomeric A proteins directly incorporated into neuronal membranes, formed cationsensitive ion channels (“amyloid channels”) and caused disruption of calcium homeostasis [112, 113]. A oligomers directly and dose-dependently modulated P/Q-type calcium channels [114]. In triple transgenic AD mice, upregulated ryanodine receptor activity led to a reduction of long-term potentiation [115]. Mutations of presenilin can affect the intracellular calcium levels via the endoplasmic reticulum calcium stores [116]. The levels of the calcium-sensing enzyme hippocalcin were elevated in AD brains [117]. Interesting reviews were published [118–121]. Drug repositioning for the treatment of Alzheimer’s disease was described recently [1703]. ARC-029 (Archer Pharmaceuticals, Roskamp Institute, Sarasota FL) is a blood-brain barrier crossing 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 of L-type voltage-gated calcium channels. The drug has been approved for a multisite Phase III clinical trial enrolling more than 500 AD patients in Europe. In February 2012, the study was expected to begin in the second half of 2012 (Thomson Reuters Pharma, update of April 16, 2012). Nilvadipine (FR-34235; Fujisawa; launched in Japan in 1989) was extensively characterized in vitro and in vivo for the potential treatment of AD. It facilitated the clearance of A across the blood-brain barrier [122, 123]. It prevented the impairment of spatial memory induced by cerebral ischemia combined with A in rats [124, 125]. It also antagonized A vasoactivity [126]. First positive clinical results on nilvadipine in AD patients were communicated. It was safe and provided short-term cognitive benefits in AD patients [127–131]. ARC-031 and ARC-031-SR (Archer Pharmaceuticals; sustained release formulation) is a non-calcium channel blocking and soluble A reducing nilvadipine derivative in Phase I clinical trials (Thomson Reuters Pharma, update of January 3, 2012). The structure was not communicated. RNS-60 (Revalesio Corporation, Tacoma WA) acts on voltage-gated ion channels. It is formulated as an inhalant anti-inflammatory charge-stabilized nanostructure. The company investigates also the potential treatment in AD and PD. A Phase I/IIa asthma trial began in May 2010. In October 2011, data from the multi-dose stage of the Phase I study were reported. RNS-60 showed a therapeutic benefit and was safe at all doses (Thomson Reuters Pharma, update of April 27, 2012). The structure was not communicated. ZSET-1446 (ST-101, Sonexa Therapeutics, San Diego 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 of essential tremor was initiated in the US in May 2011. It was recognized that ZSET-1446 (ST-101) targeted T-type voltage-gated calcium channels in mediating improved cognition in the CNS [132]. In vivo data were published previously [133, 134] (Thomson Reuters Pharma, update of July 17, 2012). AD-N02 (Adamed, Mazowieckie, Poland) is a sodium channel blocker and dopamine/5-HT stabilizer for the potential treatment of schizophrenia and bipolar disorder (Thomson Reuters Pharma, update of August 22, 2012). The structure was not communicated. Isradipine (Novartis, launched in 1997) is a L-type voltage-gated calcium channel blocker protecting MC65 neuroblastoma cells from APP 8 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes C-terminal fragment (APP-CTF)-induced neurotoxicity [135–137, 1704]. The development of many drugs interacting with ion channels was terminated (in alphabetical order): DMP-543 (Bristol-Myers Squibb; an inhibitor of Mtype K+ channels), enecadin (PAION under license from Nippon Shinyaku; a sodium and calcium channel blocker), 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-Myers Squibb; an inhibitor of M-type K+ channels; despite promising EEG brain mapping data [138] and an increase of parietal regional cerebral blood flow in AD patients [139], the clinical results were disappointing [140]), MEM-1003 (BAY-Z-4406, Memory Pharmaceuticals under license from Bayer; a L-type voltage-gated calcium channel [141]), nerispirdine (HP-184, Hoechst, now sanofi; an inhibitor of Mtype 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, formerly Neuropharma; a dual calcium channel blocker and acetylcholinesterase inhibitor [155]), NS-649 (NeuroSearch; a neuron-specific calcium channel blocker [156]), NSD-761 (NeuroSearch; a selective ion channel modulator); RGH-2716 (TDN-345; a nootropic neuroselective ion channel blocker; Takeda in collaboration with Gedeon Richter [157–159]), SNX-482 (R-type calcium channel blocker of Neurex, now Elan Pharmaceuticals [160–162]), SPI-017 (Sucampo; selective type-2 chloride channel activator), tamolarizine (NC-1100, calcium channel blocker, Nippon Chemiphar [163–165]), VRX-698 (Valeant Pharmaceuticals International; a potassium channel opener), and XE-991 (Bristol-Myers Squibb; a KCNQ potassium channel blocker [166, 167]). DRUGS INTERACTING WITH NERVE GROWTH FACTORS The role of neurotrophic factors in AD was discussed [168] as was a neurotrophic rationale for the therapy of neurodegenerative diseases [169]. The potential therapeutic use of BDNF, a key regulator for protein-synthesis dependent long-term potentiation and long-term memory, in neurological and psychiatric disorders was presented [170, 171], in particular for AD [172, 173]. Interestingly, genetic knockdown of BDNF in triple transgenic AD mice did not alter A or tau pathology [174]. Nerve growth factor (NGF)cholinergic dependency in brain aging, MCI, and AD was discussed [175–181]. NGF treatment in dementia was described [182]. NGF and AD, new facts for an old hypotheses were communicated recently [183]. Hippocampal ProNGF signaling pathways and amyloid- levels in mild cognitive impairment and Alzheimer’s disease were elucidated [1705]. It was claimed that NGF promoted long-term memory formation by activating poly(ADP-ribose) polymerase-1 (PARP-1) [184]. NeuroAid (MLC-601; Danqi Plantan Jiaonang; Moleac Pte Ltd, Singapore) is a BDNF stimulator derived from Radix Astragali, Radix Salvia Miltiorrhizae, Radix Paeoniae Rubra, Rhizoma Chuanxiong, Radix Angelicae Sinensis, Carthamus Tinctorius, Prunus Persica, Radix Polygalae, Rhizoma Acori Tatarinowii, Buthus Martensii, Hirudo, Eupolyphaga Seu Steleophaga, Calculus Bovis Synthetic or Artifactus and Cornu Saigae Tataricae, for the potential oral prevention of stroke including cerebral infarction and ischemic stroke and for the potential treatment of traumatic brain injury in a randomized, doubleblind, placebo-controlled, multicenter, Phase III trial (n = 1100) since November 2007 in Singapore and the Philippines. The study was expected to be completed in 2012 (Thomson Reuters Pharma, update of October 3, 2012). CERE-110 (AAV2-NGF; NeuroRescue AD; Ceregene, San Diego CA) is an AAV2 vector based gene delivery system containing cDNA for NGF. A Phase II clinical trial in mild-to-moderate AD patients (n = 50) started in May 2009 in the US. The therapeutic potential of CERE-110 was described in detail [185, 186] (Thomson Reuters Pharma, update of July 20, 2012). GM-607 (Genervon Biopharmaceuticals, Pasadena CA) is a motoneuronotrophic factor analogue for the potential treatment of ischemic stroke, spinal cord injuries, ALS, AD, HD, and PD. By April 2011, a Phase II ischemic stroke trial was initiated. By February 2012, further Phase II IND applications were being prepared for spinal cord injuries, ALS, PD, and AD (Thomson Reuters Pharma, update of August 28, 2012). MIM-D3 (Mimetogen Pharmaceutics, Quebec, Fig. 3) is a small molecule NGF peptidomimetic for the treatment of AD and dry eye (xerophthalmia). A Phase II trial for dry eye was initiated in November 2010 [187] (Thomson Reuters Pharma, update of May 17, 2012). W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes PYM-50028 (Cogane, P58, P63; Phytopharm, UK) (Fig. 3), a steroidal saponin, stimulated BDNF release and may be beneficial for the treatment of PD, AD, glaucoma, and ALS. PYM-50028 reversed neuronal damage induced by 1-methyl-4phenyl-1,2,3,6-tetrahydropyridine (MTPT) in a mouse model of PD [188]. In July 2011, FDA granted PYM-50028 orphan drug status for ALS. In November 2010, recruitment of patients (n = 400) began in the randomized, double-blind, proof-of-concept, placebo-controlled, dose-ranging Phase II study CONFIDENT-PD (Thomson Reuters Pharma, update of August 16, 2012). T-817MA (Toyama Chemical, Fujifilm Holding) (Fig. 3) is a neurotrophic small molecule in Phase II clinical trials for AD in the US since April 2008. T-817MA attenuated cognitive impairments in P301L tau transgenic mice [189], prevented memory deficits caused by i.c.v. A administration [190, 191], and attenuated A-induced neurotoxicity [192] (Thomson Reuters Pharma, update of July 30, 2012). NsG-0202 (ECB-AD, ECT-AD, NsGene A/s, Denmark) is an encapsulated cell biodelivery system to deliver cells expressing NGF. A Phase Ib trial started in December 2007 in Sweden. First clinical data were presented in July 2011. In January 2012, the trial was expected to be completed in 2012 [193] (Thomson Reuters Pharma, update of January 17, 2012). There are several drugs interacting with neurotrophic factors in preclinical evaluation (in alphabetical order): AL-209 (ADNF-9, SAL; Allon Therapeutics, Vancouver) is an activity-dependent neurotrophic factor derived nine amino acid sequence peptide and tubulinbinding agent that stimulated PARP-1 for the potential treatment of CNS and ocular disorders (Thomson Reuters Pharma, update of March 6, 2012). AL-309 (Allon Therapeutics, Vancouver) is an analogue of AL-209 and PARP stimulator for oral, intranasal and s.c. administration. By October 2012 preclinical development had been completed (Thomson Reuters Pharma, update of October 17, 2012). AMRS-001 (CNS-001; Amarantus BioSciences, Sunnyvale CA, formerly CNS Protein Therapeutics having acquired the relevant IP from Prescient Neuropharma) is a program of therapeutics based on mesencephalic astrocyte-derived neurotrophic factor [194], which promoted the survival of dopaminergic neurons for the potential treatment of PD, AD, and brain injury. It also protected the heart from ischemic damage [195] (Thomson Reuters Pharma, update of October 2, 2012). 9 Catecholamine derivatives (Emory University, Atlanta, GA) acted like BDNF to activate the tyrosine kinase B (TrkB) receptor. They have a potential for the treatment 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 receptor leading to its phosphorylation (Thomson Reuters Pharma, update of December 27, 2011). The structures were not communicated. 7,8-Dihydroxy-flavone, a TrkB receptor agonist and BDNF mimic, reversed memory deficits in a mouse model of AD [196]. The compound and derivatives thereof are also evaluated at Emory University in collaboration 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 receptor that inhibited p75NTR-mediated neuronal death via regulation of the Trk receptor function for the potential treatment of AD, PD, and motor neuron disease (Thomson Reuters Pharma, update of May 17, 2012). Gambogic amine and gambogic amide (Emory University, Atlanta GA), TrkA receptor agonists with robust neurotrophic activity, prevented neuronal cell death [200] (Thomson Reuters Pharma, update of August 28, 2012). Gedunin (Emory University, Atlanta GA) and its derivatives including deoxygedunin are TrkB receptor agonists for the potential treatment of PD, HD, AD, HIV-related dementia, ALS, stroke, and multiple sclerosis [201] (Thomson Reuters Pharma, update of April 26, 2012). JRP-655 (Prous Institute for Biomedical Research, Barcelona) is a BDNF secretagogue and neuroplasticity modulator (Thomson Reuters Pharma, update of September 5, 2012). The structure was not communicated. 4-methylcatechol, which increased BDNF content and stimulated BDNF mRNA expression and synthesis, effectively improved spatial learning and memory in rats [202, 203]. NeuroAiD II (MLC-901, Moleac Pte Ltd., Singapore), a BDNF stimulator, is a derivative of NeuroAiD for the potential treatment of global ischemia in preclinical development (Thomson Reuters Pharma, update of September 2, 2011). ND-602 (Neurodyn, Charlottetown, Canada) is a neuroprotective progranulin (PC cell-growth factor)- 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 treatment of PD, AD, and ALS. ND-602 was found to increase activity of neprilysin and significantly reduced A and plaque burden in the hippocampus and entorhinal cortex of Tg2576 mice (Thomson Reuters, update of May 25, 2012). The development of many drugs interacting with neurotrophic factors was terminated. The most advanced drug, xaliproden (SR-57746A, Xaprila; sanofi), a 5-HT1A agonist and stimulator of endogenous neurotrophin synthesis, did not meet the cognition endpoints in Phase III trials in AD patients, which applies also to paliroden (SR-57667B; with a biphenyl instead of the -naphtyl in xaliproden; sanofi, [204]). The lessons learnt from the xaliproden clinical trials were discussed [205]. The biological characterization was reviewed [206]. Also the development of other drugs interacting with neurotrophic factors (in alphabetical order) was terminated: ABS-200 (American Biogenetic Sciences), ADNF-14 (NIH, activity-dependent neurotrophic factor-14), AGT-120, and AGT-140 (ArmaGen Technologies); AK-30-NGF (a monoclonal antibody as carrier for the delivery of NGF; Alkermes), arundic acid (MK-0724, ONO-2506, Arocyte, Ono Pharmaceuticals [207–210]), beta NGF (tethered to a molecular shuttle; Apollo Life Sciences), CEP-427 (a neurotrophic factor enhancing small molecule; Cephalon, now Teva), coleneuramide (MCC-257; Mitsubishi Tanabe Pharma), CX-438 (Cortex Pharmaceuticals), dekafin-1 (a fibroblast growth factor receptor, agonist and tyrosine kinase modulator; Enkam Pharmaceuticals); dFGF (dimerized fibroblast growth factor; Massachusetts Institute of Technology (MIT)/ViaCell Inc.), GDF-1 (Creative Biomolecules, now Curis; a growth and differentiation factor-1 agonist), glial maturation factor (Rhone-Poulenc/Regeneron), huIM-13 (a humanized monoclonal antibody that binds to the TrkA receptor, Lay Line Genomics), inosine (an NGF, Alseres Pharmaceuticals, 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 Pharmaceuticals, formerly NeoTherapeutics), which induced the expression of NGF, neurotrophin-3, and of basic fibroblast growth factor [211–216], LM11A-31 (a p75 neurotrophin receptor ligand; Elan), nerve growth factor agonists (Chiron), neuregulin-2 (cerebellumderived growth factor, NRG2; Acorda Therapeutics under license from CeNeS and Harvard University), NGF therapy (Lay Line Genomics), NP-901 (Noscira), NS-521 (NeuroSearch), p75 NGF receptor antagonist (Circadian Technologies under license from the Walter & Eliza Hall Institute), NT-3 (Genentech and StemCells), ReN-1820 (ReNeuron under license from the University of Bristol), ST-857 (acetylL-carnitine arginine amide; Sigma-Tau [217–219]), T-588 (Toyama [220–227]), and trofexin (a peptide neurotrophic factor; Protarga). DRUGS INTERACTING WITH RE-UPTAKE TRANSPORTERS (PSYCHOSTIMULANTS) Psychostimulants exert behavioral-calming and cognition-enhancing actions in the treatment of ADHD [228]. Cognition-enhancing doses of psychostimulants exert regionally restricted actions elevating extracellular catecholamine levels and enhancing neural signal processing preferentially in the prefrontal cortex. Additional evidence suggests a prominent role of prefrontal cortex ␣2 and D1 receptors in the behavioral and electrophysiological actions of low-dose psychostimulants [229, 230]. Excellent reviews on attention-modulating effects of cognitive enhancers were published [231–236]. An analysis of student use (and abuse) of cognitive enhancers was presented [237]. A review on glycine transporter type 1 (GlyT1) inhibitors was disclosed [238]. Methylphenidate (Ritalin, Ciba, now Novartis, launched 1956) is a psychostimulant drug, which increased the levels of dopamine and noradrenaline in the brain through re-uptake inhibition of the monoamine transporters [239]. Methylphenidate preferentially increased catecholamine neurotransmission within the prefrontal cortex at low doses that enhance cognitive functions [240]. Cognitionenhancing doses of methylphenidate preferentially increased prefrontal cortex neuronal responsiveness [241]. Methylphenidate decreased the amount of glucose needed by the brain to perform a cognitive task [242]. The improvement of memory functions by methylphenidate (and modafinil) in healthy individuals was described [243]. Most of these experiments were made with the mixture of erythro and threo stereoisomers. The active principle of methylphenidate is the (+)-(␣R,2R)-threo-methylphenidate hydrochloride (Dexmethylphenidate, Ritadex; Novartis Pharma under license from Celgene launched 2002) (Fig. 4), also available as an extended-release oral formulation Focalin XR [244]. The spheroidal oral drug absorption system dexmethylphenidate was described [245] (Thomson Reuters Pharma, update of July 27, 2012). 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, later Cephalon, now Teva, launched 1998) is an inhibitor of dopamine, noradrenaline, and serotonin re-uptake and an ␣1 adrenoceptor agonist [246]. Modafinil showed improvement of cognition and attention in patients with chronic schizophrenia ADHD [247, 248], in particular in the first episode of psychosis [249, 250]. The neuronal mechanism by which modafinil affects cognitive and emotional function in schizophrenic patients was reviewed [251]. Experiments using BOLD functional magnetic resonance imaging (fMRI) in healthy volunteers showed that modafinil enhanced the efficiency of prefrontal cortical cognitive information processing, while dampening reactivity to threatening stimuli in the amygdala, the brain area implicated in anxiety [252]. Modafinil improved attention in wellrested individuals [243]. Modafinil reliably enhanced performance on several cognitive tests of planning 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 learning and delayed recall performance in healthy volunteers [253]. It may be useful in the treatment of substance abuse [254]. It improved working memory and sustained 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). The sales of armodafinil in 2011 amounted to USD 266 mil (Thomson Reuters Pharma, update of September 21, 2012). Atomoxetine (tomoxetine, Strattera, LY-139603; Lilly; launched 2003) (Fig. 4) is a selective noradrenaline re-uptake inhibitor for the treatment of ADHD in children and adults [256]. Atomoxetine inhibited noradrenaline and serotonin transporters with Ki values of 2.6 and 48 nM, respectively [257]. Atomoxetine improved response inhibition in ADHD patients and in healthy volunteers via increased activation 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 Pharmaceuticals and New River Pharmaceuticals, launched 2007) (Fig. 4) is a drug for the treatment of ADHD in children, adolescents, and adults [260]. Lisdexamfetamine itself is inactive and acts as a prodrug to dextroamphetamine upon cleavage of the lysine portion of the molecule. Dextroamphetamine inhibited noradrenaline, serotonin and dopamine transporters with respective Ki values of 39, 3,830, and 34 nM [261]. The use of lisdexamfetamine in patients with dementia was described [231]. Sales in 2011 were USD 805 million (Thomson Reuters Pharma, update of September 28, 2012). Indeloxazine (YM-08054; Elen, Noin, Yamanouchi, Schering-Plough, now Merck and Jeil Pharmaceuticals, launched in China and South Korea) (Fig. 4) blocked the reuptake of noradrenaline with IC50 of 3.2 M and of serotonin with IC50 of 0.71 M. It is also a MAO inhibitor. It showed learning and memory improving effects in many animal tests [262–267]. NS-2359 (NeuroSearch, Fig. 4) is a blocker of dopamine, noradrenaline, and serotonin re-uptake currently in Phase I clinical trials in Denmark for the potential treatment of CNS diseases. GSK was previously developing NS-2359 for the treatment for depression but discontinued the development (Thomson Reuters Pharma, update of September 26, 2012). There are several drugs interacting with re-uptake transporters in preclinical evaluation (in alphabetical order): AM-285 (Cyclocreatine, CincY; Lumos Pharma, Austin TX based on University of Cincinnati technology) (Fig. 4) is evaluated for the potential treatment of creatine transporter deficiency, an X-linked autism spectrum disorder characterized by severe cognitive impairment [268]. In June 2012, the FDA awarded cyclocreatine Orphan designation for the treatment of creatine transporter deficiency (Thomson Reuters Pharma, update of August 24, 2012). AS-1522489-00 (Astellas Pharma) is a glycine transporter-1 inhibitor, a 1,2,4-triazole derivative, of which only the (S)-atropisomer is biologically active [269, 270] (Thomson Reuters Pharma, update of January 27, 2012). The structure was not communicated. Lu-AA42202 (Lundbeck) (Fig. 4) is a triple dopamine, noradrenaline, and serotonin re-uptake inhibitor for the potential treatment of major depressive disorders and ADHD (Thomson Reuters Pharma, update of December 1, 2010). MLR-1017 (mesocarb, sydnocarb, sidnocarb, Melior Pharmaceuticals, Exton, PA) (Fig. 4) is a dopamine transporter inhibitor for the potential treatment of ADHD and levodopa-induced side effects in PD. The drug was previously launched in Russia [271, 272] (Thomson Reuters Pharma, update of April 12, 2012). PD-2005 (P2D Bioscience, Cincinnati, OH, identical with AHN 2-005, National Institute of Drug Abuse and US Naval Medical Research Center) (Fig. 4) is an inhibitor of the dopamine transporter for the potential treatment of ADHD and for cognitive impairments in traumatic brain injury. It dose-dependently improved performance of rats in a delayed-alternation task of spatial working memory [273] (Thomson Reuters Pharma, update of June 12, 2012). RO-4543338 (Roche) is a well-tolerated glycine transporter inhibitor, which facilitated drug cue extinction [274, 275]. The structure was not communicated. Selective Serotonin Re-uptake Inhibitors (SSRIs) showed cognitive enhancing effects [276]. Patients treated with citalopram showed significant improvements on a cognitive subscale. Paroxetine and fluoxetine improved most of the tested cognitive functions in a study of 242 depressed patients during one year [277]. Other studies with citalopram, fluoxetine, sertraline, and zimelidine did not report improvement of memory and/or reaction time in demented patients [278]. Thiethylperazine (Torecan) (Fig. 4), a drug approved by the FDA to relieve nausea and vomiting, 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 mouse model of AD expressing the ABCC1 transporter, a markedly reduced load of A was measured. Deficiency of ABCC1 transporter substantially increased cerebral A levels [279] (Thomson Reuters Pharma, update of April 26, 2012). The development of ASP-2535 (Astellas; a glycine transporter-1 inhibitor [280]), coluracetam (BCI-540, MKC-231; BrainCells Inc. under license from Mitsubishi Tanabe Pharma; an enhancer of high-affinity choline uptake and K+ -induced release of acetylcholine [281–288]), CP-101 (CogniPharm, an i.v. formulation of modafinil), D-serine re-uptake inhibitors (Memory Pharmaceuticals, now Roche), flufenoxina (FAES Farma; a dual serotonin and norepinephrine re-uptake inhibitor), glycine transporter-1 inhibitors (Helicon Therapeutics), JNJ-17305600 (NFPS; NPS Allelix; a glycine transporter inhibitor [289]), PD-2007 (P2D Bioscience, a dopamine transporter inhibitor), SCH-900435 (Org-25935; Organon, Schering-Plough, now Merck, a glycine transporter-1 inhibitor), 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, a noradrenaline uptake inhibitor [298],[299–301]), and tesofensine (NS-2330, NeuroSearch, a monoamine re-uptake inhibitors of serotonin, dopamine, and noradrenaline [302]) was terminated. The development of tesofensine for the once-daily oral treatment of obesity in Phase II trials was also suspended [303]. The vesicular monoamine transporter 2 (VMAT2) located on the membrane of vesicles is responsible for storing and packaging neurotransmitters into monoamine vesicles or granules. Imaging VMAT2 in the brain provides a measurement reflecting the integrity of dopaminergic, noradrenergic and serotonergic neurons [304]. Positron emission tomography (PET) ligands for the VMAT2 have become valuable diagnostic tools. (+)-[11 C]-dihydrotetrabenazine (DTBZ) (Fig. 5) allows the determination of the striatal monoaminergic presynaptic terminal density. In a study enrolling 20 patients with dementia with Lewy bodies (DLB), 25 AD patients, 7 patients with frontotemporal dementia, and 19 elderly controls clinicians could accurately differentiate between the different forms of dementia [305–307]. The uptake of (+)-11 C-DTBZ in the stria- 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 presynaptic dopaminergic system was unchanged compared to controls. The striatal uptake in DLB and PD patients was significantly decreased [308]. 18 F-Florbenazine (18 F-AV-133; 18 F-FP-DTBZ; Avid Radiopharmaceuticals, Philadelphia PA, a subsidiary of Lilly, under license from University of Michigan) (Fig. 5) is in Phase III clinical evaluation. In May 2012, an open-label, single-blind, Phase II/III study (NCT01550484; 18 F-AV-133-B04) was initiated in patients with undiagnosed movement disorders (n = 150) in the US to assess the safety and efficacy of florbenazine in differentiating PD. At that time, the study was scheduled to complete in September 2014. For the synthesis, see [309]; for the binding characteristics in rat striatum and hypothalamus homogenates (Kd = 0.19 and 0.25 nM, respectively), see [310, 311]. The whole body biodistribution was studied [312]. 18 F-AV-133 was used for noninvasive assessment of the vesicular monoamine transporter type 2 in 17 PD patients and 6 healthy controls. VMAT2 binding potential was decreased by 81% in the posterior putamen, 70% in the anterior putamen, and 48% in the caudate nucleus of PD patients [313]. This ligand is a valid measure of dopaminergic neuron integrity. The in vivo assessment in patients with DLB and AD was reported [314]. The optimal scanning time window was elucidated [315]. For PET imaging in a MPTP-induced mouse model of PD, see [316] (Thomson Reuters Pharma, update of August 7, 2012). 11 C--CFT and 18 F--CFT (Fig. 5) proved to be valuable PET ligands due to their high affinity interaction with dopamine re-uptake sites. AD patients showed a reduction of 11 C--CFT in putamen and caudate of about 20%. Thus the putamen and the caudate nucleus were equally affected in contrast to PD, which showed predominantly putaminal reduction [317]. In PD, the mean uptake of 18 F--CFT in the contralateral putamen was reduced to 31% and in the ipsilatral putamen to 45%. In the caudate nucleus, the uptake was reduced contralaterally to 67% and ipsilaterally to 77% [318]. For an exhaustive review, see [319]. 123 I-Ioflupane (123 I-FP-CIT; Amersham, now GE Healthcare) (Fig. 5) is a SPECT ligand launched in 2000. It is the ligand of choice to differentiate between AD and DLB patients [320, 321]. It is valuable for the diagnosis of Parkinsonian syndromes [322]. SERT dependent 123 I-ioflupane uptake allows a more comprehensive assessment of neurochemical disturbances in degenerative Parkinsonism [323–325] (Thomson Reuters Pharma, update of August 14, 2012). DRUGS INTERACTING WITH TRANSCRIPTION FACTORS cAMP response element-binding (CREB) protein and the discovery of cognitive enhancers was reviewed [326–328]. CREB phosphorylation as a mechanistic marker in the development of memory enhancing AD therapeutics was described [329]. The current knowledge of key calcium signal-regulated transcription factors, namely CREB, nuclear factor of activated T-cells, and downstream regulatory element antagonist modulator (DREAM) and memory formation was summarized [330]. A disrupted activity-dependent gene transcription required for memory through the CREB-regulated transcription co-activators CRTC1 [331]. The GABAB receptor antagonist SGS742 improved spatial memory and reduced protein binding to CREB2 in the hippocampus [332] (see Part 1, Chapter 1.10.2 GABAB receptors). CREB antisense oligonucleotide administration into the dorsal hippocampal CA3 region impaired long- but not shortterm spatial memory in mice [333]. The lack of DREAM protein enhanced learning and memory and slowed brain aging [334]. SRF/MYOCD inhibitors (Socratech LLC, Rochester NY) are small molecule inhibitors against serum response factor and myocardin, which are interactive transcription factors in vascular smooth muscle cells for the potential treatment of AD [335]. For a review on the imbalance of vascular molecules in AD, see [336] (Thomson Reuters Pharma, update of February 15, 2012). Structures were not communicated. The development of CREB modulators (Helicon Therapeutics) and of EPAC inhibitors (exchange protein directly activated by cAMP, a guanine nucleotideexchange factor; Scottish Biomedical [337]) was terminated. ANTIOXIDANTS Soluble A oligomers localize to mitochondria and interfere with their normal functioning causing an overproduction of reactive oxygen species (ROS), inhibiting respiration and ATP production and damaging the structure of mitochondria [338–340]. Some authors, however, claim that the early impairments of mitochondrial dysfunction and oxidative stress may precede A overproduction and deposition [341–343]. Excellent reviews were presented over years [344–347]. Possibilities of intervention of antioxidant pathways in AD were discussed [348, 349]. 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 is tightly connected with the oxidative cascade. Over the years, clinicians have explored treatment of AD patients with antioxidants. Free radicals can be scavenged by dietary means. Compounds such as acetyl-L-carnitine, curcumin [350], Gingko biloba extracts such as EGb 716, (R)-α-lipoic acid, melatonin, 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 consequence of a landmark study, clinicians even advise AD patients not to take large doses of vitamin E or ␣-lipoic acid [373]. Many clinical trials were also carried out with synthetic 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’s lazaroids, such as tirilazad or U-74006F, U-74389G, U-74500A, U-75412E, U-78518F, and U-83836E [381–384]). None of these compounds produced statistically significant therapeutic benefits for AD patients. MitoQ (redox mixture of mitoquinone and mitoquinol; Antipodean Pharmaceuticals, Menlo Park CA, under license from the University of Otago; Fig. 6 shows the oxidized form) is a brain penetrating mitochondria-targeting antioxidant [385–388]. It selectively blocked mitochondrial oxidative damage and prevented cell death [389–394]. It is evaluated in Phase II clinical trials for the treatment of PD and liver damage. Clinical trials for the treatment of Friedreich’s ataxia and sunburn were abandoned (Thomson Reuters Pharma, 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 to triphenyl-phosphonium bromide was terminated [399] (Thomson Reuters Pharma, update of June 18, 2012). MTP-131 (Bendavia, SS-31, Szeto-Schiller peptide; Stealth Peptides Inc., Newton Centre MA) (Fig. 7) is a cell-permeable mitochondria-targeting antioxidant tetrapeptide (NH2 -D-Arg-Dmt-Lys-Phe-NH2 ), which is in Phase II clinical trials for ischemia reperfusion injury since September. 2010 (EMBRACE–STEMI trial). It will further be investigated for the treatment of eye, neurological, mitochondrial, and metabolic 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, Indole3-propionic acid; ViroPharma, Exton PA, under license from Intellect Neurosciences, New York University and Mindset BioPharmaceuticals) is an antioxidant and A aggregation/deposition inhibitor. The prevention of A induced neurotoxicity by indole-3-propionic acid was described [404]. The rationale behind the selection of OXIGON as a potential disease-modifying therapy for Alzheimer’s disease was presented [1706]. Preliminary data from a multiple-dose Phase Ib trial for AD were reported in October 2010. It appears that ViroPharma wants to pursue the drug for the indication Friedreich’s ataxia only (Thomson Reuters Pharma, update of August 17, 2012). There are several antioxidants in preclinical evaluation (in alphabetical order): Catalytic manganoporphyrine antioxidants, such as AEOL-10113, AEOL-10150, AEOL-10201 and AEOL-11207 (Aeolus Pharmaceuticals, Mission Viejo 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 treatment of traumatic brain injury and stroke (Thomson Reuters Pharma, update of November 9, 2011). DL-3-n-butylphtalide (NBP, Hebei, China) is a natural antioxidant extracted from seeds of Apium and a powerful free radical scavenger [413]. It protected dopamine neurons in a rotenone model for PD [414]. FRP-0924 (gemifloxacin, Neuron BioPharma, Granada, Spain) is an antioxidant and neuroprotectant for the potential treatment of neurodegenerative diseases including AD. FRP-924 crossed the blood-brain barrier and prevented neuronal death. As fluoroquinolones are generally well tolerated with most side effects being mild and serious adverse effects being rare, it is expected that the drug will enter Phase II rapidly (Thomson Reuters Pharma, update of August 14, 2012). IAC (Cerebricon, Finland and Medestea Research, Torino, Italy) (Fig. 8) is a novel radical scavenger react- 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-centered radicals of biological interest [415–418]. Daily treatment with IAC (3–30 mg/kg i.p.) decreased mortality, enhanced cognitive functions in the water maze, and reduced the A plaque burden in hAPP transgenic mice [419]. Lipid soluble antioxidants (OXIS International, Beverley Hills CA) mimic the activity of the body’s natural cell membrane-protecting antioxidant vitamin E for the potential treatment of cardiovascular diseases, diabetes, AD, and PD (Thomson Reuters Pharma, update of June 12, 2012). The structures were not communicated. Lipocrine and Memoquin, dual acetylcholinesterase inhibitors and antioxidants (University of Bologna), were described in Part 2, Chapter 2.1.1.3. (formulae in Part 2 in Fig. 6). NPS-0155 (Neuron BioPharma, Granada, Spain) is an antioxidant and neuroprotective compound for the potential treatment of AD and other neurodegenerative diseases (Thomson Reuters Pharma, update of April 27, 2012). The structure was not communicated. PAN-811 (3-AP; NSC-663249; OCX-191; Triapine, Panacea Pharmaceuticals, Gaithersburg MD) (Fig. 8) is a ribonucleotide reductase inhibitor, calcium ion chelator, and radical scavenger for the potential treatment of AD and ischemia [420, 421] (Thomson Reuters Pharma, 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 scavenger of free radicals. It attenuated the toxicity of A to energy metabolism, mitochondrial membrane structure, and key enzymes in the electron transport chain [422]. There are numerous preclinical reports of drugs and natural products, which, in addition to their radical scavenging properties, also prevented A-induced neurotoxicity, such as peoniflorin [423], 2,2 -pyridoin [424], quetiapine [425]), stemazole (Fig. 8) [426], and zeatin (Fig. 8) [427]. Dual free radical scavengers and A binding ligands were described [428, 429]. The development of many antioxidants was terminated (in alphabetical order) of AN-808 (Athena Neurosciences, now Elan Pharmaceuticals), AO-2 and AO-3 antioxidants (Antoxis), CNSB-002 (AM-36; Relevare; AMRAD, formerly Zenyth Therapeutics [430] [431, 432]), CPI-1189 (Centaur [433, 434]), disufenton sodium (Cerovive, NXY-059, AstraZeneca, which was in Phase III clinical trials for the treatment of acute ischemic stroke [435–445]), FR-210575 (Fujisawa [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 great detail over the last fifteen years. Excellent reviews were 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 an i.v. prodrug of the calcium and zinc chelator BAPTA for the potential treatment of stroke and traumatic brain injury. It is in Phase III clinical trials for stroke since October 2009. First reports on clinical results were communicated [486–488]. Also preclincial data were 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 attenuating compound that reduced levels of soluble A and tau hyperphosphorylation [492]. A Phase IIa study in AD patients (n = 78), started in December 2006 with daily doses of 50 or 250 mg or placebo over three months, was completed in January 2008. A significant reduction of A42 levels in the cerebrospinal fluid (CSF) was seen in the 250 mg group. This dose also significantly improved executive function performance in the category fluency and the trail making cognitive tests compared to placebo [493, 494]. In January 2012, the FDA approved an IND to initiate a randomized, multicenter, double-blind, placebo-controlled, parallel-group, safety, tolerability, and efficacy Phase IIa trial in early- to mid-stage HD patients (n = 100) in the US and Australia. In April 2012, the first patient was dosed in this trial. By June 2012 the trial had been initiated in Australia. Data are expected in the second half of 2013. The compound was extensively characterized in preclinical studies [495–497] (Thomson Reuters Pharma, update of October 2, 2012). See also Chapter 17.1. Small molecules preventing tau aggregation. AEN-100 (Synthetic Biologics, formerly Adeona Pharmaceuticals, Ann Arbor, MI) is a once-daily, gastroretentive, sustained-release, oral tablet formulation of zinc acetate for the potential treatment of ALS and AD. By November 2011, a Phase I study for ALS patients was underway (Thomson Reuters Pharma, update of September 19, 2012). Other metal chelators are currently in preclinical evaluation (in alphabetical order): Aom-0937 (Hangzhou Adamerck Pharmlabs, China) is a prodrug of DP-b99 (Thomson Reuters Pharma, update of July 28, 2011). The structure was not communicated. DP-460 (D-Pharm, Rehovot Israel) (Fig. 9) is a membrane active chelator derivative of the calciumspecific chelator BAPTA that modulates copper and zinc homeostasis to inhibit oxygen radicals and plaque formation. It is in preclinical evaluation for the potential oral treatment of AD and ALS (Thomson Reuters Pharma, update of November 1, 2011). It appears that DP-460 was preferred to DP-109 [498]. Both compounds have been evaluated as neuroprotectants in a transgenic mouse model of ALS [499] (Thomson Reuters Pharma, update of August 7, 2012). Deferoxamine (SAN-121; Sanomune, a subsidiary of DiaMedica under license from HealthPartners Research Foundation, Winnipeg, Canada) (Fig. 10) is a nasally-delivered iron chelator improving performance in a radial arm water maze in P3301L tau transgenic mice [500] (Thomson Reuters Pharma, update of December 23, 2011). HLA20A (Technion Haifa) (Fig. 10) is a combination of an 8-hydroxy-quinoline metal chelator, which was carbamoylated at the 8-hydroxy function to interact with acetylcholinesterase as in rivastigmine [501, 502]. See Part 2, Chapter 2.1.1.10. Dual acetylcholinesterase inhibitors and metal chelators. PA-1637 (Palumed, France) is an A plaque production-inhibiting copper chelating agent (Thomson Reuters Pharma, update of May 28, 2012). The structure was not communicated. PBT-3 and PBT-4 (Prana Biotechnology, Parkville, Australia) are non-8-hydroxyquinoline metal protein attenuating compounds, potential follow-ups of PBT-2. (For both Thomson Reuters Pharma, update of November 2, 2011). The structures were not communicated. Triazole-pyridine derivatives as inhibitors of metal-induced A aggregation were described [503]. VAR-10200 (HLA-20; Varinel, West Chester, PA) (Fig. 10) is a dual iron-chelating agent and MAO-B inhibitor for the potential treatment of age-related macular 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-hydroxyquinoline with the MAO inhibitor moiety of rasaglinine [504–517] (Thomson Reuters Pharma, update of September 24, 2012). See also Part 2, Chapter 2.26. Drugs interacting with Monoamine Oxidase. The development of many metal chelators was terminated (in alphabetical order): desferrioxamine [518–520], Gero-46 (clioquinol, Gerolymatos); NG-1 and PBT-1 (clioquinol, Prana Biotechnology [521, 522]); phanquinone (PN Gerolymatos); tetrathiomolybdate (a copper chelating agent of 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 license from the University of Michigan); the iron chelator VAR-10100 (VK-28) [523–526] and VK-11 and VK12 (Prana Biotechnology). NATURAL PRODUCTS Excellent reviews on naturally occurring phytochemicals for the prevention and treatment of AD have 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 “natural substances and AD: from preclinical studies to evidence based medicine” was published recently [533]. Mentat (BR-16A; Himalaya Drug Co.) is a standardized mixture of herbal extracts, which has been launched in India and the US (as MindCare) in November 2000 for the treatment of cognitive deficits (Thomson Reuters Pharma, update of November 6, 2000). YY-280 (Yucrid; Yuyu, South Korea) is a combination therapy of ticlopidine and EGb-761 (tanamin, a Ginkgo biloba extract) administered as a tablet for the treatment of apoplexy and myocardial infarction. 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, South Korea) is derived from the dried root of Pulsatella koreana (baekduong). A randomized, double-blind, placebo-controlled Phase III clinical trial for the treatment of mild-to-moderate AD patients (n = 256) was initiated in November 2010 in South Korea. SK-PCB70M improved scopolamine-induced impairments of memory consolidation in rats [535, 536]. It had antioxidant activity and reduced A levels in the brains of Tg2576 mice [537, 538]. It alleviated the neurologic symptoms in G93A-SOD1 ALS mice [538] (Thomson Reuters Pharma, update of February 28, 2012). Circadin (KI-1001, NSC-56423; Neurim Pharmaceuticals, Tel Aviv) is an oral controlled-release formulation of melatonin (i.e., melatonin acetamide) and was launched in 2007 for the treatment of primary insomnia. A Phase II clinical trial was initiated in Israel in patients with MCI (n = 50) in October 2007. Another Phase II clinical trial was initiated in the US, UK, and Israel in patients with mild-to-moderate AD (n = 140) treated with an acetylcholinesterase inhibitor to evaluate the effects of add-on Circadin (2 mg) on decline in cognitive skills, global functioning, and daytime somnolence 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 is in Phase II clinical trials for AD since March 2009 (Thomson Reuters Pharma, update of February 11, 2011). PPLs (NatureWise Biotech, Taipeh, Taiwan) are prenylflavanone compounds extracted from Taiwanese propolis in Phase II clinical trials. A pilot AD program of PPLs in combination with Aricept was initiated in November 2010 (Thomson Reuters Pharma, update of July 10, 2012). PTX-200 (Phytrix, Munich, Germany) is a plantderived neuroprotectant for the potential treatment of PD. A Phase I/IIa clinical evaluation in the UK was successfully completed (Thomson Reuters Pharma, update of February 1, 2012). Resveratrol is a natural phyto compound, which activated Sirtuin-1 [539–544]. It reduced A accumulation [545–548]. It remodeled soluble oligomers and fibrils of A into off-pathway conformers [549]. Resveratrol is not a direct activator of SIRT1 enzyme activity [550]. Resveratrol improved memory deficits in mice fed a high-fat diet [551]. Subchronic oral toxicity and cardiovascular safety pharmacology studies were carried out [552]. The biosynthesis of resveratrol in yeast and in mammalian cells was achieved [553]. The Georgetown University Medical Center in Washington DC started at Phase II, randomized, double-blind, placebo-controlled study in patients with mild-to-moderate AD (n = 120) in the US in May 2012 (NCT01504854). A novel application in HD was discussed recently [554]. See also in Part 2, Chapter 2.40, Drugs interacting with Sirtuin, formula in Part 2, Fig. 23 (Thomson Reuters Pharma, update of June 22, 2012). RPh-201 (Regenera Pharma, Rehovot, Israel) is a subcutaneous formulation of an agent derived from a traditional medicinal plant. A Phase I/IIa randomized, double-blind, placebo-controlled trial in healthy volunteers and adults with AD (n = 56) started in January 2012 in Canada. A topical formulation is currently evaluated in a Phase II study in patients with chronic ulcerated wounds (n = 15) in Israel (both Thomson Reuters Pharma, update of January 31, 2012). VR-040 (apomorphine as dry powder inhaled formulation, Vectura, Chippenham, UK) is being developed in Phase II clinical trials for the potential treatment of PD since June 2006 (Thomson Reuters Pharma, update of October 3, 2012). Exebryl-1 (ProteoTech, Kirkland, WA and Chinese licensee Tasly Pharmaceuticals) is a synthetic compound with molecular weight of about 300 Da, one of the components of an Amazonian vine Uncaria tomentosa extract, which reduced aggregation of both A and tau. A Phase I clinical trial for AD was initiated in July 2008 (Thomson Reuters Pharma, update of June 8, 2012). Taisi (Beijing SL Pharmaceutical under license from Xuanwu Hospital of Capital Medical University), a capsule formulation of a stilbene analogue isolated from an unspecified organism, is in clinical studies in China since February 2011 (Thomson Reuters Pharma, update of June 29, 2012). W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes There are many natural products in preclinical evaluation as potential drugs for the treatment of AD (in alphabetical order): Alpha-mangostin, a polyphenolic xanthone derivative from mangosteen, concentration-dependently attenuated neurotoxicity induced by A1-40 or A1-42 oligomers (EC50 = 3.89 nM and 4.14 nM, respectively) [555]. Anatabine (Star Scientific, Glen Allen VI through its subsidiary Rock Creek Pharmaceuticals in collaboration with the Roskamp Institute) reduced A when applied to cells [556] (Thomson Reuters Pharma, update of January 3, 2012). Andrographis paniculata leaves extract showed cerebroprotective and nootropic activities in rats [557]. Apomorphine is an inhibitor of A fibril formation [558]. Apomorphine treatment of AD mice promoted A degradation [559]. Roles of apomorphine for regulated ␣-cleavage, autophagy, and antioxidation were discussed [560]. AX-00111 (Axonal Consultoria Tecnologica Ltda, Sao Paulo Brazil) is a plant-derived compound for the potential treatment of AD (Thomson Reuters Pharma, update of June 1, 2012). Axona (Accera Inc., Broomfield, CO) is a new medical food therapy for AD patients [561, 562]. It contains the proprietary formulation of mediumchain triglycerides, mostly caprylic triglyceride. The rationale for the use of Axona is based on the finding that the cerebral hypometabolism (i.e., impaired glucose metabolism) is an early sign of AD. Mediumchain triglycerides are metabolized in the liver resulting in the production of the ketone body betahydroxybutyrate, which is transported to the brain to provide an alternative fuel source for cerebral metabolism [563, 564]. See also [565]. Bacopa monnieri has shown memory free recall enhancing effects in adult humans [566]. Neuroprotective effects in experimental models of dementia were described [567]. Baicalein (5,6,7-trihydroxyflavone) protected cortical neurons from A25-35 -induced toxicity [568] and in a one dose pre-treatment at 5 and 10 mg/kg i.p. attenuated A25-35 -induced amnesia in mice in a stepthrough passive avoidance paradigm. Post-treatment for 7 or 13 days (10–15 mg/kg i.p.) also attenuated A25-35 -induced amnesia [569]. Baicalin prevented the production of hydrogen peroxide and oxidative stress induced by A aggregation in SH-SY5Y cells [570]. Apigenin (4 ,5,7-trihydroxyflavone), baicalein, and nordihydroguaiaretic acid were potent inhibitors of liposome permeabilization by A42 oligomers 21 [571]. Baicalein inhibited the formation of ␣-synuclein oligomers within living cells and prevented A peptide fibrillization and oligomerization [572]. Beta-asarone improved cognitive functions in rats after injection of A into the hippocampus [573] probably via JNK signaling and modulation of bcl-2 family proteins [574] and attenuation of neuronal apoptosis [575, 1708]. BT-11 is an extract of the dried root of Polygala tenuifolia (Willdenow) and effectively enhanced cognitive functions in elderly humans [576–578]. BV-7003 (Bioved Pharmaceuticals) is a natural product for the potential treatment of memory loss (Thomson Reuters Pharma, update of September 11, 2012). The structure was not communicated. Cabernet Sauvignon attenuated A neuropathology in a mouse model of AD [579]. Carvacrol showed cognition enhancing activity in two rat models of dementia [580]. Catechins showed potent anti-amyloidogenic and fibril-destabilizing effects in vitro [581–583]. Celastrol, a triterpenoid antioxidant compound isolated from the Chinese Thunder of God vine (T. wilfordii) reduced A pathology in a transgenic mouse model of AD [584]. Celastrus paniculatus seeds showed nootropic activity [585]. Chelerythrine showed promising cholinesterase inhibition, good inhibition of amyloid- aggregation and the ability to disaggregate preformed amyloid- aggregates [1709]. Cinnamon extract reduced A oligomerization and corrected cognitive impairment in animal models of AD [586]. Coumarins are naturally occurring -secretase inhibitors [587] and acetylcholinesterase inhibitors [588]. Cryptotanshinone (CTS), an active component of the medicinal herb Salvia miltiorrhiza, inhibited A aggregation and protected SH-SY5Y cells from damage by A [589]. It upregulated ␣-secretase by activation PI3K pathway in cortical neurons [590]. Curcumin showed potent anti-amyloidogenic effects for AD A fibrils in vitro and in vivo [591–605]. Clinical trials in AD patients showed disappointing results [350, 600, 606]. A novel nanoparticle formulation of curcumin (NanoCurc) was developed at the Indiana University School of Medicine [607]. For curcumin-decorated nanoliposomes see [608]. Structure-activity relationships of A aggregation inhibitors based on the curcumin scaffold were presented [609–611]. 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 from seeds of Apium and a powerful free radical scavenger [413]. It protected dopamine neurons in a rotenone model for PD [414]. DX-9386 is a traditional Chinese medicinal prescription, which improved thymectomy-induced impairment of learning behaviors in mice [612]. Ecdysterones, steroidal hormones in insects and terrestrial plants, inhibited A aggregation, disaggregated preformed fibrils, and inhibited A42 -induced cytotoxicity [613]. (-)-Epigallocatechin-3-gallate (EGCG; Sunphenon) present in green tea reduced A-mediated cognitive impairment presumably via flavonoidmediated presenilin-1 phosphorylation, which reduced A production [614–616]. EGCG prevented lipopolysaccharide-induced elevation of A generation [617]. EGCG remodeled mature ␣-synuclein and A fibrils and reduced cellular toxicity [618]. The cell signaling pathways and iron chelation were described [582, 619–624]. Also ERK and NF- κB pathways are involved [617]. The structural properties of EGCG-induced, nontoxic AD A oligomers were described [625]. EGCG functions through estrogen receptor mediated activation of ADAM10 in the promotion of non-amyloidogenic processing of APP [626, 627]). A special formulation of EGCG in nanolipidic particles to improve its bioavailability was presented [628]. See also Part 1, Chapter 1.9. Drugs interacting with estrogen receptors and Part 1, Fig. 10. ESP-102, a standardized combined extract of Angelica gigas, Saururus chinensis, and Schizandra chinenis, significantly improved scopolamine-induced memory impairment [629] and A1-42 -induced memory impairment in mice [630]. An aqueous extract of Eucommia ulmoides Oliv. Bark (EUE) showed beneficial effects on learning and memory impairments in mice [631]. Flavonoids in AD and neuroinflammation were discussed in depth [621, 632–635]. Some act as acetylcholinesterase inhibitors [636]. Biflavanoids were superior to monoflavonoids in inhibiting A toxicity [637]. Fortasyn Connect is a multi-nutrient diet comprising docosahexanoic acid (DHA), eicosapentenoic acid, uridine-mono-phosphate, choline, phospholipids, folic acid, vitamins B6, B12, C, and E and selenium. It reduced AD-like pathology in young adult APPSWE /PS1dE9 mice [638]. Fulvic acid inhibited aggregation and promoted disassembly of tau fibrils associated with AD [639]. Gallic acid from grape seed polyphenol extract may be useful for the treatment of AD [583]. Garlic extract attenuated the cytotoxicity of A on undifferentiated PC12 cells [640] and suppressed lipid peroxidation induced by A in PC12 cells [641]. It protected against A-induced apoptosis [642]. Garlic extract inhibited A fibril formation and defibrillated A preformed fibrils [643]. Aged garlic extract ameliorated the early cognitive deficits in Tg2576 mice [644, 645]. A review on the “aged garlic extract” was published [646]. Gastrodin protected primary cultured rat hippocampal neurons against amyloid- peptide-induced neurotoxicity via ERK1/2-Nrf2 pathway [1710]. Ginger root extracts (Zingiber officinale, Cognition Therapeutics) effected inhibition of A42 aggregation (Thomson Reuters Pharma, update of May 23, 2012). Gingko biloba (EGb 761, Tanakan, Tanamin) treatment prevented age-related spatial memory deficits in a transgenic mouse model of AD [647]. It enhanced adult hippocampal neurogenesis and phosphorylation of CREB [648]. For reviews on Ginkgo biloba extract and CNS functions, see [649, 650]. For results of clinical studies, see (in chronological order) [651–665]. Grape-derived polyphenolics from Vitis vinifera grape seeds attenuated cognitive deterioration in a mouse model of AD [666]. Ultrastructural alterations of AD paired helical filaments by grape seed-derived polyphenols was studied [667]. Guanosine protected human neuroblastoma cells from oxidative stress and toxicity induced by A peptide oligomers [668]. Hederacolchidide-E from Pulsatilla koreana showed cognition-enhancing and neuroprotective effects by reversing scopolamine-induced cognitive impairments in rats. It increased viability of human neuroblastoma SK-N-SH cells incubated with A42 [536]. Heme prevented A1-40 aggregation and its cytotoxicity [669]. Hopeahainol A attenuated memory deficits by targeting amyloid- in APP/PS1 transgenic mice [1711]. HSH-971 (Ocean University of China, Shandong) is a marine sulfated oligosaccharide for the potential treatment of AD [670]. (Thomson Reuters Pharma, update of November 7, 2011). HX-106 (HX-106N; ViroMed Co., Seoul) is a natural product inhibiting acetyl-cholinesterase for the potential treatment of AD (Thomson Reuters Pharma, update of June 13, 2012). The structure was not disclosed. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Hyperoside protected primary rat cortical neurons from neurotoxicity induced by A via the PI3K/Akt/Bad/Bcl(XL)-regulated mitochondrial apoptotic pathway [671]. IB-10C179(Instituto Biomar, Leon, Spain) is a compound derived from marine organisms which protected primary neurons from apoptosis and reduced free radical production (Thomson Reuters Pharma, update of March 7, 2012). The structure was not communicated. Icariin is a flavonoid isolated from Epimedii herba. It inhibited A25-35 induced expression of -secretase in rat hippocampus [672]. It attenuated A-induced neurotoxicity by inhibition of tau protein hyperphosphorylation in PC12 cells [673]. It attenuated lipopolysaccharide-induced microglial activation and resultant death of neurons by inhibiting TAK1/IKK/NF-κB and JNK/p38 MAPK pathways [674]. It improved memory impairment in AD model mice and attenuated A-induced neurite atrophy [675]. There seems to be a synergistic effect to improve learning and memory deficits in rats by co-administration of Icariin and Panax notoginseng saponins [676]. IDN-5706, a hyperforin derivative, decreased the content of acetylcholinesterase associated with different types of A plaques in 7-month-old double APPSWE /PS1 transgenic mice after treatment with IDN 5706 for 10 weeks [677]. Kaempferol protected PC12 and T47D cells from A toxicity [581, 678, 679]. Loganin isolated from Cornus officinalis showed cognitive-enhancing activity in scopolamine-induced amnesic mice [680]. Luteolin exerted ameliorating effects on Ainduced impairment of water maze performance and passive avoidance in rats [681–684]. Medical Food Cocktail consisting of the dietary supplements curcumin, piperine, epigallocatechin gallate, ␣-lipoic acid, N-acetylcysteine, B vitamins, vitamin C, and folate administered for 6 months to Tg2576 mice resulted in improvement of cognitive functioning [685]. Melatonin inhibited AD -fibrillogenesis [686–689]. The neuro-protective activities of melatonin against the A are not mediated by melatonin membrane receptors. The role of melatonin in AD-like neurodegeneration was described [690–693]. Melatonin facilitated short-term memory [694]. Menthol exerted a protective effect on A-induced cognitive deficits in mice [695]. Morin destabilized A42 protofibrils [696]. It inhibited the early stages of A aggregation [697]. It 23 attenuated tau hyperphosphorylation by inhibiting GSK-3 [698]. Myrcetin is a naturally occurring regulator of metalinduced A aggregation and neurotoxicity [699, 700]. Naringin showed memory enhancing activity in mice [701]. NeurocentRX Pharma is investigating a natural product-based compound for the treatment of cognitive diseases (Thomson Reuters Pharma, update of February 17, 2012). Nordihydroguaiaretic acid inhibited growth arising from direct A protofibril association [599, 702, 703]. O4 (an orcein-related small molecule) was able to convert toxic A oligomers to nontoxic -sheet-rich amyloid fibrils [704]. Obovatol, a biphenolic compound isolated from Magnolia obovata, attenuated scopolamine-induced cognitive dysfunctions [705], improved cognitive functions in animal models of AD [706], and attenuated LPS-induced memory impairments in mice via inhibition of NF-κB signaling pathway [707, 708]. Oleuropein and derivatives from olives were recognized as tau aggregation inhibitors [709]. Oren-gedoku-to exerts its potential use for the treatment of AD as a weak, reversible inhibitor of indoleamine-2,3-dioxygenase [710]. Oroxylin A is a flavonoid compound that is found in the root of Scutellaria baicalensis Georgi. It attenuated the memory impairment induced by transient bilateral common carotid artery occlusion in mice [711]. 1,2,3,4,6-penta-O-galloyl-beta-D-glucopyranose is the active constituent of the traditional medicinal herb Paeonia suffruticosa. It showed potent A anti-aggregation effects in vitro and in vivo [712]. Piceatannol showed protective effects against Ainduced neuronal cell death [713]. Pinocembrin, a flavonoid abundant in propolis, protected against A-induced toxicity in neurons [714]. Polyphenol derivatives (Pharma Eight, Nagoya) probably interacted with A through – stacking interactions 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 seedpod reversed memory impairment in cognitively impaired aged rats associated with decreased hippocampal CREB phosphorylation [717, 718]. Prosopis cineraria (L.) Druce (Leguminosae), a plant of the Thar Desert of India and Pakistan, is used traditionally by local people for the treatment of memory disorders and to arrest wandering of the mind. 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 the Morris water maze test, which may be attributed to the inhibition of brain acetylcholinesterase [719]. Puerarin, a phytoestrogen isolated from Pueraria lobata, attenuated A-induced cognitive impairment [720–723, 1712]. Quercetin-O-glucuronide significantly reduced the generation of amyloid- peptide [1713]. Pycnogenol protected neurons from A-induced apoptosis [724]. Quercetin showed protective effects against A42 in primary neurons [586, 725]). Rosmarinic acid protected PC12 cells from A-induced neurotoxicity [599, 726]. Subchronic administration of rosmarinic acid, a natural prolyl oligopeptidase inhibitor, enhanced cognitive performance [727]. Rutin inhibited A aggregation and cytotoxicity, attenuated oxidative stress, and decreased the production of nitric oxide and proinflammatory cytokines [728]. Saffron from Crocus sativus had an inhibitory effect on A aggregation [729, 730]. It was administered to 46 patients with probable AD in a 16-week double blind study (15 mg twice a day). Saffron was safe and effective [731]. A 22 week study corroborated the first results [732]. Saffron may be a source of novel acetylcholinesterase inhibitors [733]. Salidroside showed neuroprotective effects against A-induced oxidative stress in SH-SY5Y human neuroblastoma cells [734]. S-allyl-L-cysteine, the main constituent of garlic, protected against A-induced apoptosis and attenuated caspase-3 activation, DNA fragmentation and PARP cleavage [648]. It exerted protective effects on Ainduced cell death in NGF-differentiated PC12 cells [735] and protected against A-induced neurotoxicity in organotypic hippocampal cultures [736]. For a review on garlic extract and one of its active ingredients, S-allyl-L-cysteine, see [646]. Silibinin, a flavonoid derived from Silybum marianum, prevented memory impairment induced by A25-35 in the Y-maze and novel object recognition tests in mice [737]. Sinapic acid is a phenylpropanoid compound with anti-inflammatory and neuroprotective effects in a mouse model of A1-42 protein-induced AD [738]. Souvenaid (Nutricia, Châtel-St-Denis, Switzerland) is a mix of nutrients including the omega-3 fatty acid docosahexanoic acid, uridine monophosphate, and choline, dietary precursors for the synthesis of phospholipids. Two clinical trials, Souvenir I, which lasted 12 weeks, and Souvenir II, which lasted 24 weeks, were concluded [739–741]. Substance P, the tachykinin undecapeptide, protected cerebellar granule cells against A-induced apoptosis [742]. Syringin from the dried stem barks of Fraxinus rhynchophylla protected against A-induced toxicity in neuronal cells [743]. Tannic acid destabilized A fibrils in vitro [744, 745]. It is a natural -secretase inhibitor [746]. ␣-Tocopherol quinone inhibited A aggregation and cytotoxicity, disaggregated preformed fibrils and decreased the production of ROS, nitric oxide, and inflammatory cytokines [747]. Thymol showed cognitive-enhancing activity in two rat models of dementia [580]. Total coptis alkaloids produced a protective effect on A25-35 -induced learning and memory dysfunction in rats [748]. Ursolic acid attenuated A-induced apoptosis in a dose dependent manner [749]. Vitamin A has anti-oligomerization effects on A in vitro [750]. Vitamin B12 deficieny is associated with cognitive impairment [1714]. Vitamin B12 supplements administered orally or parenterally at high dose (1 mg daily) were effective in correcting biochemical defiency, but improved cognition only in patients with pre-existing vitamin B12 deficiency [1715]. Vitamin D defiency is associated with increased odds of cognitive impairment [1716–1718]. Withania somnifera (also known as Ashwagandha, an Indian ginseng) is a nootropic agent promoting cognition including memory [751]). W. somnifera administered once daily over 30 days reversed behavioral deficits, plaque pathology, accumulation of A peptides and oligomers in the brains of middle-aged and old APP/PS1 AD mice. Enhanced expression of low-density lipoprotein receptor-related protein (LRP) in brain microvessels and the A degrading protease neprilysin occurred 14–21 days after a substantial decrease in brain A levels [752]. Wuzi Yanzong Granule improved memory functions in patients with MCI [753]. Yokukansan (TJ-54), a traditional Japanese herbal medicine, was tested in clinics for potential antidementia effects [754]. Spatial memory in a rat model of early AD was improved [755]. Zokumei-to (ZMT) attenuated A25-35 -induced memory impairment [756]. The Central Drug Research Institute (India) was developing bacosides A and B, which are saponin W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes nootropic agents. The compounds were in Phase II clinical evaluation, but it appears that the development was terminated. Also the development of CBNU06 (Chungbuk University), DCB-AD1 (Development Center for Biotechnology), dysiherbaine (Daiichi Suntory Biomedical Research Co., a compound isolated from the Micronesian sponge Dysidea herbacea [757]), of the nordihydroguaiaretic acid derivative EM-4232 (Eromos Pharmaceutical LLC), HSS808, HSS-818, HSS-838, HSS-848 and HSS-888 (HerbalScienceLLC Singapore; standardized turmeric curcuma longa extracts), of LHM-123 (Mazal Plant Pharmaceuticals), MDF-004 and MDF-005 (Neuron BioPharma), and of YY-1224 and YY-1824 (Yuyu; orally available terpene trilactones extracted from Ginkgo biloba [758]) was terminated. NOOTROPICS (“DRUGS WITHOUT MECHANISM”) In this chapter compounds are described, whose mechanism(s) of action are unknown. Initiation of clinical trials was based on positive effects on impaired brain functions in experimental animals after proof of good tolerability. LSL-001 (Laboratoire S. Lasnier, Etaules, France) is a nootropic compound in Phase II clinical trials since June 2011 for the potential treatment of AD (Thomson Reuters Pharma, update of June 5, 2012). The structure was not disclosed. N-251 (Neurokos Inc., Palo Alto, CA) is a compound of an undisclosed mechanism of action for the potential treatment of AD. Phase II clinical trials started in May 2009 (Thomson Reuters Pharma, update of January 16, 2012). The structure of N-251 was not communicated. PF-03049423 (Pfizer) is a neurorestorative compound for the potential treatment of neurological disorders including stroke recovery. In December 2010, a randomized, double-blind, placebo-controlled Phase II trial was initiated in the US and in South Korea (n = 240) (Thomson Reuters Pharma, update of August 13, 2012). The structure was not communicated. TPM-189 (Teikoku Pharma USA, San Jose, CA) is a transdermal formulation of a small molecule therapeutic for the potential treatment of AD in Phase II development (Thomson Reuters Pharma, update of May 17, 2012). The structure was not communicated. VI-1121 (VIVUS Inc., Mountain View, CA) is a nootropic agent in Phase II clinical trials (n = 50) since August 2011 in the US (NCT01428362) (Thomson 25 Reuters Pharma, update of September 9, 2011). The structure of VI-1121 was not communicated. ASP-0777 (Astellas Pharma) is in Phase I clinical trials since May 2009 (Thomson Reuters Pharma, update of September 19, 2012). The structure was not disclosed. Lilly is developing a small molecule for the treatment of cognitive impairment in schizophrenia in Phase I clinical trials since July 2012. (Thomson Reuters Pharma, update of November 13, 2012). The structure was not disclosed. RO-5508887 (Roche) is a nootropic agent in Phase I clinical trials in healthy male volunteers in France since October 2011 (Thomson Reuters Pharma, update of August 16, 2012). The structure was not disclosed. SEP-363856 (Sunovion Pharmaceuticals, Marlborough MA and Sumitomo Chemical) is an orally active compound with novel mechanism of action for the treatment of schizophrenia. The drug is in Phase I in the US. (Thomson Reuters Pharma, update of November 5, 2012). The structure was not disclosed. TAK-357 (Takeda) is a cognitive enhancer with unspecified 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 preclinical evaluation (in alphabetical order): AC-0523 (Neuera, a subsidiary of Accera, Broomfield, CO) (Fig. 11) is indicated for the treatment of mitochondrial dysfunction-related HD (Thomson Reuters Pharma, update of February 15, 2012). AFX-929 (Afecta Pharmaceuticals; Irvine, CA) is a nootropic agent in preclinical development (Thomson Reuters Pharma, update of November 10, 2011). The structure was not disclosed. Alzheimer’s disease therapeutics (Berg Pharma, Nashville TN) are nootropic compounds for the potential treatment of AD (Thomson Reuters Pharma, update of April 17, 2012). The structures were not disclosed. Alzheimer’s disease therapeutics (SienaBiotech/ Roche) are neuroprotectant compounds for the potential treatment of AD (Thomson Reuters Pharma, update of February 8, 2012). The structures were not disclosed. Alzheimer’s disease therapy (Reyon Pharmaceuticals, Seoul) is a therapeutic program for the potential treatment of AD (Thomson Reuters Pharma, update of June 20, 2012). The structures were not disclosed. AVN-457, AVN-458, and AVN-492 (Avineuro Pharmaceuticals, San Francisco, CA) are nootropic agents 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 Thomson Reuters Pharma, update of August 19, 2011). The structures 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., Choongwae Holdings, Seoul) is a nootropic agent in development for the Korean and Japanese markets (Thomson Reuters Pharma, update of May 11, 2012). The structure was not disclosed. D-130 (Envoy Therapeutics, Jupiter, FL) is a compound targeting the cortex region of the brain using bacTRAP technology for the potential treatment of cognition deficits (Thomson Reuters Pharma, update of June 1, 2012). The structure was not disclosed. D-180 (Envoy Therapeutics, Jupiter, FL) is a compound targeting the striatum region of the brain using bacTRAP technology for the potential oral treatment of PD (Thomson Reuters Pharma, update of September 12, 2012). The structure was not disclosed. GSK-2647544 (GSK) is a neuroprotectant for the potential treatment of Alzheimer’s disease. In October 2012, a single-blind, randomized, placebo-controlled, Phase I trial was expected to begin later that month in Australia in healthy male subjects (expected n = 16). At that time, the trial was expected to complete in April 2013 (Thomson Reuters Pharma, update of November 20, 2012). The structure was not disclosed. J-147 (Salk Institute for Biological Studies) (Fig. 11) is a drug that improved memory in normal rodents and prevented cognitive decline in transgenic mice of AD (Thomson Reuters Pharma, update of December 22, 2011). JAY 2-22-33 (Medical College of Georgia) (Fig. 11) significantly reduced A toxicity and improved cognitive performances in transgenic AD mice [759]. JWB1-84-1 (Medical College of Georgia) (Fig. 11) is a tertiary amine analogue of choline from the laboratory of the late Prof. Jerry J. Buccafusco. It produced a dose-dependent decrease in the number of errors made by well-trained AD-transgenic mice in the radial arm water maze test [759, 760]. KD-901 (Kwang Dong Pharmaceutical Co., Seoul) is a nootropic drug in preclinical evaluation (Thomson Reuters Pharma, update of March 1, 2011). The structure was not disclosed. KU-046 (Kareus Therapeutics and Connexios Life Sciences, Atlanta, GA) is a combination drug, which demonstrated significant improvement of cognition (Thomson Reuters Pharma, update of May 29, 2012). The structure was not disclosed. LNK-3186 and LNK-3248 (AstraZeneca after its acquisition of Link Medicine Corp., Waltham, MA) are nootropic compounds (For both Thomson Reuters W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Pharma, update of July 13, 2012). The structures were not disclosed. Maltoyl p-coumarate attenuated cognitive deficits in rats treated with scopolamine or with A42 [761]. MeN061016-1 (Lijun International Pharmaceutical, 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 Pharmaceuticals, Emmetten, Switzerland) is an intranasal gel formulation of pregnenolone for the potential enhancement of memory (Thomson Reuters Pharma, update of January 27, 2012). NNZ-2591 (Neuren Pharmaceuticals, Auckland, NZ) (Fig. 11) is an orally active neuroprotectant diketopiperazine derivative for the treatment of brain injury and PD [762] (Thomson Reuters Pharma, update of August 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) is a novel small molecule neuroprotectant for the potential treatment of CNS disorders, including Alzheimer’s disease, Parkinson’s disease and age-related cognitive decline (Thomson Reuters Pharma, update of October 29, 2012). The structure was not disclosed. OG-635 (Oryzon Genomics, Barcelona) is a nootropic agent for the potential treatment of AD and PD (Thomson Reuters Pharma, update of May 31, 2012). The structure was not disclosed. Pentylenetetrazole (Balance Therapeutics, Hillsborough, CA) is investigated for the potential treatment of cognitive impairment in Down’s syndrome (Thomson Reuters Pharma, update of September 4, 2012). PNB-03, PNB-04, and PNB-05 (PharmaNeuroBoost NV, Limburg, Belgium) are nootropic agents for the potential treatment of PD (PNB-03), AD (PNB-04), and obsessive compulsive disorders (PNB-05) (Thomson Reuters Pharma, update of February 1, 2012). The structures were not communicated. PTI-125 (Pain Therapeutics Inc., Austin, TX) reduced A-related AD pathogenesis by targeting filamin A [763] (Thomson Reuters Pharma, update of August 3, 2012). The structure was not communicated. RP-4000 (Reviva Pharmaceuticals, San Jose, CA) is a small molecule nootropic agent (Thomson Reuters Pharma, update of December 27, 2011). The structure was not communicated. SEL-103 (Selvita Life Sciences Solutions, Krakow, Poland and Orion, Espoo, FL) is a program inves- 27 tigating small molecule nootropic agents (Thomson Reuters Pharma, update of July 4, 2012). Structures were not communicated. SKL-A4R (SK Biopharmaceuticals, formerly SK Life Science, Fair Lawn, NJ) is a small molecule nootropic agent (Thomson Reuters Pharma, update of March 19, 2012). The structure was not communicated. TYP-1 (NOBEL ILAC, Istanbul, Turkey) is a small molecule neuroprotectant (Thomson Reuters Pharma, update of July 30, 2011). The structure was not communicated. The development of ABIO-08-01 (BTG-1640; Abiogen under license from British Technology Group, BTG), ADS-8703 (Adamas Pharmaceuticals), AIP-002 (American Home Products, Wyeth, now Pfizer), AIT-034 (Spectrum Pharmaceuticals, formerly NeoTherapeutics), Alzheimer’s disease therapeutics (DuPont/Scios), ALE-26015 (Allelix Pharm-Eco LP), aloracetam (Hoechst, now sanofi), Alzene (Bar Ilan University and Ivax); AQW-051 (Novartis; Phase II trials for treatment of schizophrenia and PD are ongoing), ASP-2535 and ASP-2905 (Astellas), AVN-397 (Avineuro Pharmaceuticals; in Phase II trials for the treatment of anxiety), AWD-52-39 (Alzneimittelwerke 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-200406 (RS-0406; SEN-1269 [767]; Senexis under license from BTG under license from Sankyo), BGC20-0466 (BTG), BGC-20-1178 (Senexis under license from 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; Lederle Laboratories, now Pfizer), of CLL-502 (CLL Pharma), CM-2433 (Cenomed, a subsidiary of Abraxis BioScience in collaboration with Medical College of Georgia), CX-417, CX-423 and CX-438 (Cortex Pharmaceuticals), DW-514 and DW-0811 (Daewon Pharm), DWJ-209 (Daewoong Pharmaceutical Co.), of the coumarin derivative ensaculin (KA-672; Dr. Willmar Schwabe [771–777]), ETX-6765 (eTherapeutics), F-94517 (Pierre Fabre), gedocarnil (Bayer Schering Pharma), GM-1416 (GliaMed Inc.), GT-4035 and GT-4054 (Gliatech), HL-026 (HanAll Biopharma), HMR-2420 (Hoechst Marion Roussel, now sanofi), HSB-13 (EncephRx, a spin-out of the Southern Methodist University and the University of Texas [1719]), HTC-867 (Wyeth, now Pfizer), IQ200 and IQ-201 (Immune Network under license from 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, Amarin Corp.), of the MBARC program (EnVivo Pharmaceuticals), MK-0249 (Merck & Co), MK-5757 (Merck & Co), MG-19649 (Molecular Geriatrics, now Applied Neuro Solutions), MP-100 (Addiction Therapeutix), MR-708 (Medea Research), NDD-094 (Novartis [778]), NeoEA-1001 (neoCodex), nooglutil (Russian Academy of Medical Sciences [779]), NSA789 (Wyeth, now Pfizer), OF-5858 and OF-6145 (All Russian Research Institute of Pharmaceutical Chemistry), Oligotropin (HF-0420; Loyola University of Chicago), Org-31433 (Organon Biosciences, now MSD OSS BV), PF-05297909 (Pfizer), Prisotinol (CGS-5649B; Ciba-Geigy, Novartis [780–783]), procaine hydrochloride (Samaritan Pharmaceuticals), PRX-4001 and PRX-4006 (Proximagen), R-641 and Ro-40-1641 (both Roche), RU-47067 and RU-52583 (Roussel-Uclaf, now sanofi), sabeluzole (Janssen Pharmaceutica NV, Johnson & Johnson [784–789]), SCH-54388 (Schering-Plough, now Merck; a metabolite of felbamate), sibopirdine (EXP921; Du Pont Merck [790]), silicon-containing pyridylsubstituted isoxazoles (Latvian Institute of Organic Synthesis), SLV-351 (Solvay Pharmaceuticals, now Abbott Laboratories), SN-104 (Sention), SNK-882 (Sanwa Kagaku Kenkyusho), SPPI-339 (SPI-339, NEO-339; Spectrum Pharmaceuticals, formerly NeoTherapeutics), SRA-997 (Novartis), ST-587 (Boehringer Ingelheim), T-9021 and T-9022 (QRxPharma), TAK065 (Takeda), tenilsetam (Hoechst Marion Roussel, now sanofi [791, 792]) trifusal (Grupo Uriach), V-0191 (Pierre Fabre) and of Z-4105 (Zambon) was terminated. PEPTIDES Cerebrolysin (Ebewe Pharmaceuticals, Vienna, Austria, launched, available in 1 ml, 5 ml, and 10 ml ampoules and in vials of 30 ml and 50 ml for intramuscular or intravenous injection or intravenous infusion) is “derived through a biotechnological procedure from highly purified porcine brain proteins and is comprised of free amino acids and biologically active, short chain peptides” [793]. Cerebrolysin acted as a presynaptic GABAB receptor agonist [794]. The nootropic effects of cerebrolysin were investigated in depth [795–799]. Cerebrolysin protected neurons from ischemia-induced loss of microtubule-associated protein 2 [800]. Effects of cerebrolysin on A deposition in transgenic mice were studied extensively [801–807]. The pharmacology of neurotrophic treatment with cerebrolysin was reviewed [808]. Clinical data were published (in chronological order) [793, 809–823]. First clinical results of a combination treatment of cerebrolysin and donepezil were reported [824]. Data of cerebrolysin in AD were compiled [825]. The safety profile of cerebrolysin in dementia and stroke trials was described [826]. Cortexin (Geropharm, St. Peterburg, Russia) is a launched polypeptide with neuroprotective, nootropic and antioxidant properties, for the intramuscular treatment of central nervous system injury caused by cerebrovascular accidents, encephalitis, encephalopathies, epilepsy and trauma (Thomson Reuters Pharma, update of October 05, 2012). The structure was not communicated. Davunetide (intranasal, AL-108, NAP, Allon Therapeutics, Vancouver) is an 8-amino acid peptide (NAPVSIPQ) derived from the activity-dependent neuroprotective protein. A Phase III clinical trial was initiated for the treatment of patients with progressive supranuclear palsy in December 2010. A Phase II clinical trial in AD patients started in January 2007 (Thomson Reuters Pharma, update of July 17, 2012). Allon Therapeutics is also developing injectable intravenous and subcutaneous formulations of davunetide. There are numerous publications on the extensive preclinical characterization of davunetide describing protection of the brain against ischemic injury in rats [827], inhibition of the aggregation of A [828], decrease of anxiety-like behavior in aging mice [829], reduction of the severity of traumatic head injury [830], reduction of accumulation of A and of tau hyperphosphorylation [831–833], stimulation of microtubule assembly [834, 835], enhancement of cognitive behavior in transgenic mice [836]), and improvement in motor function and reduction of ␣-synuclein inclusions in mice overexpressing ␣-synuclein [837]. Reviews on davunetide were published [838–847]. The effects of davunetide on cognition and functional capacity in schizophrenia were described [848] (Thomson Reuters Pharma, update of September 21, 2012). AM-111 (XG-102, D-JNKI-1; Auris Medical and Xigen, a spin off from the Centre Hospitalier Universitaire Vaudois and the University of Lausanne) is an injectable protease-resistant peptidic derivative of the JNK-inhibiting protein IB-1 for the i.v. treatment of acute sensorineural hearing loss, stroke, and AD [849–852]. A Phase IIb trial for acute sensorineural hearing loss was initiated in January 2009 (n = 210) and results are expected in autumn of 2012. Auris Medical W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes is also investigating a topical gel formulation of AM111 (Thomson Reuters Pharma, update of August 31, 2012). Etanercept (ENBREL, NK-001; Neurokine Pharmaceuticals, Vancouver) is a recombinant fusion protein of 934 amino acids (mol. weight: 150 kDa) composed of a dimer of the extracellular portion of human TNFR-2 fused to the Fc fragment of human IgG1 [853, 854]. A rapid cognitive improvement following perispinal etanercept administration to 15 probable-AD patients treated once weekly for 6 months was reported [855–857]. See also the commentary [858]. In December 2010, the drug was in Phase II development for the potential treatment of neurocognitive impairment following coronary artery bypass graft surgery (Thomson Reuters Pharma, update of July 9, 2012). FGL (ENKAM Pharmaceuticals, Copenhagen, Denmark) is a peptide neural cell adhesion molecule mimetic for the potential treatment of AD and stroke. In December 2011, clinical studies including three Phase I studies, one proof-of-concept Phase IIa study in AD, and a pilot study in patients recovering from stroke was planned to begin in 2012 (Thomson Reuters Pharma, update of April 23, 2012). Enkam was previously developing FGLL, a peptide neural cell adhesion molecule mimetic, for the potential intranasal treatment of AD and stroke. In May 2005, the drug was shown to be safe and well tolerated in a Phase I study for AD. By December 2011, the drug was no longer being developed due to the lack of flexibility for the route of administration. Glypromate (Gly-Pro-Glu) is naturally cleaved from the N-terminal sequence of IGF-I and displayed neuroprotective actions in vitro and in vivo [859–861]. It protected against A-induced somatostatin depletion in rat cortex [862, 863]. NNZ-2566 (Neuren Pharmaceuticals, Auckland, NZ) (Fig. 11) is an analogue of glypromate with an additional ␣-methyl-group on the proline moiety, which resulted in an improved half-life and better oral bioavailability. In March 2012, a randomized, double-blind, placebo-controlled Phase I study was initiated in healthy volunteers (n = 32) in Australia. Potential indications are mild traumatic brain injury, Rett syndrome, PD, and AD [864– 867]. Excellent synthetic organic chemistry was described [868–871]. In November 2012 the IND was filed for a Phase II trial. At that time an application was submitted to the Texas Children’s Hospital IRB and enrollment was expected to begin pending approval by the FDA and the Texas Children’s Hospital IRB. (Thomson Reuters Pharma, 29 update of November 23, 2012). (Thomson Reuters Pharma, update of August 31, 2012). Neuren is also developing a formulation for intravenous infusion. There are many peptides in preclinical evaluation for the potential treatment of AD (in alphabetical order): AL-408 (Allon Therapeutics, Vancouver) is the orally active D-amino acid derivative of davunetide (AL-108) and an active element of the PARP-1 activating activity-dependent neuroprotective protein (Thomson Reuters Pharma, update of November 9, 2011). Alzimag (IMAGENIUM, Paris, France) is a peptide for the potential treatment of AD (Thomson Reuters Pharma, update of December 22, 2011). The structure of the compound were not communicated. C3bot peptides (Charité Medical School Berlin and Hannover Medical School) are short neuron-specific neuritogenic peptides derived from the C3 exoenzyme of Clostridium botulinum, which transiently activated RhoA for the potential treatment of neurodegenerative 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 (Cognosci Inc., Research Triangle Park, NC) are apoE-mimetic peptides that showed consistent reduction of both A deposition and of tau hyperphosphorylation in three tau transgenic mouse models and in two APP transgenic mouse models [876, 877] (Thomson Reuters Pharma, update of June 5, 2012 for COG-112 and of September 11, 2012 for COG-133 and COG-1410). G-79 (BN-201; Bionure, Barcelona) is a peptide for the potential treatment of multiple sclerosis, ALS, AD, PD, and glaucoma (Thomson Reuters Pharma, update of July 4, 2012). KIBRA pathway modulators (Amnestix Inc., a wholly owned subsidiary of SYGNIS Pharma; Heidelberg, Germany) offer a new genetic link to cognition that may benefit patients suffering from AD. For the association of KIBRA and late onset AD, see [878]; for enhancement of cognition in anxiety disorders [879] (Thomson Reuters Pharma, update of August 14, 2012). Leptin reduced the accumulation of A and phosphorylated tau in rabbit organotypic slices [880–882]. The company Neurotez (Bridgewater, NJ) is investigating leptin as an A synthesis inhibitor for the potential treatment of AD. The company planned to file an IND in 2012. Effects of leptin on memory processing were described [883–885]. Leptin induced proliferation of neuronal progenitor cells [886] (Thomson Reuters Pharma, update of August 24, 2012). 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 fragments; Socratech LLC, Rochester NY) is produced by stable transfected baby hamster kidney cells expressing LRP-IV. The preparation improved cerebral blood flow, 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 with APP and regulated A production [887, 888]. Netrin1 increased soluble APP␣ and decreased A1-40 and A1-42 levels in a J20 mouse model of AD. An intranasal delivery is planned (Thomson Reuters Pharma, 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 for the potential treatment of multiple sclerosis and motor neuron disease, respectively [889] (Thomson Reuters Pharma, updates of June 18, 2012 and September 7, 2012, respectively). NRG-101 (Mind-NRG, Geneva, Switzerland under license from ProteoSys, Mainz, Germany) is an injectable neuregulin peptide for the potential treatment of PD, AD, and schizophrenia (Thomson Reuters Pharma, update of July 6, 2012). NT-1 and NT-2 (Neurotez, Bridgewater, NJ) are small peptides, which block the interaction between mutant presenilin and cytoplasmic linker protein 170 (CLIP-170) that is linked to increased A levels (Both Thomson Reuters Pharma, update of June 6, 2012). NX-210 (Neuronax, Saint Beauzire, France) is the lead compound of a series of peptides for the potential treatment of spinal cord injury and other neurological disorders, such as stroke, AD, PD, and traumatic brain injury (Thomson Reuters Pharma, update of June 29, 2012). The structure was not communicated. Pepticlere (DP-68 and DP-74; ProteoTech, Kirkland, WA) is the name of small molecule nasal sprays, 6- to 9-mer peptides that inhibit A fibril formation (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, Switzerland) is a small stabilized receptor active peptide targeting the CCR5 receptor for the potential treatment of AD (Thomson Reuters Pharma, update of October 1, 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 neurodegenerative diseases including AD (Thomson Reuters Pharma, update of June 27, 2012). The structures were not communicated. SX-AZD1 (Serometrix, Pittsford, NY) is a peptide mimetic interacting with APOE4 (Thomson Reuters Pharma, update of July 29, 2011). The structure was not communicated. XD4 is a heptapeptide isolated from a Ph.D.-C7 library through phage display that rescued memory deficits in AD transgenic mice by significantly inhibiting A42 -induced cytotoxicity, increasing microglial phagocytosis of A, and decreasing A-induced generation of ROS and nitric oxide [890]. Colostrinin (ReGen Therapeutics, London) is a polypeptide complex isolated from ovine colostrum containing a high proportion of proline (25%) and hydrophobic amino acids (40%). It is composed of peptides of molecular mass up to 3,000 Da. It reduced aggregation of A [891, 892] and alleviated Ainduced toxicity [893]. It facilitated learning and memory in rats [894] and chicks [895]. Effects on gene expression were described [896]. Clinical results were presented (in chronological order) [897–901]. Reviews on colostrinin were published [902, 903]. The development of colostrinin for the treatment of AD patients was discontinued. The compound was launched as a nutraceutical in Australia in July 2007 (Thomson Reuters Pharma, update of May 17, 2011). The development of ANA-1 and ANA-5 (Alzhyme Pty; phage peptides, which bind to A to inhibit its generation of hydrogen peroxide [904]), adrenomedullin peptides (National Institutes of Health), AS-602704 (Merck Serono under license from Axonyx), C-AVP(4-9) (Yakult Central Institute for Microbiological Research), ebiratide (Hoe-427; Hoechst, now sanofi; an ACTH (6-9) derivative [905–907]), gilatide (a nonapeptide of Axonyx under license from Thomas Jefferson University), metallothionein (Neurosciences Victoria and the University of Tasmania [908, 909]) and of noopept (GVS-111, SGS-111; DVD-111; Saegis Pharmaceuticals under license from the Russian Academy of Sciences; a nootropic dipeptide analogue of piracetam [910–920]) was terminated. The development of NC-1900 (Nippon Chemiphar, the active fragment analog of arginine vasopressin [921–925]), PR-21C (Pharmaxon; a polysialylated form of the neural cell adhesion molecule) and of RGX-100 and RGX200 (RemeGenix Inc., Cortica Neuroscience; A production-inhibiting BRI2-derived peptides for the intranasal administration to AD patients [926–933]) W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes was discontinued. The development of SEM-606 (University of Manchester) and of TKP-1001 (EUSA Pharma, formerly Tasliker under license from The Open University) was terminated. DRUGS PREVENTING AMYLOID- AGGREGATION Plasma A levels can be linked directly to specific cognitive changes that constitute the conversion from MCI to AD [934–939]. The relationship between atrophy and A deposition in AD was investigated thoroughly [940–942]. A review of the literature correlating Alzheimer disease neuropathologic changes with cognitive status was provided [947]. The molecular and neurophysiological mechanisms of amyloid- and cognition in Alzheimer’s disease were outlined [1720]. A rapid decline of memory in healthy older adults 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 mediated rapid disruption of synaptic plasticity and memory [943]. The topic “A toxicity in Alzheimer’s disease” was reviewed [944, 945]. A critical review on the amyloid cascade hypothesis was published [946]. Although A plaques may play a key role in AD pathogenesis, the severity of cognitive impairment correlates best with the burden of neurofibrillary tangles [947]. The key interactions of apolipoprotein E and A pathology were reviewed [948]. The inhibition and reversion of A misfolding and aggregation is an approach, which has been followed up by many research groups during years. Excellent reviews dealing with this subject were published (in chronological 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 Pharmaceuticals under license from the Scripps Research Institute) (Fig. 12) is small-molecule transthyretin stabilizer for the oral treatment of transthyretin familial amyloid polyneuropathy [977–980]. The drug was approved by EMA in November 2011 was launched in Europe in March 2012 [1724]. In June 2012, the FDA issued a complete response letter and requested the completion of a second efficacy study. For the identification of A binding sites on transthyretin see [981]; 31 for transthyretin amyloidosis see [982, 983] (Thomson Reuters 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 mimetic designed to inhibit the formation and deposition of amyloid fibrils. A Phase III study was initiated in December 2010 for the treatment of renal disease in patients with AA amyloidosis [984–987] (Thomson Reuters Pharma, update of September 5, 2012). For ARC-029 (Archer Pharmaceuticals, Roskamp Institute, 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 administration; Allon Therapeutics, Vancouver, Phase III clinical trials), see Chapter 15, Peptides. For APH-0703 (Aphios Corp., Woburn MA), a nanoparticle formulation of APPH-9601 in Phase II clinical trials since May 2010 (Thomson Reuters Pharma, update of July 5, 2012), see Part 2, Chapter 2.35, Drugs interacting with Protein Kinase C. Doxycycline hyclate (Fondazione IRCCS Policlinico San Matteo, Pavia, Italy) is in a Phase II study (NCT01171859) in patients with transthyretin amyloidosis (n = 40) since July 2010. At oral doses of 200 mg for three months, doxycycline produced a significantly lower decline in the ADAScog scores in 101 mild-to-moderate AD patients [988] (Thomson Reuters Pharma, update of April 27, 2012). ELND-005 (AZD-103; scyllo-inositol; scyllocyclohexanehexol, Elan, Dublin, Ireland, and Ellipsis Neurotherapeutics, formerly Transition Therapeutics, Toronto, Canada) (Fig. 12) is an orally available inhibitor of A peptide aggregation. Results of a Phase II dose-ranging, randomized, placebo-controlled study in 351 patients with mild-to-moderate AD treated for 78 weeks with either 250, 1,000, or 2,000 mg/day were reported. Patients with mild AD receiving 250 mg of ELND-005 had higher scores on the Neuropsychological Test Battery compared with placebo, which were significant. In contrast ADCS-ADL scores were not significant. The two higher doses were discontinued [989]. In August 2012, a Phase II study was initiated in bipolar disorder patients in the US. The study is expected to be completed in August 2014 (Thomson Reuters Pharma, update of September 14, 2012). The endogenous brain inositols stabilized small aggregates of A, which are non-toxic to NGFdifferentiated PC-12 cells and primary human neuronal 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-. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes cultures [990]. In particular scyllo-inositol, when given orally to transgenic mice, reversed AD phenotype, improved impaired cognition, and altered cerebral A pathology [991, 992]. Scyllo-inositol dose-dependently rescued long-term potentiation in mouse hippocampus from the inhibitory effects of soluble oligomers of cell-derived human A [993, 994]. Elevated scyllo-inositol concentrations were found in patients with AD [995]. Syntheses of scyllo-inositol derivatives have been reported [996]. The synthesis of the PET ligand [18 F]1-deoxy-1-fluoro-scyllo-inositol was described [997]. A comparison of three amyloid assembly inhibitors, scyllo inositol, epigallocatechin gallate and the molecular tweezer CLR01 (vide infra) was published [998]. SOM-0226 (SOM Biotech SL, Barcelona) is a drug for the potential treatment of transthyretin amyloidosis in Phase II clinical development (Thomson Reuters Pharma, update of May 30, 2012). The structure was not communicated. For AAD-2004 (GNT Pharma, Suwon, South Korea) in Phase I clinical trials (Thomson Reuters Pharma, update of May 4, 2012). See Part 2, Chapter 2.34. Drugs interacting with Prostaglandin D & E Synthases, Part 2, Fig. 22. Beta amyloid modulator (Medipost, Seoul) is in Phase I clinical trials in South Korea since September 2011 for the potential treatment of AD (Thomson Reuters Pharma, update of May 7, 2012). The structure was not communicated. BLU-8499 (formerly NRM-8499; Bellus Health, formerly Neurochem, Quebec) is a prodrug of tramiprosate (vide infra). It was evaluated in a singlecenter, randomized, double-blind, placebo-controlled Phase I study in 84 young and elderly healthy subjects to assess safety, tolerability and pharmacokinetics, which started in March 2010. The data were reported in January 2011. It showed improved gastrointestinal tolerability and lower inter-individual variability of drug exposure compared to comparable doses of tramiposate (Thomson Reuters Pharma, update of September 5, 2012). DWP-09031 (presumed to be DWJ-301; Daewong Pharmaceutical Co. in collaboration with Medifron, both South Korea) inhibited the production and aggregation of A. By January 2012, the Korean FDA had approved an IND for a Phase I trial. The randomized, double-blind, placebo-controlled study (NCT01522586) is planned in healthy male volunteers (n = 64) to assess the safety, pharmacokinetics and pharmacodynamics of DWP-09031 (Thomson Reuters 33 Pharma, update of July 27, 2012). The structure was not disclosed. For Exebryl-1 (ProteoTech, Kirkland, WA and China licensee Tasly Pharmaceuticals; in Phase I clinical trials), see Chapter 13, Natural Products. For NP-61 (NP-0361; Noscira, previously Neuropharma, Madrid; Phase I clinical trials; Thomson Reuters Pharma, update of May 10, 2012), see Part 2, Chapter 2.1.1.2., Dual acetylcholinesterase and A inhibitors. Systebryl (ProteoTech, Kirkland, WA) is the lead of a series of small molecules including PTI-19 and PTI-51 for the potential treatment of systemic AA amyloidosis. In December 2011, Systebryl was in Phase I studies (Thomson Reuters Pharma, update of December 21, 2011). Structures were not communicated. There are many A aggregation inhibitors in preclinical evaluation (in alphabetical order): Alzheimer’s disease therapeutics (BioChromix Pharma AB, Solna, Sweden) are compounds, which achieved a significant reduction in plaque load and neurotoxic A aggregates. (Thomson Reuters Pharma, update of August 24, 2012). The structures were not communicated. A oligomer cellular prion protein binding inhibitors (AstraZeneca under a sublicense from Axerion Therapeutics under license from Yale University) are interacting with the cellular prion protein, the high affinity receptor for A oligomers [999–1003] (Thomson Reuters Pharma, update of August 17, 2012). No structures were disclosed. A protein inhibitors (Neuropore Therapies, San Diego CA) are peptidomimetic compounds that interfere with A aggregates (Thomson Reuters Pharma, update of July 16, 2012). Structures were not communicated. A/tau protein aggregation inhibitors (CNRS, FIST SA, Paris, France) are investigated for the potential treatment of AD (Thomson Reuters Pharma, update of August 24, 2012). Structures were not communicated. Amyloid-derived diffusible ligands are investigated by Merz Frankfurt, Germany under license from Acumen Pharmaceuticals (Thomson Reuters Pharma, update of June 12, 2012). The structures of the compounds were not communicated. ARN-4261 (New York University and Aria Neurosciences, Hamden CT) (Fig. 12) and ARN-2966 (Fig. 12) are inhibitors of A aggregation. ARN-2966 reduced A deposition and memory deficit in transgenic mice. Following treatment a significant 25% reduction in A plaque load was noted compared with 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 demonstrated that ARN-2966 was biostable and well tolerated in transgenic mice with blood-brain barrier penetration after both oral and intravenous dosing (Thomson Reuters Pharma, update of August 28, 2012). AVCRI-104P4 (University of Barcelona) (Fig. 12) inhibited A aggregation including acetylcholinesterase- and self-induced-A aggregation, -secretase, acetylcholinesterase, and butyrylcholinesterase (Thomson Reuters Pharma, update of September 28, 2012). AVN-457, AVN-458 and AVN-492 (Avineuro Pharmaceuticals, San Francisco) are nootropic agents for the potential treatment of cognitive disorders (Thomson Reuters Pharma, update of October 22, 2012). The structures were not disclosed. AZP-2006 (AlzProtect, Loos, France in collaboration with INSERM and the University of Lille II) is an APP modulator of undisclosed structure (Thomson Reuters Pharma, update of July 4, 2012). Beta-amyloid aggregation inhibitors (Medisyn Technologies, Minnetonka, MN / Mount Sinai School of Medicine) demonstrated significant A-lowering and anti-aggregation activity in vitro. Two compounds also 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 aggregation inhibitors (Snowdon Inc., Vancouver) are expected to enter Phase I trials in 2012 (Thomson Reuters Pharma, update of August 19, 2011). The structures of the compounds were not communicated. Beta amyloid beta sheet formation inhibitors (AC Immune, Lausanne, Switzerland) are small molecules, which inhibited the aggregation of A to oligomeric and fibrillar species for the potential treatment of AD and glaucoma [1004]. Two 3-aminopyrazole moieties carrying additional aryl substituents were connected via a linker. The concept of 3-aminopyrazoles with a donor-acceptor-donor hydrogen bond pattern complimentary to that of the -sheet of A42 was first investigated by Schrader and colleagues [1005–1013] (Thomson Reuters Pharma, update of September 24, 2012). Beta-amyloid inhibitor (Icogenex, Seattle, WA) is a small molecule therapeutic that normalizes the production of A (Thomson Reuters Pharma, update of June 26, 2012). The structure was not disclosed. Beta-amyloid inhibitor (Star Scientific, Glen Allen VI through its subsidiary Rock Creek Pharmaceuticals in collaboration with the Roskamp Institute) reduced A when applied to cells (Thomson Reuters Pharma, update of January 3, 2012). The structure was not disclosed. Beta-amyloid modulators (Crossbeta Biosciences, Utrecht, The Netherlands) are small molecules, which target A oligomers and misfolded proteins (Thomson Reuters Pharma, update of August 24, 2012). Structures were not communicated. Beta-amyloid precursor protein modulators (Alzcor Pharmaceuticals, Arlington, MA) are investigated for the potential treatment of AD (Thomson Reuters Pharma, update of July 27, 2011). Structures were not disclosed. BMS-869780 (Bristol-Myers Squibb) (Fig. 12) reduced A42 levels in transgenic mice at oral doses and displayed an IC50 value at A42 in the low nM range. It was not hepatotoxic (Thomson Reuters Pharma, update of August 6, 2012). BTA-EG4 (Johns Hopkins University and Georgetown University) is a benzothiazole aniline derivative and amyloid- synthesis inhibitor for the potential treatment of Alzheimer’s disease (Thomson Reuters Pharma, update of October 30, 2012). The structure was not communicated. C36 (Medisyn Technologies, Minnetonka, MN in collaboration with the Mount Sinai School of Medicine) is an A40 and A42 lowering small molecule for the potential treatment of AD (Thomson Reuters Pharma, update of December 6, 2011). The structure was not disclosed. Caprospinol (SP-233; Samaritan Pharmaceuticals, Las Vegas, NV under license from Georgetown University) (Fig. 12), a spirostenol drug, blocked the oligomerization of A42 exerting a direct effect on mitochondria [1014–1017] (Thomson Reuters Pharma, update of March 18, 2011). Carvedilol is a launched unselective ␣1 , 1 and 2 blocker, which inhibited A fibril formation [1018]. CLR01 (University of Duisburg-Essen, Rensselaer Polytechnic Institute, NY and UCLA) (Fig. 12) is a molecular tweezer, which binds to lysine residues with micromolar affinity and interferes with a combination of hydrophobic and electrostatic interactions that are important in the self-assembly of most amyloidogenic proteins including A, tau, and ␣-synuclein [1007, 1018–1024]. CLR-097(Clera Inc., Toronto) inhibited the process of A plaque formation (Thomson Reuters Pharma, update of December 5, 2011). The structure was not disclosed. Cotinine, a natural metabolite of nicotine, may be a potential new therapeutic agent against AD [1025]. It reduced amyloid- aggregation and improved memory in Alzheimer’s disease mice [1725]. Daunomycin inhibited A fibril formation [1018]. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes DBT-1339 (Medifron, Seoul and licensee Daewoong, South Korea) (Fig. 12) is the lead compound from A protein deposition inhibitors (Thomson Reuters Pharma, update of June 1, 2012). Enoxaparin (Enox, a low molecular weight heparin) lowered brain A load in a mouse model of AD [1026]. It also improved cognition in APPSWE /PS1dE9 mice [1027, 1028]. GAG/carbohydrate compounds (ProteoTech, Kirkland, WA) are glycosaminoglycan modulators for the potential treatment of AD (Thomson Reuters Pharma, update of June 7, 2012). Structures were not disclosed. Galantamine inhibited A aggregation and cytotoxicity [1029]. haw-AD-14 (Hawthorn, Madison, Mississippi under license from CoPlex) is an A synthesis and tau phosphorylation inhibitor (Thomson Reuters Pharma, update of September 26, 2011). The structure was not communicated. HO-4160 (University of California at Davis) (Fig. 12) is a spin-labeled fluorene compound that specifically disrupted A oligomers [1030]. Imipramine in part through inhibition of TNF-␣ prevented cognitive decline and A accumulation in a mouse model of AD [1031]. IPS-04001, IPS-04001 and IPS-04003 (InnoPharmaScreen, South Korea) is a specific inhibitor of A peptide and VEGF-165 interaction. By June 2012, in vivo efficacy studies had shown a significantly enhanced effect on the memory of NSE-PS2/N141Ltransgenic mice (Thomson Reuters Pharma, update of July 16, 2012). The structure were not communicated. KMS-88 series (Hanmi Pharmaceutical, South Korea, the Korea Institute of Science and Technology and Seoul National University) (Fig. 12) is a series of aminostyryl-benzofuran derivatives inhibiting A fibril formation [1032, 1033] (Thomson Reuters Pharma, update of September 12, 2012). Minocycline is a second generation tetracycline that can effectively cross the blood-brain barrier. It improved cognitive impairment in AD models [1034]. It provided protection against A25-35 induced alterations of the somatostatin signaling pathway [1035, 1036]. It reduced microglial activation [1037, 1038]) and A derived neuroinflammation [1039]. It recovered MTT-formazan exocytosis impaired by A [1040]. It reduced the development of abnormal tau species in models of AD [1041–1043]. Minocycline protected PC12 cells against NMDA-induced injury via inhibiting 5-lipoxygenase activation [1044, 1045]. 35 Minocycline improved negative symptoms in patients with early schizophrenia [1046]. See also Part 2, Chapter 2.25. Drugs interacting with 5-Lipoxygenase. NPT-4003 (Neuropore Therapies, San Diego CA) is the lead of heterocyclic compounds that interfere with amyloid- aggregates. Detailed data were presented at the ICAD Vancouver in July 2012 [1726]. (Thomson Reuters Pharma, update of July 16, 2012). The structure was not communicated. Rifampicin inhibited A aggregation and neurotoxicity [1047–1049]. In a clinical trial in 101 mild-to-moderate AD patients, statistically significant improvements in the ADAScog scores after treatment with 300 mg rifampicin for three months were found [988]. Rifampicin and caffeine caused an upregulation of LRP1 [1050]. The brain efflux index of A in rifampicin and caffeine treated mice was significantly higher (82% and 80%, respectively) than the brain efflux index of control mice (62%). It appears that a yet to be identified transporter/receptor plays a significant role in A clearance, which is upregulated by rifampicin and caffeine. Rolitetracycline is effective as inhibitor of A fibril formation [1018]. SD-1002 (Synaptic Dynamics, Farmington CT) is a lysosomal modulator that reduced protein deposition of amyloid-1-42 oligomers for the potential treatment of Alzheimer’s disease (Thomson Reuters Pharma, updates of October 22, 2012). The structure was not communicated. SEN-1500 (Senexis, Cambridge, UK) (Fig. 12) and follow-up compound SEN-1576 (structure not disclosed) are small molecule A aggregation inhibitors [1727] (Thomson Reuters Pharma, updates of March 23 and August 2, 2012, respectively). Small molecule A modulators (Asceneuron, a spun-out of Merck Serono, Geneva, Switzerland) are drugs for the potential treatment of AD (Thomson Reuters Pharma, update of October 3, 2012). Structures were not disclosed. SP-08 (Samaritan Pharmaceuticals, Las Vegas, NV under 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 from Takeda) 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 activity in vitro [1051] and protected Caenorhabditis elegans from A-induced toxicity by targeting oligomers [1052]. 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; INOX1; OXIGON, ViroPharma, Exton PA, under license from Intellect Neurosciences under license from New York University and Mindset BioPharmaceuticals; Thomson Reuters Pharma, update of August 10, 2012), see Chapter 11. Antioxidants. AGT-160 (ArmaGen Technologies, Santa Monica, CA) is a recombinant IgG fusion protein formed by the fusion of a single chain Fv antibody against A plaque formation to the company’s human insulin receptor-targeting monoclonal antibody Trojan horse for transport across the blood-brain barrier for the potential detection and treatment of AD (Thomson Reuters Pharma, update of January 23, 2012). See also Part 1. Chapter 1.16. Insulin receptors. EDN-OL1 (Edunn Biotechnology, St. Louis, Missouri under license from St. Louis University) is an antisense oligonucleotide targeting A production, which improved memory and learning in three different AD mouse models. By February 2012, EDN-OL1 had demonstrated activity in patients with Down syndrome (Thomson Reuters Pharma, update of August 24, 2012). NPT-001 (NeuroPhage Pharmaceuticals, Cambridge, MA) is a filamentous M13 bacteriophage that disrupted plaque aggregation through binding of A. Direct binding of M13 bacteriophage to fibrils of aggregated A was observed with high affinity of 4 nM. In an AD mouse model, A plaques were reduced by 70% after a single intracranial administration (Thomson Reuters Pharma, update of July 16, 2012). Spheron-based therapeutics (Nymox Pharmaceutical Corp., Quebec) are compounds capable of blocking the transformation of human spherons into plaques [1053, 1054] (Thomson Reuters Pharma, update of February 22, 2012). A-aggregation inhibitor research programs were ongoing at Elan [1055], at GrenPharma [1056], at GSK [1057, 1058], at Johnson & Johnson [1059], at Pfizer [1060], at Pharmacia-Farmitalia Carlo Erba [1061] and at Scios [1062]. Excellent papers on A aggregation inhibitors from universities 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 on A-aggregation inhibitors were published (in chronological 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, NC531, Alzhemed, Cerebril; Bellus Health, former Neurochem), an orally available glycosaminoglycan mimetic, was terminated in 2007 [1145–1151]. The compound was launched as a nutraceutical for memory protection [1152]. Also the development of ALS-499 and ALS-633 (Advanced Life Sciences, in collaboration with Argonne National Laboratory), amyloid beta aggregation inhibitor (Zyentia), amyloid oligomer specific antagonists (Treventis), amyloid protein deposition inhibitor (ProteoTech), AN-531 (Athena Neurosciences, now Elan Pharmaceuticals), AS-602704 (Ac-FPFFD-NH2 , Merck Serono [1153]), beta amyloid aggregation inhibitors (Cortex, Elan, Hybridon, Oceanix, Queens University at Kingston and University of Wisconsin-Madison), beta-amyloid modulators (Inflame Therapeutics, Neuro-Hitech and Q-RNA), beta-amyloid precursor protein modulators (SIBIA Neurosciences in collaboration with BMS), beta amyloid synthesis inhibitors (BTG), CLR-01 (Clear Therapeutics, a spin-off from UCLA), CT-500 (Creabilis Therapeutics), ID-1135 and ID-1567 (Bellus Health), lysosomal modulators (Synaptic dynamics), MPI-442690 and MPI-442691 (Myriad Genetics in collaboration with the Mayo Clinic), NC-503 and NC-531 (Neurochem; Bellus Health), NHT-0112 (Neuro-Hitech), NOX-A42 (Noxxon Pharma), NVP-AAM147 and NVP-AAM155 (Novartis), phenserine-based amyloid inhibitors (Message Pharmaceuticals), PPI-368, PPI-558 and PPI-1019 (Apan; all Praecis Pharmaceuticals [953, 957]), PTI-777 (ProteoTech), Reumacon (Meda under license from Conpharm), Ro-47-1816-001, Ro-65-7652-000, Ro-65-8564-001 and Ro-8815-001 (Roche), RS-1178 (BGC-20-1178; Senexis under license from BTG under license from Sankyo [1154]), RS-0406 (BGC-20-0406; Senexis under license from BTG under license from Sankyo [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, formerly Cytokine Networks), a cytokine inhibitor, which inhibited A production, plaque formation and cognitive deterioration in an animal model of AD [39, 40, 1729]), SEN-304, SEN-606, SEN-1186, SEN-1203, W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes and SEN-1329 (Senexis), SIB-1848 (SIBIA Neurosciences, now Merck and Bristol-Myers Squibb), SKF-746532 (SmithKline Beecham, now GSK), synthetic anti-beta sheet peptides (Mayo Foundation), THUR-24 (Thuris Corp.), transthyretin (Research Foundation of the State University of New York), and of VIP-SSM (University of Illinois) was terminated. Ligands interacting with amyloid-β Since the discovery of the 11 C-PiB-PET ligand, tremendous progress has been made in the development of new PET ligands. Considerable evidence suggests that A deposition precedes decline in cognition [1159, 1160]. The development of PET amyloid- imaging agents was described [1730]. 11 C-PiB (University Pittsburgh) (Fig. 13) had a KD = 1.4 nM for AD frontal cortex and KD = 4.7 nM for A fibrils and did not bind to neurofibrillary tangles [1161]. The log P value is 1.2 [1162]. Patients with AD typically showed a marked 11 C-PiB retention (with a significant 1.5 to 2-fold increase) in cortical areas known to contain large amounts of A plaques [1163–1165]. 11 C-PiB positivity in MCI indicated already advanced A pathology [1166]. Significant correlations between 11 C-PiB retention and CSF biomarkers were found in MCI patients [1167]. Clinical severity of AD measured by the Clinical Dementia Rating scale sum of boxes correlated with 11 C-PiB uptake in PET [1168]. 11 C-PiB imaging of human immunodeficiency virus-associated neurocognitive disorder was described [1169]. For a direct comparison of 11 C-PiB, 18 F-Florbetapir, 18 FFlorbetaben, and 18 F-Flutemetamol, see [1170] for a comparison between Pittsburgh Compound B and Florbetapir see [1731]. Longitudinal imaging of AD pathology with 11 C-PiB, 18 F-FDDNP, and 18 F-FDG was described [1171–1173]. For the prediction of cognitive decline via PET of brain A and tau, see [1174]. The clinical perspective of imaging of cerebral A plaques was discussed [1175, 1205]. The correspondence between in vivo 11 C-PiB-PET and postmortem assessment of plaques was discussed [1176]. Imaging brain amyloid in nondemented adults with Down syndrome using Pittsburgh Compound B was reported [1732]. Fred Van Leuven and coworkers carried out a systematic evaluation of the isomeric compounds with the OH groups attached in 4, 5, and 7 positions. The 5-hydroxy analogue had the most favorable in vivo pharmacokinetics in brain [1177, 1178]. 18 F-Florbetapir 37 (18 F-AV-45; Amyvid; Avid Radiopharmaceuticals, Philadelphia, PA, a subsidiary of Eli Lilly, under license from the University of Pennsylvania) (Fig. 13) is a launched 18 F PET imaging agent targeting A. It had a Ki of 2.87 nM, did not bind to tau, and can easily be labeled with 18 F by an automated synthesis [1179–1187]. By March 2010, more than 700 patients were enrolled in a Phase III trial presumed to be the 18 F-AV-45-A07 study. Several clinical papers appeared describing imaging with 18 F-Florbetapir [1188–1195]. Correlations between 18 F-Florpetapir and FDG images were reported [1196]. 18 F-Florpetapir is useful to quantify brain amyloid load [1197]. For a direct comparison of 11 C-PiB, 18 F-Florbetapir, 18 FFlorbetaben, and 18 F-Flutemetamol, see [1170]; for a clinical comparison of 18 F-Florbetapir and 18 F-FDG PET in patients with AD and controls, see [1198]. A prospective cohort study was described [1199]. A longitudinal assessment of 18-month of cognitive decline in mild cognitive impairment and Alzheimer’s disease patients using florbetapir was communicated [1733]. “From Alois to Amyvid” see [1734]. PET imaging was also carried out in a murine model of amyloid- plaque deposition [1735]. FDA approved 18 F-Florpetapir as a PET imaging agent to estimate A neuritic plaque density in patients with cognitive impairment on April 6, 2012. Lilly launched the imaging agent in June 2012. The compound will be manufactured and distributed by Cardinal Health [1200] (Thomson Reuters Pharma, update of September 17, 2012). 18 F-Florbetaben (18 F-BAY 94-9172; 18 F-AV1/ZK; Piramal Healthcare, Mumbai, India from an asset acquisition from Bayer under license from Avid Pharmaceuticals and the University of Pennsylvania) (Fig. 13) is the phenyl analogue of florbetapir. It has a Ki of 2.22 nM [1181]. In November 2009, a Phase III clinical trial in AD patients (n = 292) began to assess safety and efficacy of florbetaben to detect or exclude cerebral A. Data from clinical studies were reported [1201–1208]. An in vitro characterization was disclosed [1736]. The one-step radiosynthesis was described [1209], as was the radiation dosimetry [1210]. In the NCT01138111 trial, 18 F-Florbetaben PET imaging was studied in MCI patients. For a direct comparison of 11 C-PiB, 18 F-Florbetapir, 18 FFlorbetaben, and 18 F-Flutemetamol, see [1170]; for a comparison of 11 C-PiB and 18 F-Florbetaben, see [1211]; for the impact of 18 F-Florbetaben PET imaging on confidence in early diagnosis of AD, see [1212] (Thomson Reuters Pharma, update of September 6, 2012). 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-. 18 F-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 a Phase I study were reported [1213], as were results of a Phase II study [1214, 1215]. In December 2009, a Phase III trial sponsored by GE Healthcare was initiated in the US. By April 2010, Phase III trials were also underway in Europe. In April 2012, preliminary data from the two Phase III studies in terminally ill patients and young healthy subjects were reported. Data showed that the primary endpoint was met. Patients (who underwent brain autopsy) showed concordance between 18 F-Flutemetamol PET images and AD-associated A brain pathology and healthy subjects showed concordance with the known lack of brain A. The association between in vivo 18 F-Flutemetamol PET imaging and in vivo cerebral cortical histopathology was described [1216], as was a combination of biomarkers PET 18 F-Flutemetamol and MRI [1217]. For a direct comparison of 11 C-PiB, 18 F-Florbetapir, 18 F-Florbetaben, and 18 F-Flutemetamol, see [1170]. Binary classification of 18 F-Flutemetamol PET using machine learning was disclosed [1218]. The pharmacokinetics of 18 F-Flutemetamol in wild-type rodents were reported [1219] (Thomson Reuters Pharma, update of October 3, 2012). 18 F-AZD-4694 (Navidea Biopharmaceuticals, Dublin, Ohio, formerly Neoprobe Corporation under license from AstraZeneca) is an i.v. PET ligand for the potential imaging of A depositions in AD in Phase II development [1220]. In December 2011, a Phase III program was planned to start in early 2013 (Thomson Reuters Pharma, update of September 21, 2012). The structure was not communicated. For syntheses of additional 18 F-ligands of AstraZeneca see [1737]. BAY-1006578 (Bayer) is in Phase I clinical trials in Finland and Sweden since June 2010 (n = 36). By October 2011, the trial was completed (Thomson Reuters Pharma, update of April 30, 2012). The structure was not communicated. A potential follow up compound is BAY-1008472 [1221]. 123 I-MNI-168 (Institute for Neurodegenerative Disorders, IND, New Haven, CT) is a SPECT ligand to detect A deposition in patients with AD. A Phase I clinical trial was initiated in the US in January 2009 (n = 34). In February 2011, the trial was terminated (Thomson Reuters Pharma, update of February 10, 2012). The structure was not communicated. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 18 F-MNI-558 (Molecular NeuroImaging in collaboration with the Institute for Neuro-degenerative Disorders, IND, New Haven, CT) is a PET agent that binds A. The compound was in a Phase 0 trial in AD patients and in volunteers (n = 10) from October 2010 to July 2011 (Thomson Reuters Pharma, update of April 30, 2012). The structure was not communicated. 11 C-BF-227 (Fig. 14) was used for amyloid PET imaging [1222–1225], for PET imaging of ␣-synuclein deposition [1226, 1227], and for in vivo detection of prion amyloid plaques [1228]. 123 I-DRM-106 (Fujifilm Pharma) (Fig. 14) is a radioiodinated imidazopyridine derivative for the potential use as SPECT imaging agent for A for the diagnosis of AD (Thomson Reuters Pharma, update of September 21, 2011). 18 F-Hexethal (Pfizer, under license from the University of Aberdeen), a barbiturate derivative, is an A imaging agent (Thomson Reuters Pharma, update of September 24, 2012). 11 C-SB-13 (Fig. 14) was evaluated first in postmortem brain tissues [1229] and in AD patients [1230]. 18 F-Florbetapir dimer (Avid Radiopharmaceuticals, Philadelphia, PA, Eli Lilly) is a PET imaging agent for the potential diagnosis of cerebral amyloid angiopathy [1231] (Thomson Reuters Pharma, update of May 8, 2012). Dicyanovinylnaphtalenes were described for neuroimaging of A [1232]. Also 18 F-labeled benzoxazoles [1233] and 18 F-labeled 2-pyridinylbenzoxazoles and 2-pyridinylbenzothiazoles were presented for PET imaging of A plaques [1234]. The development of 11 C-AZD-2184 and 11 CAZD-2995 (both AstraZeneca [1235–1237, 1738]) and 18 F-FDDNP (Siemens Medical Solutions Molecular Imaging and UCLA) (Fig. 14) was terminated. 18 F-FDDNP was the first PET tracer used in vivo for detection of cerebral A plaques in 2002 [1238]. 18 FFDDNP binds to A and neurofibrillary tangles in AD, to prion plaques in Creutzfeldt-Jakob disease, to A deposits in cerebral amyloid angiopathy, and to Lewy bodies in PD and DLB [1161]. It was used for measuring amyloid- and tau levels in adults with Down’s syndrome [1739]. The ligand is still used for prediction of cognitive decline [1174, 1740]. Longitudinal imaging of AD pathology using 11 C-PiB, 18 F-FDDNP, and 18 F-FDG PET was described [1239]. 18 F-FDDNP PET allowed a prediction of cognitive decline based on hemispheric cortical surface maps [1240]. 123 I-IMPY (Avid Radiopharmaceuticals, Philadelphia, PA, under license from University of Pennsylvania, Fig. 14) is a SPECT ligand for imaging studies 39 [1241, 1242]. Its Ki is 15 nM [1243]. The safety and biodistribution was evaluated [1244]. The signal-tonoise ratio for plaque labeling is not as robust as that of 11 C-PiB. 11 C-PiB showed a S/N ratio of about 2.5, while 123 I-IMPY displayed a ratio of 1.8–2.0, between 30 and 50 min after an i.v. injection [1181]. Its development was terminated. The development of amyloid binding PET ligands (Aventis), fluoropegylated indolyl-phenylacetylenes (Avid Radio-pharmaceuticals), 11 C-6-MeBTA (University of Pittsburgh), and of 18 F-SMIBRW372 (Siemens Medical Solutions Molecular Imaging) was terminated. Inhibitor of serum amyloid P component binding The normal plasma protein serum amyloid P component (SAP) binds to fibrils in all types of A deposits and contributes to the pathogenesis of amyloidosis [1245–1247]. Ro-63-8695 (CPHPC; Pentraxin Therapeutics, London under license from Roche) (Fig. 14) is a competitive inhibitor of serum amyloid P component binding to amyloid fibrils (IC50 = 0.9 M). First results from in vivo mouse and human studies have been communicated [1245, 1248–1250]. In addition, Pentraxin Therapeutics and GSK are investigating a combination of CPHPC with a humanized antibody for the potential treatment of amyloidosis (Thomson Reuters Pharma, update of March 22, 2012). Vaccines against amyloid-β AN-1792 (AIP-001; Elan and American Home Products, later Wyeth, now Pfizer) was the first vaccine against AD in clinical trials. The antigen was synthetic A42 , which was administered with QS-21 from the Stimulon family of saponins purified from the bark of Quillaja saponaria as adjuvant [1251, 1252]. The multicenter double-blind Phase IIa study in the US and Europe involving 375 patients with mild-tomoderate AD started in the third quarter of 2001. Treatment of patients with AN-1792 plus adjuvant and the adjuvant alone as placebo was in a ratio of 4 : 1. 18 patients developed meningoencephalitis probably due to an inappropriate T cell activation and/or the proinflammatory Th1 type of adjuvant. The study was discontinued in March 2002 [1253]. Nevertheless, many important data could be collected. From 129 patients immunized with the drug, 25 were classified as antibody responders after 4.5 years. These patients showed significantly slower decline on the 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 significant difference on a dependence scale compared to placebotreated patients. Antibody responders also showed less of a decline in a memory test. The scientific harvest of this single clinical trial is listed in chronological order [1254–1286] (Thomson Reuters Pharma, update of August 24, 2012). Affitope AD-02 (Affiris, Vienna AT and licensee GlaxoSmithKline Biologicals) is a six amino acid peptide vaccine targeting the N-terminus of A only, when it is free. The adjuvant is aluminum hydroxide. The antibody response is focused exclusively on A and did not show crossreactivity to APP [1287, 1288]. A European Phase II clinical trial in 420 patients in Austria, Germany, France, the Czech Republic, Slovakia, and Croatia started in April 2010 (Thomson Reuters Pharma, update of August 1, 2012). CAD-106 (Cytos Biotechnology, Zurich and Novartis) is an A1-6 peptide linked to a Q virus-like particle 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 October 2008 (n = 30). The safety, tolerability, and antibody response of active A immunotherapy with CAD106 in patients with AD was published [1289]. For comments, see [1290, 1291]. It was found that 80% of the patients involved in the trial developed their own protective antibodies against A without suffering any side-effects over the three years of the study. The researchers believe that the CAD106 vaccine is a tolerable treatment for patients with mild-to-moderate AD. 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 were presented [1294] (Thomson Reuters Pharma, update of July 26, 2012). Vanutide cridificar (ACC-001, PF-05236806; Janssen Alzheimer Immunotherapy, Dublin, Ireland acquiring Elan’s Alzheimer’s Immunotherapy Program and Pfizer) is a peptide fragment of A conjugated to the mutated diphtheria toxin protein CRM197. The adjuvant is QS-21. In May 2007, a randomized, multicenter, double-blind, placebo-controlled, parallel assignment, safety/tolerability/immunogenicity Phase II trial in patients with mild-to-moderate AD (n = 56) began [1295]. In July 2009, Pfizer began a multicenter, 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 was September 2014. In August 2009, an additional study W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes was initiated in Japanese subjects (n = 32) [1276] (Thomson Reuters Pharma, update of August 13, 2012). ACI-24 (AC Immune, Lausanne, Switzerland) is an A1-15 peptide to which on both ends two lysines were attached, which are tetrapalmitoylated on the -nitrogens. The antigen is embedded in a liposome membrane. Adjuvant is a mixture of the lipids DMPC, DMPG, cholesterol, and MPLA in a ratio of 9 : 1 : 7 : 0.06, respectively [1296, 1297]. The drug restored memory defects of transgenic AD mice. The IgF subclasses of the antibodies generated from the vaccine were mostly IgG2b indicating a noninflammatory Th2 isotype. CD and NMR revealed predominantly -sheet conformation. The drug is currently evaluated in a Phase I/II clinical trial in Denmark, Finland, and Sweden (Thomson Reuters Pharma, update of June 19, 2012). Affitope-AD-03 (Affiris, Vienna, Austria and licensee GlaxoSmithKline Biologicals) entered a Phase I clinical trial in October 2010. In November 2011, the study was completed (Thomson Reuters Pharma, update of April 4, 2012). UB-311 (United Biomedical, Hauppauge, NY) is an intramuscularly administered vaccine targeting Nterminal amino acids 1-14 of A in Phase I clinical trials in Taiwan in patients with mild-to-moderate AD (n = 18) [1298] (Thomson Reuters Pharma, update of June 25, 2012). V-950 (Merck, Whitehouse Station, NJ) is an N-terminal A peptide conjugated to an aluminumcontaining adjuvant with or without ISCOMATRIX (an adjuvant containing saponin, cholesterol, and phospholipids) [1299]. Mild-to-moderate AD patients (n = 124) received six injections/year of increasing doses of either placebo or V-950 in the presence or absence of varying concentrations of ISCOMATRIX in Phase I clinical trials in the US and Sweden over a four year period [1276] (Thomson Reuters Pharma, update of February 24, 2012). There are numerous vaccine preparations in preclinical evaluation (in alphabetical order): ABvac40 and ABvac42 (Araclon Biotech, Zaragoza, Spain) are active therapeutic antibody vaccines awaiting approval for clinical trials [1741] (Thomson Reuters Pharma, update of June 25, 2012). ADepVac (Ichor Medical Systems, San Diego, CA in collaboration with the University of California at Irvine and the Institute for Molecular Medicine) is a DNA vaccine using the TriGrid electroporation technology [1300] (Thomson Reuters Pharma, update of July 13, 2012). 41 ALZ-101 (Alzinova, Goteborg, Sweden, a spin-off of MIVAC Development) is a specific oligomerdirected therapeutic vaccine (Thomson Reuters Pharma, update of July 18, 2012). ALZ-301 (Alzinova, Goteborg, Sweden, a spin-off of MIVAC Development) is an A40 oligomer targeted replacement therapy (Thomson Reuters Pharma, update of July 18, 2012). Alzheimer’s disease vaccine (VLP Biotech, San Diego CA) is a vaccine that activates antibodies against A protein (Thomson Reuters Pharma, update of May 3, 2012). Amyloid- 3-10 DNA vaccination (China Medical University, Shenyang) suggested a potential new treatment for AD [1301–1305]. Active immunization with Ankyrin G, a neuronal cytoskeletal protein, with Freund’s adjuvant complete of arcA mice reduced A pathology [1306]. BAN-2203 (BioArctic Neuroscience, Stockholm, Sweden) is an immunotherapeutic vaccine targeting A protofibrils (Thomson Reuters Pharma, update of January 7, 2011). BBS-1 BACE inhibitor mAb vaccine (Nasvax, Ness Ziona, IL under license from Ramot at Tel Aviv University) is based on a lead mAb candidate blocking -site-1, which inhibited the ability of BACE to cleave APP (Thomson Reuters Pharma, update of July 30, 2012). C12 (Pharma Bio, Moscow, Russia) is a program of therapeutic vaccines comprised of synthetic peptide fragments of the ␣7 nicotinic acetylcholine receptor used to generate antibodies that block A binding to neurons (Thomson Reuters Pharma, update of March 29, 2012). EB-101 (Atlas Pharmaceuticals, Sunnyvale, CA) is a synthetic human A42 /sphingosine-1-phosphate/ liposome vaccine (Thomson Reuters Pharma, update of July 27, 2012). A T cell-based vaccination with Glatimer acetate of AD double-transgenic (APP/PS1) mice resulted in decreased plaque formation and induction of neurogenesis. Dendritic-like (CD11c) cells produced insulin-like growth factor 1 [1307]. MER-5101 (Mercia Pharma, Scarsdale, NY) is a vaccine comprised of an A peptide conjugate coupled to an immunogenic carrier protein and the company’s Th2-biased adjuvant MAS-1 (Thomson Reuters Pharma, update of January 27, 2012). Mimovax (MV-01; Affiris, Vienna AT) is an AFFITOPE-based vaccine targeting truncated and modified forms of A (Thomson Reuters Pharma, update of April 2, 2012). 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 license from Vitruvian BioMedical under license from the University of Texas Southwestern Medical Center) is an adjuvant free, gene-base (DNA) A42 vaccine for the potential treatment or prevention of AD (Thomson Reuters 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 potential intranasal treatment of AD (Thomson Reuters Pharma, update of May 2, 2012). RV-03 (Intellect Neurosciences, New York, NY) is a peptide vaccine developed using the RECALL-VAX technology targeting both A and a truncated delta tau protein (Thomson Reuters Pharma, update of August 21, 2012). SeV-amyloid beta RNA vaccine (DNAVEC, Tsukuba, Japan) is an intranasal vaccine comprising a Sendai virus vector encoding the A gene [1308] (Thomson Reuters Pharma, update of September 21, 2011). Excellent papers on A vaccines from universities were presented (in chronological order) 2000: [1309, 1310]; 2001: [1311]; 2004: A1-42 gene vaccination [1312]; 2005: a synthetic universal Th cell pan HLA DR epitope, pan HLA DR-binding peptide (PADRE), in which the PADRE-A1-15 sequence lacks the T cell epitope of A [1313]; 2006: an A derivative in alum adjuvant [1314], an intranasal dendrimeric A1-15 vaccine [1315–1319]; 2007: an oral vaccination using a recombinant adeno-associated viral vector carrying A cDNA (AAV/A) [1320], a transcutaneous administration of aggregated A1-42 plus the adjuvant cholera toxin [1321], a mannan-A28 conjugate [1322, 1323]; 2008: a DNA epitope vaccine that expresses epitopes of A42 [1324]; 2009: K6 A1-30-NH2 in alum adjuvant [1325], an A1-33-MAP peptide to markedly reduce intracellular A deposits [1326], K6A1-30 for immunizations of old primates [1327], A1-42 vaccinations also reducing mouse tau pathology [1328]; 2010: an oral vaccination with GFP-A [1329], immunization of aged beagle dogs with aggregated A1-42 formulated in an alum adjuvant [1330], active immunization with A1-42 emulsified in CFA containing Mycobacterium tuberulosis extract [1331], lowering of A plaque burden by the SDPM1 peptide, a 20 amino acid peptide bound by cysteines that binds tetramer forms of A1-40 and A1-42 amyloids [1332]; 2011: two novel anti-A vaccines consisting of virus like particles [1333], the identification of the shortest A-peptide generating specific antibodies [1334]; 2012: preventive immunization of aged primates [1335]. Excellent reviews on A vaccines have been provided (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 immunotherapeutic approaches in AD targeting A was presented [1375, 1376]. The development of A retroparticles (University of Heidelberg and Novartis [1377]), Abloid (Virionics), ACC-002 (Elan), AFFITOPE AD-01 (Affiris), Alzheimer’s vaccine (Lundbeck under license from Pharmexa), Alzheimer’s vaccine (Mindset BioPharmaceuticals), Alzheimer’s disease vaccines (University of South Florida and University of New Mexico), anti-amyloid- vaccines (Wyeth, now Pfizer and Janssen Alzheimer Immunotherapy), DNA vaccine (University of Texas, Southwestern Medical Center), immunotherapy vaccine (Prana Biotechnology), KNX-Vaccine3A (Kinexis under license from the University of California at Irvine), PEVIPRO (Pevion Biotech [1378]), PX-106 (Lundbeck/Pharmexa), and RV-01 (Intellect Neurosciences) was terminated. Antibodies against amyloid-β Bapineuzumab (AAB-001; Janssen Alzheimer Immunotherapy, Dublin, Ireland following the acquisition of Elan’s Alzheimer’s Immunotherapy Program and Pfizer) is a humanized monoclonal antibody raised against eight amino acids in the N-terminus of A for the potential i.v. and s.c. treatment of AD. A Phase III clinical study started in December 2007. The Phase III program included two trials in ApoE4-positive patients (n = 800 in the US and EU) and two in ApoE4-negative patients (n = 1,250 in the US and EU) for 18 months each and three extension studies for both ApoE4 and non-ApoE4 arms with three doses, 0.5, 1, and 2 mg/kg. The highest dose had to be abandoned due to vasogenic edema safety concerns. One extension study started in July 2009 (n = 1,350), two additional extension studies in December 2009 in the EU and Australia (n = 800 and 1,000, respectively). Clinical results from the Phase I and Phase II studies were reported [1276, 1361, 1379–1382] as was a 11 C-PiB PET assessment [1383] and the effects on CSF biomarkers [1384]. Reviews on bapineuzumab were published (in chronological order) [23, 1385–1392]. For the scientific basis see the Nature W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Medicine paper [1393]. The pharmacokinetic profile of 125 I-labeled 3D6, the mouse parent antibody of bapineuzumab, in transgenic mice was reported [1742]. In July 2012, Pfizer reported that the ’302 study in ApoE4 carriers did not meet its primary endpoints. Pfizer, Johnson & Johnson, and Elan announced that development of i.v. bapineuzumab was terminated (Thomson Reuters Pharma, update of September 18, 2012). Solanezumab (LY-20622430; Lilly) is a middomain humanized monoclonal antibody selective for soluble A. Three Phase III studies were initiated in patients with mild-to-moderate AD (n = 1,000) in May 2009, patients received 400 mg of solanezumab or placebo i.v. every 4 weeks for 80 weeks. First results were disclosed (in chronological order) [1276, 1394–1398]. Safety and biomarker effects of solanezumab in patients with AD was communicated [1399]. Solanezumab failed to meet the cognitive and functional primary endpoints of two Phase III AD trials. But it showed a significant reduction in cognitive decline in patients with mild AD. The open-label EXPEDITION-EXT extension trial, which is fully enrolled, will continue as planned (Thomson Reuters Pharma, update of September 7, 2012). Gantenerumab (R-1450; Roche under license from MorphoSys) is a human anti-A monoclonal antibody. 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 administered by s.c. injection every 4 weeks for 104 weeks to evaluate effects on cognition and functioning, safety, and pharmacokinetics. In June 2012, the trial was expanded to a Phase II/III trial. The trial size will be increased from 360 to 770 participants. The mechanism of A removal was described [1400, 1401]. An expert opinion was provided [1402] (Thomson Reuters Pharma, update of September 5, 2012). Crenezumab (MABT-5102A; RG-7412; Genentech under license from AC Immune, Lausanne, Switzerland) is an anti-A humanized monoclonal antibody as a conformation-specific, passive immunotherapy for the potential i.v or s.c. treatment of AD. A Phase II clinical trial (n = 372; NCT01397578) was initiated in April 2011. The first Alzheimer’s Prevention Initiative will study the effects of crenezumab in 300 people from a large family in Columbia with a rare genetic mutation that typically triggers AD symptoms around age of 45. It is a collaboration between the National Institutes of Health (NIH), Banner’s Alzheimer’s Institute, and Genentech over five years starting in 2013. Data and findings will be shared 43 publicly after the study completion. The scientific basis for the selection of crenezumab was published recently [1403] (Thomson Reuters Pharma, update of September 5, 2012). GSK-933776A (GSK) is a monoclonal antibody for the i.v. treatment of AD and age-related macular degeneration. A Phase II trial (n = 162) of age-related macular degeneration started in the US in August 2011 (Thomson Reuters Pharma, update of August 24, 2012). The topic intravenous immunoglobulins as a treatment for AD, the rationale and current evidence was described in reviews [1404, 1405, 1743]. Immune globulin intravenous human (IVIg; launched as Gammagard in the US and as Kiovig in Europe; Baxter International, Deerfield, IL) is a highly purified solvent/detergent-treated, sterile, freeze-dried preparation derived from human plasma for the treatment of primary immunodeficiency disease. This pool of human immunoglobulins obtained from healthy donors contains naturally occurring anti-A antibodies. In a Phase II trial in 24 patients, the eight-person placebo group worsened, whereas the 16 treated patients improved moderately on both cognitive and quality-of-life measures over the first 6 months [1406–1408]. A Phase III clinical trial in AD patients enrolling 390 patients was initiated in September 2008. Trial completion is expected by the end of 2012 (NCT00818662, NCT00299988) [1276]. The first report of long-term (three-year) stabilization of AD symptoms with IVIG (Gammagard, Baxter), including no decline on measures of cognition, memory, daily functioning, and mood, was reported in July 2012 at the Alzheimer’s Association International Conference in Vancouver. Sales in 2011 amounted to 1,541 million USD. For preclinical data, see [1409, 1410] (Thomson Reuters Pharma, update of October 3, 2012). Octagam (Octapharma, Lachen, Switzerland) is a 10% liquid intravenous immunoglobulin launched for the treatment of primary immunodeficiency. By December 2008, a double-blind, randomized, Phase II clinical trial (NCT 00812565) in 58 patients with AD had been initiated in the US. Clinical data were presented at the AAIC in Paris in July 2011 (Thomson Reuters Pharma, update of April 3, 2012). Flebogamma DIF 10% (or 5%) (Grifols SA, Barcelona) is a 10% (or 5%) double inactivated and filtered immunoglobulin therapy for the treatment of primary immunodeficiency. Data were presented on a single-center, open-label, pilot study on the use of Flebogamma DIF 0.5 g/kg i.v., q2w for 6 months in AD 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 of March 23, 2012). BAN-2401 (Eisai under license from Bioarctic Neuroscience, Stockholm, Sweden) is a humanized monoclonal antibody that targets the large soluble amyloid product (protofibril A). A Phase I trial in 80 patients with mild-to-moderate AD began in September 2010. Eisai plans to co-administer BAN-2401 with donepezil 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 human IgG1 mAb targeted against A. A Phase I clinical trial (n = 40) in patients with mild-to-moderate AD started in the US in July 2011 (Thomson Reuters Pharma, update of September 24, 2012). PF-05236812 (AAB-003; Janssen Alzheimer Immunotherapy and Pfizer) is a humanized monoclonal antibody targeted against A. In October 2010, a randomized, double-blind, placebo-controlled, adaptive, Phase I trial was initiated in patients with mild-tomoderate AD (n = 80) in the US [1411] (Thomson Reuters Pharma, update of August 13, 2012). RN6G (Rinat Neuroscience Corp., New York, now Pfizer) is an anti-A antibody for the potential i.v. treatment of age-related macular degeneration in Phase I trial (n = 45) since April 2009 in the US. In April 2012, a randomized, double-blind, placebo-controlled, Phase II study was planned in patients with geographic atrophy secondary to age-related macular degeneration (expected n = 276) to assess the safety and efficacy of RN6G [1412] (Thomson Reuters Pharma, update of September 11, 2012). SAR-228810 (sanofi) is an anti-protofibrillar A monoclonal antibody. A randomized, double-blind, placebo-controlled, Phase I trial in patients with mildto-moderate AD (n = 48) was initiated in Sweden in January 2012. The study is scheduled to complete in December 2013 (Thomson Reuters Pharma, update of July 27, 2012). There are currently many antibodies for the potential treatment of AD and closely related diseases in preclinical evaluation (in alphabetical order): 4E10 (Prana Biotechnology, Parkville, Australia) is a monoclonal antibody targeting a proprietary pathological A target epitope (Thomson Reuters Pharma, update of November 2, 2011). 6F6 (GSK) is an anti-A monoclonal antibody for the potential treatment of age-related macular degeneration (Thomson Reuters Pharma, update of June 9, 2011). 9D5 (University of Göttingen, MBM ScienceBridge) targets pyro-Glu-A peptide oligomers for the potential diagnosis and treatment of AD [1413] (Thomson Reuters Pharma, update of August 7, 2011). A-887755 (Abbott Laboratories) is an A, oligomer-selective, mouse monoclonal antibody generated using a homogenous, synthetic A20-42 oligomer peptide (Thomson Reuters Pharma, update of February 27, 2012). A-992401 (Abbott Laboratories) is a mAb targeting the receptor for advanced glycation-end-products (Thomson Reuters Pharma, update of December 1, 2010). Ab40-4-42 (Ilsung Pharmaceutical, Seoul) is a biological therapeutic, which acted by inhibiting A aggregation and cytotoxicity (Thomson Reuters Pharma, update of May 14, 2012). ACU-193 (Acumen Pharmaceuticals, Livermore CA) is a monoclonal antibody against amyloid-derived diffusible ligands (Thomson Reuters Pharma, update of August 16, 2012). AD-0802 (Bioarctic Neuroscience, Stockholm, Sweden) is a humanized mAb with affinity for binding to A protofibrils. It is a potential follow-up compound to 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 the fusion of a single chain Fv antibody against A plaque formation to the company’s human insulin receptortargeting monoclonal antibody Trojan horse for the transport across the blood-brain barrier. A 1 mg/kg i.v. dose of AGT-160 was administered twice a week for 12 weeks to transgenic AD mice resulting in a 40% decrease in the brain concentration of A (Thomson Reuters Pharma, update of January 23, 2012). See also Part 1. Chapter 1.16. Drugs interacting with the insulin receptor. ALZ-201 (Alzinova, Goteborg, Sweden, a spinoff of MIVAC Development) is an oligomer-specific A monoclonal antibody (Thomson Reuters Pharma, update of July 20, 2012). Amyloid precursor protein C-terminal fragment-targeted monoclonal antibodies (Ecole Polytechnique Fédérale de Lausanne, EPFL) is a monoclonal antibody targeted to the 99 amino acid C-terminal fragment of APP (APP-C99) (Thomson Reuters Pharma, update of March 31, 2011). Anti-amyloid beta antibodies (Kyowa Hakko Kirin under license from Immunas Pharma, OncoTherapy Science Inc.) are antibodies targeting A W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes oligomers (Thomson Reuters Pharma, update of August 6, 2012). Brain-targeted BACE1 antibody (Genentech, Roche Holding) is a bi-specific antibody against BACE1 and the transferrin receptor to allow transcytosis 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) is a combination of CPHPC, which lowers blood levels of serum amyloid P component (see Chapter 16.2) and a humanized antibody for the treatment of amyloidosis [1249] (Thomson Reuters Pharma, update of March 22, 2012). DLX-212 (Delenex Therapeutics, Zurich, Switzerland) is an anti-A antibody fragment (Thomson Reuters 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 treatment of AD (Thomson Reuters Pharma, update of June 13, 2012). IN-N01 (Intellect Neurosciences under license from Immuno-Biological Laboratories, IBL, New York, NY) is an antibody drug conjugate consisting of a A-specific humanized mAb employing its ANTISENILIN technology, for the potential treatment of AD, glaucoma, age-related macular degeneration and traumatic brain injury. In September 2012, Intellect planned to conduct studies in combination with tau mAbs 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 conjugated to melatonin developed using its CONJUMABA technology for the potential treatment of age-related macular degeneration and Alzheimer’s disease (Thomson Reuters Pharma, update of October 2, 2012). Lpathomab (LT-3015; Lpath Inc., San Diego, CA) is a systemic formulation of a humanized antilysophosphatidic acid monoclonal antibody for the potential treatment of fibrosis, ocular inflammation, and CNS disorders including AD [1423] (Thomson Reuters Pharma, update of August 28, 2012). MDT-2007 (Medronic, Minneapolis, MN) is a humanized anti-A monoclonal antibody, the humanized version of the mouse mAb 6E10 (Thomson Reuters Pharma, update of January 6, 2012). Nanobody therapeutics (Ablynx, Gent Belgium and licensee Boehringer Ingelheim) are naturallyoccurring single chain antibodies of Camelidae. In 45 April 2012, a CTA was submitted to European regulatory authorities for a Phase I study (Thomson Reuters Pharma, update of August 23, 2012). NEOD-001 (Onclave Therapeutics, Dublin, Ireland and Neotope Biosciences, South San Francisco, CA and Elan) is a proprietary monoclonal antibody for the potential treatment of AL amyloidosis. In February 2012, the drug was awarded Orphan status by the FDA for AA amyloidosis and AL amyloidosis (Thomson Reuters Pharma, update of June 27, 2012). STC-103 (STC Biologics, Cambridge, MA) is a novel Fc fusion protein therapeutic for the potential treatment of AD (Thomson Reuters Pharma, update of August 10, 2012). Excellent papers on A antibodies from universities and basic research from companies were presented (in chronological 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 chronological 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 challenge of adverse effects of immunotherapy for AD was addressed [1480]. A clinical review of active and passive immunotherapeutic approaches in AD targeting A was presented [1375, 1376]. The development of 11A1 (a monoclonal antibody against the toxic conformer of A42 ; Tokyo Metropolitan Institute of Gerontology, Kyoto University and Immuno-Biological Laboratories), A-11 (A deposition-inhibiting antibody; Kinexis under license from the University of California), AAB002 (Janssen Alzheimer Immunotherapy and Pfizer), ABP-102 (Abiogen Pharma; a catalytic monoclonal antibody, an abzyme), ACU-5A5, ACU-0101979 and huC091 (Acumen Pharmaceuticals), anti-Abeta monoclonal antibodies (Mapp Biopharmaceutical in collaboration with Johns Hopkins University), the anti-amyloid beta antibody therapy targeting specific forms of the A parenchymal plaque (sanofi under license from Rockefeller University), anticalin (Pieris, an antibody against residues 16-26 of A), AZD3102 (AstraZeneca), bapineuzumab (AAB-001; Janssen Alzheimer Immuno-therapy and Pfizer), beta amyloid deposition-inhibiting antibodies (amyloidosis, Kinexis), beta amyloid-targeting antibodies (Mindset BioPharmaceuticals), encapsulated scFv- 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 tetrapeptide region of A (scFv-beta 1; Novartis in collaboration with the Institute for Research in Biomedicine and the Ecole Polytechnique Fédérale de Lausanne), ESBA-212 (ESBA Tech, Nestlé, a humanized single-chain antibody directed against A), KNXMonoclonal204 and KNX-Monoclonal205 (Kinexis under license from the University of California at Irvine), m266 (an antibody specific for the central domain of A; Lilly [1481, 1482]), and ponezumab (PF-4360365; RN-1219; RI-1219; Rinat Neuroscience and Pfizer) [1276, 1483–1485] a humanized monoclonal antibody specific for the C-terminus of A40 , was terminated. DRUGS INTERACTING WITH TAU A hallmark of the AD brain is the presence of inclusions within neurons that are comprised of fibrils formed from hyperphosphorylated microtubulestabilizing protein tau (paired helical filaments and neurofibrillary tangles [1486–1489]. The formation of misfolded multimeric tau species is believed to contribute to the progressive neuron loss and cognitive impairments of AD [1490]. Tau suppression in a neurodegenerative mouse model improved memory function [1491]. The precise composition of the neurotoxic species Tau-P* is not defined yet [1486]. The levels of early multimeric tau-aggregates that preceded the neurofibrillary tangles were found to correlate with memory deficits [1492]. The sites of phosphorylation and the involved kinases were compiled [1493, 1494]. The tau phosphorylation pathway genes were described [1495]. Although A plaques may play a key role in AD pathogenesis the severity of cognitive impairment correlated best with the burden of neurofibrillary tangles described in an exhaustive review of the literature [1496, 956]. A and tau influence each other in the mediation of toxicity. A formation in APP transgenic mice caused hyperphosphorylation of tau, whereas there was no overt A plaque pathology in tau transgenic mice. Immunization against A in triple transgenic mice resulted in reduced levels of hyperphosphorylated tau. Tau reduction prevented A induced defects in the axonal transport of mitochondria. Combination of these data led to the two step tau axis hypothesis of AD [1497–1499]. First, postsynaptic toxicity of A is tau-dependent, because tau interacts with FYN [1500], which is localized to the dendritic compartment, where it phosphorylates the NMDA receptor subunit 2B, mediates their inter- action with the postsynaptic density protein 95, an interaction required for A toxicity and second, A triggers progressively increased phosphorylation of tau compromising the binding of tau to microtubules leading to increasing dendritic tau levels [1501]. For a very instructive figure depicting these interactions, see [1502]. The synaptic accumulation of hyperphosphorylated tau oligomers is associated with dysfunction of the ubiquitin-proteasome system [1503]. A cascade of biomarkers was proposed: A precedes tau-mediated neuronal injury; A- and tau-mediated abnormalities precede neuroimaging biomarkers such as changes in hippocampal or ventricular volumetry measured by MRI; and all of these precede cognitive change marked by progressive deterioration in episodic memory and other cognitive domains such 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 of biomarkers were examined. Results indicated that CSF A effects on cognition change were substantially attenuated by CSF tau and measures of brain structure and function and CSF tau effects on cognitive change were attenuated by neuroimaging variables. Contrary to hypotheses, CSF A and CSF tau were observed to have independent effects on neuroimaging and CSF tau had a direct effect on baseline cognition independent of brain structure and function [1507]. Small molecules preventing tau aggregation TRx-0014 (methylthioninium chloride, methylene blue, Rember; TauRx Therapeutics, Singapore, a spinout from the University of Aberdeen) (Fig. 15) is a tau protein aggregation inhibitor. In August 2004, a placebo-controlled, dose-ranging Phase II trial was initiated in subjects (n = 275) with AD associated dementia in the UK. The doses were 30, 60, or 100 mg tid. In July 2007, a Phase II open-label continuation study of two doses of 30 or 60 mg tid gelatin capsules started for 52 weeks. The drug reduced cognitive decline by 81% over 1 year and no significant loss of cognitive function was seen over 19 months [1508, 1509]. In November 2010, the EC granted the drug Orphan status for frontotemporal dementia, progressive non-fluent aphasia and progressive supranuclear palsy. In February 2012, an open-label trial in patients with frontotemporal dementia was ongoing. For the preclinical characterization, see [1510–1516]. Only when levels of soluble tau protein were concomitantly W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes reduced by a very high concentration of methylene blue cognitive improvement was observed in a mouse model of human tauopathy [1517]. Methylene blue also inhibited the aggregation of A [1518, 1519] (Thomson Reuters Pharma, update of September 10, 2012). LMT-X (TauRx Therapeutics, Singapore) is a potential follow-up compound and prodrug of Rember. In September 2012, a Phase III study for frontotemporal dementia began in 180 subjects for 12 months. In October 2012 two global phase III studies were initiated for AD. The first study will involve 833 patients with mild to moderate Alzheimer’s disease over 12 months, while the second study will include 500 patients with mild Alzheimer’s disease over 18 months. The clinical trials will be conducted in parallel and on a global basis including sites in Australia, Belgium, 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-protein attenuating compound that reduced levels of soluble A and tau hyperphosphorylation in a Phase IIa study in AD patients. (Thomson Reuters Pharma, update of October 2, 2012) See also Chapter 12. Metal Chelators. 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 by October 2012. First positive results of a clinical pilot study 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 small molecule microtubule stabilizer aimed at preventing the production of abnormal tau protein. A randomized, double-blind, placebo-controlled Phase I study was initiated in patients with mild AD (expected n = 100) in the US and in Europe in March 2012 (Thomson Reuters Pharma, update of August 6, 2012). The structure was not communicated. PP2A stimulator (Se-015; Ve-015; Velacor Therapeutics, Victoria, Australia) is a selenium derivative acting as protein phosphatase 2A (PP2A) stimulator resulting in dephosphorylation of Akt kinase and tau. By February 2011, a Phase I trial had been completed and the company received the approval to initiate a Phase IIa trial in AD patients in Australia (Thomson Reuters Pharma, update of February 10, 2011). The structure has not been communicated. 47 There are many compounds inhibiting tau aggregation in preclinical evaluation (in alphabetical order): A/tau protein aggregation inhibitors (CNRS, FIST SA, Paris, France) are investigated for the potential treatment of AD (Thomson Reuters Pharma, update of August 24, 2012). The structures of the compounds were not communicated. Astemizole and Lansoprazole have been found to selectively interact with tau polymers [1521]. Berberine attenuated tau hyperphosphorylation in HEK293 cells [1745]. Bezafibrate (Bezalip, Bezatol; Boehringer Mannheim, now Roche, co-marketing with Kissei Pharmaceuticals, launched 1977) (Fig. 15), a drug for the treatment of hyperlipidemia, is a pan-PPAR agonist, which improved behavioral deficits and tau pathology in P301S mice exerting a preventive effect [1522]. BLV-0703 (BLV-200703; Bioalvo, Lisbon, Portugal) (Fig. 15) is a tau aggregation inhibitor in preclinical development (Thomson Reuters Pharma, update of May 9, 2011). Epothilone D improved microtubule density, axonal integrity and cognition in a transgenic mouse model of tauopathy [1523]. Fulvic acid inhibited aggregation and promoted the disassembly of tau fibrils associated with AD [639]. Insulin intranasal ameliorated tau hyperphosphorylation in a rat model of type 2 diabetes [1524]. L-3-n-butylphthalide reduced tau phosphorylation and improved cognitive deficits in APP/PS1Alzheimer’s transgenic mice [1746]. NBB BSc3504 (TU Darmstadt and Max-PlanckUnit for Structural Molecular Biology Hamburg) (Fig. 15) is a N -benzylidene-benzohydrazide showing remarkable tau aggregation inhibition (IC50 > 0.97 M) and paired helical filament depolymerization (DC50 > 1.14 M), but poor affinity towards A1-42 fibrils (IC50 > 33 M) [1525]. NC-11813 (Eli Lilly and Nymirum, Ann Arbor, MI) is an inhibitor of tau exon 10 splicing, which stabilized the tau splice regulatory element (Thomson Reuters Pharma, update of August 10, 2012). The structure was not communicated. NP-111001 derivatives (Noscira, Madrid) act as tau phosphorylation inhibitors (Thomson Reuters Pharma, update of January 12, 2012). The structures were not communicated. NPT-002 (NeuroPhage Pharmaceuticals, Cambridge, MA) targets A and tau aggregates for the potential treatment of AD (Thomson Reuters Pharma, 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. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes update of July 16, 2012). The structure was not communicated. Protein phosphatase methylesterase 1 (PME1) inhibitors (Signum Biosciences and GSK) are small molecules blocking the enzyme which demethylates PP2A. Normally PP2A is almost all methylated, but in Alzheimer’s disease PP2A methylation is reduced in half. The structural mechanism of demethylation and inactivation of protein phosphatase 2a was described [1747–1749] (Thomson Reuters Pharma, update of November 15, 2012). The structures were not communicated. Protein phosphatase 2A stimulators (Cognosci, Research Triangle Park, NC) inhibit the formation of apoE/inhibitor 2 of PP2A (I2PP2A) complex. The topic protein phosphatases and AD was reviewed [1526, 1527] (Thomson Reuters Pharma, update of February 24, 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 tau detoxifying compounds targeting tau-mediated cytotoxicity (Thomson Reuters Pharma, update of September 14, 2012). The structures were not communicated. The ReS9-S7 and ReS12-S programs (reMYND, a spin-off from the University of Leuven, Belgium and licensee Roche) investigate the attenuation of the cytotoxic effects of ␣-synuclein aggregation in dopaminergic neurons for the potential treatment of PD (Thomson Reuters Pharma, update of September 14, 2012). The structures were not communicated. SIG-1012 (Cognion) and SIG-1106 (Signum Biosciences, Monmouth Junction NJ in collaboration with Princeton Univ.) are phosphoprotein 2A modulators and presumed to also include the PP2A methylesterase 1 inhibitor eicosanoyl-5-hydroxytryptamide (EHT), all extracted from coffee and acting as tau aggregation inhibitors. (Thomson Reuters Pharma, update of October 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 of AD (Thomson Reuters Pharma, update of October 3, 2012). Structures were not disclosed. Small molecule tau modulators (Yuma Therapeutics, Brookline, MA) target neurofibrillary tangles resulting from abnormal forms of the protein tau (Thomson Reuters Pharma, update of March 29, 2011). Sodium selenate mitigated tau pathology and functional deficits in AD models [1528]. Tau aggregation inhibitors (Catholic University of Leuven, Belgium) are currently in the preclinical Phase 49 (Thomson Reuters Pharma, update of July 30, 2012). Structures were not communicated. Tau aggregation inhibitors (ProteoTech, Kirkland, 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 (Thomson Reuters Pharma, update of April 24, 2012). Tau phosphorylation inhibitors (ProQinase, Freiburg im Breisgau, Germany) (Fig. 15) reduced in vivo GSK-3 selective phosphorylation of Ser396/Ser404 and Ser262 in the brains of triple transgenic mice after i.p. administration. It appears that the development was terminated (Thomson Reuters Pharma update of October 11, 2012). Therapeutic program (B & C Biopharm and Equispharm, both South Korea) is targeting tau kinase (Thomson Reuters Pharma update of May 18, 2012). Thiamet-G (Simon Fraser University) is a potent inhibitor of human O-GlcNAcase (Ki = 21 nM) (Fig. 15) and efficiently reduced phosphorylation of tau at Thr231, Ser396, and Ser422 in both rat cortex and hippocampus [1529–1533]. For commentaries, see [1534, 1535]. See also Part 2, Chapter 2.27. Drugs interacting with O-GlcNAcase. THQ-4S and THQ-55 (Fig. 15) interact specifically with oligomeric forms of tau inhibiting their assembly into AD filaments [1536, 1537]. TRx-0237 (TauRx Therapeutics, Singapore) is the lead compound of second-generation tau aggregation inhibitors. Safety and efficacy studies in AD and frontotemporal 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 selective HDAC6 inhibitor [1750]. Chronic tubastatin treatment for two months decreased total tau levels in rtg4510 mice [1751]. Natural products as a source of tau-targeting drugs were described [1538]. Excellent papers on small molecule inhibitors of tau aggregation were published by researchers at universities (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 discovery for AD were presented (in chronological order) 2000: [1562]; 2002: [1563], 2006: [1564], 2007: 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 interactions between repeats in tau protein and A were described [1582]. The development of the tau aggregation inhibitors aminothienopyridazines (University of Pennsylvania), AZD-1080 (AstraZeneca), BPU-410 and BPU-430 (Champions Biotechnology under license from Johns Hopkins University), LDN-33960 (a striatal-enriched protein tyrosine phosphatase (STEP) inhibitor, Yale University), NHT-0112 (NeuroHitech), protein phosphatase methylesterase 1 (PME1) inhibitors (Scripps Research Institute and Massachusetts Institute of Technology), SDGIT200801 and SDGII-T200801 (both Bioalvo), SRN-003-556 (SIRENADE Pharmaceuticals) and of TRx-0037 (TauRx) were terminated. Ligands interacting with tau T-777, T-807, and T-808 (Siemens Medical Solution Molecular Imaging) (Fig. 15) are tau-binding molecules as PET tracers for the diagnosis of AD. They are benz[4,5]imidazo[1,2-a]pyrimidines. 18 FT808 displayed a high level of binding affinity (Kd = 22 nM) and good selectivity for tau aggregates over A plaques [1583] (Thomson Reuters Pharma, update of September 19, 2011). Novel in vivo tau imaging agents 18 F-THK-523 (University of Melbourne) [1584] and 11 C-THK-951 (Tohoku University) (both Fig. 15) were published [1585] after preliminary results of 2005 [1752] (Thomson Reuters Pharma, update of September 219, 2012). The University of Melbourne is investigating 18 F labeled arylquinoline derivatives, such as 18 F-THK-5105, 18 F-THK-5116, 18 F-THK-5117, and 18 F-THK-5125 for the potential use in imaging tau deposition in neurofibrillary tangles for AD (Thomson Reuters Pharma, update of June 13, 2012). The structures were not communicated. Researchers of Amersham (GE Healthcare) published a patent describing ligands with an exceptional selectivity to tau: compound 18 (Fig. 15): Kd = 0.9 nM for tau and Kd = 30 M for A1-40 [1586]. A 125 I-3-oxindole derivative stained neurofibrillary tangles in AD brain sections [1587]. Also bis(arylvinyl)pyrazines, -pyrimidines, and -pyridazines were described as imaging agents for tau fibrils and A plaques [1588] as were 2-styrylindolium based fluorescent probes [1753]. Research toward tau imaging was reviewed [1553, 1589, 1590] stating that ligand polarizability contributes to tau fibril binding affinity. Vaccines against tau Recombinant misfolded truncated tau protein vaccine (Axon Neurosciences, Vienna, Austria) attenuated pathology in vivo in a transgenic rat model of tauopathy (Thomson Reuters Pharma, update of February 17, 2011). RV-03 (Intellect Neurosciences, New York, NY) is a peptide vaccine developed using the RECALLVAX technology targeting both the A and a truncated delta tau protein (Thomson Reuters Pharma, update of August 21, 2012). Phosphorylated tau peptides were used to immunize transgenic mice to produce robust anti-neurofibrillary tangle effects [1591]. The development of the chimeric peptide vaccine RV-02 (Intellect Neurosciences) was terminated. Antibodies against tau and α-synuclein Humanized tau monoclonal antibodies (AC Immune, Lausanne, Switzerland) had high specific affinity binding to pTau. The compound was outlicensed to Genentech (Thomson Reuters Pharma, update of June 19, 2012). Lilly presented data on passive immunization with anti-tau antibodies in two transgenic mouse models [1754]. NI-105 (Biogen Idec, Weston, MA through the acquisition of Panima, previously a subsidiary of Neurimmune Therapeutics, Zurich Switzerland) is a human recombinant monoclonal antibody targeted to tau protein created using Neurimmune’s reverse translational 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 avian antibody to its cognate phosphopeptide pT231/pS235 at 1.9 Å resolution [1755]. T01-OX2 (Intellect Neurosciences, New York, NY) is an antibody drug conjugate that consists of a mAb targeting oligomeric forms of tau protein conjugated to melatonin (Thomson Reuters Pharma, update of August 17, 2012). W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes TauC3 monoclonal antibody (Intellect Neurosciences, New York, NY under license from Northwestern University) is targeting the neoepitope tauC3 (Thomson Reuters Pharma, update of September 20, 2012). Tau protein modulator (iPierian, South San Francisco, CA) is a monoclonal antibody for the potential treatment of AD, frontotemporal dementia and progressive supranuclear palsy (Thomson Reuters Pharma, update of May 22, 2012). Tau-targeted antibody therapy (Neotope Biosciences, South San Francisco, CA and Elan) is a monoclonal antibody targeted to tau (Thomson Reuters Pharma, update of August 13, 2012). TOC-1 (Intellect Neurosciences, New York under license from Northwestern University) is a tauoligomer-targeting monoclonal antibody [1592, 1593] (Thomson Reuters Pharma, update of September 20, 2012). The detection of naturally occurring anti-tau antibodies was described [1594]. Already in 1985 monoclonal antibodies to AD neurofibrillary tangles were described [1595]. Excellent papers on passive immunization targeting pathological phospho-tau protein were published (in chronological order) 2005: [1596], 2007: [1597], 2008: [1598, 1599], 2009: [1600, 1601], 2010: [1602, 1603], 2011: [1604–1607], 2012: [1756]. The development of TTRY78F (University of Porto), an anti-transthyretin monoclonal antibody, was terminated. STEM CELLS Substantial progress has been achieved in research of stem cells for the potential treatment of AD communicated 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 choline acetyltransferase restored cognitive function of kainicacid-induced learning and memory deficits in rats [1643, 1644]. Neural stem cells reduced hippocampal tau and reelin accumulation [1645]. Intracerebral transplantation of bone marrow-derived mesenchymal stem cells reduced A deposition and rescued memory deficits in AD mice by modulation of immune responses [1626]. Neural stem cells improved cognition via BDNF in a transgenic model of AD 51 [1646–1648]. Human neural stem cells genetically modified to express human NGF gene restored cognition in mice [1649]. Human neural stem cells genetically modified to overexpress BDNF promote functional recovery and neuroprotection in a mouse stroke model [1650]. Neural stem cells improved learning and memory in rats with AD [1651] and improved neuronal survival in cultured postmortem brain tissue from aged and AD patients [1652]. Induced pluripotent stem cells may be used to model patient-specific AD in vitro [1653, 1654]. For a commentary, see [1655]; to model pathology in Down syndrome [1656] and in HD [1657]. They may create new opportunities for disease modeling and drug discovery [1758]. GDNF/BDNF-producing glial and brain-derived stem cells (NurOwn; BrainStorm Cell Therapeutics, New York, NY) is investigated as a potential treatment of PD, sciatica, and multiple sclerosis [1658, 1659]. A Phase I/II clinical trial in ALS patients started in June 2011 in Israel. In January 2012 positive data were reported (Thomson Reuters Pharma, update of July 24, 2012). Human neural stem cell therapy (HuCNS-SC; StemCells, Newark, CA, formerly CytoTherapeutics under license from NeuroSperes) is a proprietary transplantable human neural stem cell therapy for the potential treatment of spinal cord injury, age-related macular degeneration, and AD. Human neural stem cells induced functional myelination in mice [1759] and in the human brain [1760]. A Phase II trial for spinal cord injury was initiated in Switzerland in March 2011. The neuroprotective effects in retinal degeneration were described [1660] (Thomson Reuters Pharma, update of September 28, 2012). Mesenchymal bone marrow-derived stem cell therapy (Stemedica Cell Technologies, San Diego, CA) is being developed for the intravenous treatment of ischemic stroke in Phase I/II since February 2011 in the US (n = 35). The study was expected to be completed in February 2013 (Thomson Reuters Pharma, update of April 11, 2012). NSI-189 (Neuralstem, Rockville, MD, in collaboration with Japanese licensee Summit Pharmaceuticals) (Fig. 16) is an orally bioavailable small-molecule neurogenesis stimulator in Phase I, which was completed in October 2011. The FDA approved the Phase Ib trial in December 2011 (three cohorts of eight patients each ). In June 2012, the first patients were dosed (Thomson Reuters Pharma, update of September 28, 2012). NSI-566RSC (Neuralstem, Rockville, MD) are human neural stem cells including spinal cord stem 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 connectivity of neural stem cells after severe spinal cord injury were investigated [1661]. A Phase I clinical trial for ALS started in January 2010 in the US. The results of a Phase I trial in 12 ALS patients receiving lumbar intraspinal injection of neural stem cells were described [1662] (Thomson Reuters Pharma, update of September 28, 2012). Neurostem-AD (Medipost, South Korea) is an umbilical cord blood-derived mesenchymal stem cell therapy, which regenerates nerve cells. A Phase I trial has been initiated in February 2011 in patients (expected n = 9) with dementia of AD-type (Thomson Reuters Pharma, update of September 25, 2012). There are several stem cell preparations in preclinical evaluation (in alphabetical order): Adult mesenchymal precursor stem cell therapy (Mesoblast, Melbourne, Australia and Cephalon, a wholly-owned subsidiary of Teva) is evaluated for the potential treatment of AD and PD and stroke (Thomson Reuters Pharma, update of September 6, 2012). Allogenic umbilical cord stem cell therapy (U-CORD; CellPraxis Bioengenharia, Bela Vista, Brazil) is investigated for the potential treatment of Alzheimer’s disease. (Thomson Reuters Pharma, update of November 07, 2012). Brain-derived stem cell therapy (Celprogen, San Pedro, CA, in collaboration with the Indiana University School of Medicine) can be differentiated into neurons. In vivo preclinical studies demonstrated improvement in short and long term memory in AD experimental models (Thomson Reuters Pharma, update of July 14, 2011). CPG23NEUR (Celprogen, San Pedro, CA) derives from cord blood stem cells, which are transdifferentiating into neuronal cells (Thomson Reuters Pharma, update of January 30, 2012). Glial progenitor cell therapy (Q Therapeutics, Salt Lake City, UT) has the potential to replace myelin on damaged neurons for the treatment of transverse myelinitis, spinal cord injury, multiple sclerosis, PD, and AD (Thomson Reuters Pharma, update of January 20, 2012). Human neural progenitor cells (Cedars-Sinai Medical Center, Los Angeles, CA), which secrete growth factors including glial cell-derived neurotrophic factor, may be useful for the treatment of ALS (Thomson Reuters Pharma, update of July 31, 2012). Human umbilical cord blood cells (Saneron CCEL Therapeutics, Tampa FL) are cerebroprotective agents which showed cognition enhancing and amyloid- reducing activities for the potential treatment of stroke and Alzheimer’s disease (Thomson Reuters Pharma, update of October 26, 2012). NBI-18 (NeoCytex Biopharma, Covington, KY), a heterocyclic pyrimidine derivative, is a stem cell stimulator for the potential treatment of neurodegenerative diseases including ALS, AD, and PD (Thomson Reuters Pharma, update of January 19, 2012). The structure was not communicated. Neural stem cell therapy (Stemedica Cell Technologies, San Diego, CA) is an allogeneic therapy for the potential treatment of AD (Thomson Reuters Pharma, update of February 22, 2012). Another stem 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-microencapsulated porcine choroid plexus cell product for the potential treatment of neurodegenerative diseases including HD, PD, and dementia [1663, 1664] (Thomson Reuters Pharma, update of May 16, 2012). NGN-9079 (neural stem cell therapy; NeuroGeneration, Los Angeles, CA) is evaluated for the potential W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes treatment of AD. Transplantation of neuronal cells into the brain was shown to improve and delay the symptoms of AD in animal models (Thomson Reuters Pharma, update of March 2, 2012). ReN-004 (ReNeuron, Guildford, UK) is a neural stem cell therapy for PD and AD (Thomson Reuters Pharma, update of September 5, 2012). ReN-005 (ReNeuron, Guildford, UK) is a neural stem cell therapy for HD (Thomson Reuters Pharma, update of December 24, 2010). Stem cell therapeutics (Samaritan Pharmaceuticals, Las Vegas, NV) are non-embryonic neuronal stem cell differentiation therapeutics including the naturally occurring compounds SP-sc4 and SP-sc7 to induce stem cell differentiation for the potential treatment of AD (Thomson Reuters Pharma, update of March 16, 2011). Allopregnanolone (University of Southern California, Los Angeles, CA) is a potent and highly efficacious proliferative agent in vitro and in vivo of both rodent and human neural stem cells [1665–1667]. Granulocyte colony-stimulating factor is known to mobilize hematopoietic stem cells from the bone marrow into the peripheral blood. Subcutaneous administration of granulocyte colony-stimulating factor into two different A-induced AD mouse models substantially rescued their cognitive/memory functions [1668]. Valproic acid can induce neurogenesis of neural progenitor/stem cells both in vitro and in vivo via multiple signaling pathways [1669]. The development of GRNOPC-1 (Geron), oligodendrocyte precursor cells differentiated from a human embryonic stem cell line that produce neurotrophic factors and remyelinate axons for the potential injectable treatment of PD, stroke, AD, multiple sclerosis, and spinal cord injury in Phase I since October 2010 was terminated. Also the development of the human embryonic stem cell therapy (GRNIC-1, Geron), the cell-based therapy MDA-200C (Medeia Therapeutics), NEBO-176 (Neuro Bioscience and RLI), propentofylline (Endogenous Stem Cells Activators (ESAI) under license from sanofi) and of stem cell therapies for glaucoma, macular degeneration, PD, traumatic brain injury, and vascular dementia (Stemedica) was terminated. MISCELLANEOUS Autophagy inducer JRP-900 (Prous Institute for Biomedical Research, Barcelona) is investigated for 53 the potential treatment of neurodegenerative disease including AD, ALS, HD, and PD (Thomson Reuters Pharma, update of July 23, 2012). The structure was not communicated. For reviews on autophagy in Alzheimer’s disease and tauopathies see [1761–1764]. Cellular homeostasis modulator CNS-102 (Coyote Pharmaceuticals) acts on protein misfolding for the potential treatment of AD, multiple sclerosis, and ALS (Thomson Reuters Pharma, update of October 2, 2012). The structure was not communicated. Glycan inhibitors (Stelic Institute, Tokyo) are investigated for the potential treatment of Parkinson’s and Alzheimer’s disease. An exhaustive review was provided [1765]. (Thomson Reuters Pharma, update of October 16, 2012). Structures were not communicated. Macrophage migration inhibitory factor (MIF) inhibitor INV-88 (Innovimmune Biotherapeutics, New York) is investigated for the potential oral treatment of inflammatory diseases, central nervous system diseases including Alzheimer’s disease and multiple sclerosis and age-related macular degeneration (Thomson Reuters Pharma, update of October 18, 2012). The structure was not communicated. MicroRNA (miRNA) mimetics (University of Göttingen, MBM Science) are evaluated for the potential diagnosis and treatment of memory impairment. By June 2012, proof of concept was achieved in an AD mouse model and in human AD patient samples. The relationship of miRNAs in the brain and A generation was discussed [1670] (Thomson Reuters Pharma, update of August 7, 2012). Proteasome-gating modulators are investigated by Proteostasis Therapeutics, Cambridge, MA under license from Harvard University for the potential treatment of neurodegenerative diseases including AD and PD (Thomson Reuters Pharma, update of August 15, 2012). Synaptic vesicle glycoprotein 2A (SV2A) ligand levetiracetam (L-059, Keppra; UCB Pharma, Brussels, Belgium and Japanese licensee Otsuka) (Fig. 16) is an antiepileptic drug launched in the US and EU in 2000 and in Japan in 2010 [1671–1676]. It binds to the same binding site as botulinum neurotoxin A and tetanus neurotoxin [1677, 1678]. It is still unclear how levetiracetam modulates SV2A’s function(s). It inhibited presynaptic Ca2+ channels through an intracellular pathway [1679, 1680]. It may promote glutamate uptake by increasing glutamate transporter expression [1681]. There are hints that levetiracetam may interact with GABAA receptors [1682–1684]. Levetiracetam had a positive impact on emotional learning and memory in naı̈ve mice [1685] and 54 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes reversed cognitive deficits in hAPP transgenic mice [1686]. Levetiracetam improved cognitive function in drug-naı̈ve epilepsy patients [1687] and in children aged 4 to 16 years [1688]. The sales of levetiracetam reported by UCB for 2011 were USD 1,345 million and by Otsuka, USD 60.5. An injectable formulation was launched in the US in August 2006, an extended release formulation in September 2008 (Thomson Reuters Pharma, update of October 1, 2012). Low-dose therapy Levetiracetam (AgeneBio, Carmel, IN under license from John Hopkins University) is investigated for the potential treatment of amnestic MCI (aMCI) in Phase II clinical trials since December 2009 in 144 aMCI patients in the US. Reduction of hippocampal hyperactivity in aMCI patients by using a low dose of levetiracetam improved cognition [1766]. It appears that the development was terminated (Thomson Reuters Pharma, update of September 7, 2011). Brivaracetam (UCB-34714; Rikelta; UCB Pharma, Brussels, Belgium) (Fig. 16), the n-propyl analogue of levetiracetam, is an orally active ligand of synaptic vesicle protein 2A (SV2A) currently in Phase III clinical trials for the treatment of epilepsy since November 2008 (n = 600). The study is expected to complete in June 2015 [1689, 1690]. Its binding characteristics as a high affinity SV2A ligand in rat, mouse and human brain were reported [1691] as was its effect on the inhibition of Na+ currents [1692]. Its neurocognitive effects were similar to the ones of levetiracetam [1693, 1694] (Thomson Reuters Pharma, update of August 8, 2012). The development of the second Phase III monoclonal antibody bapineuzumab (Janssen Alzheimer Immunotherapy and Pfizer) was terminated. The Phase III clinical trials of other A PET imaging agents, 18 F-florbetaben (Piramal) and 18 Fflutemetamol (GE Healthcare), may be successfully completed by next year. The completion of Phase III clinical trials of the VMAT2 PET ligand 18 Fflorbenazine (Avid Radiopharmaceuticals, Lilly) is planned for September 2014. Phase II clinical trials of other disease modifying drugs, of monoclonal antibodies against A gantenerumab, crenezumab, GSK-93377A, intravenous immune globulin, and octagam, of vaccines against A AD-02, CAD-106, and vanutide cridificar and the PET ligand 18 F-AZD-4694 (Navidea) will require several years. This applies also for the Phase II clinical trials of small molecules preventing A aggregation or inhibiting formation of transthyretin amyloid fibrils, such as doxycycline hyclate, ELND005, 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-PCB70M (a natural product) followed by 22 Phase II compounds, RV-208 (a gene expression stimulator), ARC-029 and ZSET-1446/ST-101 (calcium channel blockers), CERE-110, GM-607, PYM-50058, and T-817MA (trophic factor stimulators), circadin, KD501, PPL, PTX-200, resveratrol, RPh-201 (natural products), LSL-001, N-251, PF-03049423, TRM189, VI-1121 (nootropics), davunetide, AM-111, etanercept, and FGL (peptides). CONCLUSION DISCLOSURE STATEMENT Two products were launched in 2012: tafamidis (Vyndaquel, Pfizer) for the treatment of transthyretin familial amyloid polyneuropathy in Europe in March and 18 F-florbetapir (Amyvid; Avid Radiopharmaceuticals, Lilly) as a PET imaging agent to estimate A neuritic plaque density in patients with cognitive impairment in the US in June. Phase III Alzheimer’s disease trials of a disease modifying drug, the monoclonal antibody against A solanezumab (Lilly), are close to completion. First results showed that solanezumab failed to meet the cognitive and functional primary endpoints. But it showed a significant reduction in cognitive decline in patients with mild AD. For a commentary, see [1695]. The open-label EXPEDITION-EXT extension trial, which is fully enrolled, is continuing as planned. Authors’ disclosures available online (http://www.jalz.com/disclosures/view.php?id=1543). REFERENCES [1] [2] [3] Giurgea C (1972) [Pharmacology of integrative activity of the brain. Attempt at nootropic concept in psychopharmacology. Actual Pharmacol (Paris) 25, 115-156. Giurgea C (1973) The “nootropic” approach to the pharmacology of the integrative activity of the brain. Cond Reflex 8, 108-115. 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, Whittington M, Young LJ (2012) Cognitive dysfunction in psychiatric W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [4] [5] [6] [7] [8] [9] [10] [11] [12] [13] [14] [15] [16] [17] [18] [19] [20] [21] [22] disorders: Characteristics, causes and the quest for improved therapy. Nat Rev Drug Discov 11, 141-168. Froestl W, Maitre L (1989) The families of cognition enhancers. Pharmacopsychiatry 22(Suppl 2), 54-100. Froestl W, Muhs A, Pfeifer A (2012) Cognitive Enhancers (Nootropics). Part 1: Drugs interacting with receptors. J Alzheimers Dis 32, 793-887. Citron M (2004) Strategies for disease modification in Alzheimer’s disease. Nat Rev Neurosci 5, 677-685. Pangalos MN, Jacobsen SJ, Reinhart PH (2005) Disease modifying strategies for the treatment of Alzheimer’s disease targeted at modulating levels of the beta-amyloid peptide. Biochem Soc Trans 33, 553-558. Hull M, Berger M, Heneka M (2006) Disease-modifying therapies in Alzheimer’s disease: How far have we come? Drugs 66, 2075-2093. Vellas B, Andrieu S, Sampaio C, Wilcock G (2007) Disease-modifying trials in Alzheimer’s disease: A European task force consensus. Lancet Neurol 6, 56-62. Sadowski M, Wisniewski T (2007) Disease modifying approaches for Alzheimer’s pathology. Curr Pharm Des 13, 1943-1954. Salloway S, MintzerJ, Weiner MF, Cummings JL (2008) Disease-modifying therapies in Alzheimer’s disease. Alzheimers Dement 4, 65-79. Sabbagh MN, Richardson S, Relkin N (2008) Diseasemodifying approaches to Alzheimer’s disease: Challenges and opportunities-Lessons from donepezil therapy. Alzheimers Dement 4, S109-S118. Vellas B, Coley N, Andrieu S (2008) Disease modifying trials in Alzheimer’s disease: Perspectives for the future. J Alzheimers Dis 15, 289-301. Duara R, Barker W, Loewenstein D, Bain L (2009) The basis for disease-modifying treatments for Alzheimer’s disease: The Sixth Annual Mild Cognitive Impairment Symposium. Alzheimers Dement 5, 66-74. 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 of Alzheimer’s disease: From alpha-secretase activators to gamma-secretase inhibitors and modulators. Drugs Aging 26, 537-555. Citron M (2010) Alzheimer’s disease: Strategies for disease modification. Nat Rev Drug Discov 9, 387-398. Frisardi V, Solfrizzi V, Imbimbo PB, Capurso C, D’Introno A, Colacicco AM, Vendemiale G, Seripa D, Pilotto A, Capurso A, Panza F (2010) Towards disease-modifying treatment of Alzheimer’s disease: Drugs targeting betaamyloid. Curr Alzheimer Res 7, 40-55. Dash SK (2011) Future targeted disease modifying drugs for Alzheimer’s disease. Recent Pat CNS Drug Discov 6, 65-76. Galimberti D, Scarpini E (2010) Treatment of Alzheimer’s disease: Symptomatic and disease-modifying approaches. Curr Aging Sci 3, 46-56. Galimberti D, Scarpini E (2011) Disease-modifying treatments for Alzheimer’s disease. Ther Adv Neurol Disord 4, 203-216. Galimberti D, Scarpini E (2011) Alzheimer’s disease: From pathogenesis to disease-modifying approaches. CNS Neurol Disord Drug Targets 10, 163-174. Salomone S, Caraci F, Leggio GM, Fedotova J, Drago F (2012) New pharmacological strategies for treatment of Alzheimer’s disease: Focus on disease modifying drugs. Br J Clin Pharmacol 73, 504-517. [23] [24] [25] [26] [27] [28] [29] [30] [31] [32] [33] [34] [35] [36] 55 Panza F, Frisardi V, Solfrizzi V, Imbimbo BP, Logroscino G, Santamato A, Greco A, Seripa D, Pilotto A (2012) Immunotherapy for Alzheimer’s disease: From anti-beta-amyloid to tau-based immunization strategies. Immunotherapy 4, 213-238. Cummings JL, Doody R, Clark C (2007) Diseasemodifying therapies for Alzheimer disease: Challenges to early intervention. Neurology 69, 1622-1634. Cummings JL (2008) Optimizing phase II of drug development for disease-modifying compounds. Alzheimers Dement 4, S15-S20. Cummings JL (2009) Defining and labeling diseasemodifying treatments for Alzheimer’s disease. Alzheimers Dement 5, 406-418. Jiang T, Yu JT, Tan L (2012) Novel disease-modifying therapies for Alzheimer’s disease. J Alzheimers Dis 31, 475-492. Lee KS, Chung JH, Choi TK, Suh SY, Oh BH, Hong CH (2009) Peripheral cytokines and chemokines in Alzheimer’s disease. Dement Geriatr Cogn Disord 28, 281-287. Gitter BD, Cox LM, Rydel RE, May PC (1995) Amyloid beta peptide potentiates cytokine secretion by interleukin1 beta-activated human astrocytoma cells. Proc Natl Acad Sci U S A 92, 10738-10741. Harries LW, Bradley-Smith RM, Llewellyn DJ, Pilling LC, Fellows A, Henley W, Hernandez D, Guralnik JM, Bandinelli S, Singleton A, Ferrucci L, Melzer D (2012) Leukocyte CCR2 expression is associated with mini-mental state examination score in older adults. Rejuvenation Res 15, 395-404. Ralay Ranaivo H, Craft JM, Hu W, Guo L, Wing LK, Van Eldik LJ, Watterson DM (2006) Glia as a therapeutic target: Selective suppression of human amyloid-beta-induced upregulation of brain proinflammatory cytokine production attenuates neurodegeneration. J Neurosci 26, 662-670. Bachstetter AD, Norris CM, Sompol P, Wilcock DM, Goulding D, Neltner JH, St CD, Watterson DM, Van Eldik LJ (2012) Early stage drug treatment that normalizes proinflammatory cytokine production attenuates synaptic dysfunction in a mouse model that exhibits age-dependent progression of Alzheimer’s disease-related pathology. J Neurosci 32, 10201-10210. 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 brain proinflammatory cytokine up-regulation that attenuates synaptic dysfunction and behavioral deficits. Bioorg Med Chem Lett 17, 414-418. Karpus WJ, Reynolds N, Behanna HA, Van Eldik LJ, Watterson DM (2008) Inhibition of experimental autoimmune encephalomyelitis by a novel small molecular weight proinflammatory cytokine suppressing drug. J Neuroimmunol 203, 73-78. Lloyd E, Somera-Molina K, Van Eldik LJ, Watterson DM, Wainwright MS (2008) Suppression of acute proinflammatory cytokine and chemokine upregulation by post-injury administration of a novel small molecule improves long-term neurologic outcome in a mouse model of traumatic brain injury. J Neuroinflammation 5, 28. Somera-Molina KC, Robin B, Somera CA, Anderson C, Stine C, Koh S, Behanna HA, Van Eldik LJ, Watterson DM, Wainwright MS (2007) Glial activation links early-life seizures and long-term neurologic dysfunction: Evidence using a small molecule inhibitor of proin- 56 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [37] [38] [39] [40] [41] [42] [43] [44] [45] [46] [47] [48] [49] [50] [51] [52] flammatory cytokine upregulation. Epilepsia 48, 17851800. O’Hare E, Scopes DI, Treherne JM, Monaghan J, Palmer PM, Amijee H, Kim EM (2011) Novel anti-inflammatory compound SEN1176 alleviates behavioral deficits induced following bilateral intrahippocampal injection of aggregated amyloid-beta. J Alzheimers Dis 25, 219-229. Shi JQ, Wang BR, Jiang WW, Chen J, Zhu YW, Zhong LL, Zhang YD, Xu J (2011) Cognitive improvement with intrathecal administration of infliximab in a woman with Alzheimer’s disease. J Am Geriatr Soc 59, 1142-1144. Bacher M, Dodel R, Aljabari B, Keyvani K, Marambaud P, Kayed R, Glabe C, Goertz N, Hoppmann A, Sachser N, Klotsche J, Schnell S, Lewejohann L, Al-Abed Y (2008) CNI-1493 inhibits Abeta production, plaque formation, and cognitive deterioration in an animal model of Alzheimer’s disease. J Exp Med 205, 1593-1599. Bach JP, Mengel D, Wahle T, Kautz A, Balzer-Geldsetzer M, Al-Abed Y, Dodel R, Bacher M (2011) The role of CNI1493 in the function of primary microglia with respect to amyloid-beta. J Alzheimers Dis 26, 69-80. Nilsson P, Iwata N, Muramatsu S, Tjernberg LO, Winblad B, Saido TC (2010) Gene therapy in Alzheimer’s disease–potential for disease modification. J Cell Mol Med 14, 741-757. Goldberg TE, Weinberger DR (2004) Genes and the parsing of cognitive processes. Trends Cogn Sci 8, 325-335. Zetterstrom TS, Pei Q, Madhav TR, Coppell AL, Lewis L, Grahame-Smith DG (1999) Manipulations of brain 5-HT levels affect gene expression for BDNF in rat brain. Neuropharmacology 38, 1063-1073. Coppell AL, Pei Q, Zetterstrom TS (2003) Bi-phasic change in BDNF gene expression following antidepressant drug treatment. Neuropharmacology 44, 903-910. Bourhis E, Maheux J, Rouillard C, Levesque D (2008) Extracellular signal-regulated kinases (ERK) and protein kinase C (PKC) activities are involved in the modulation of Nur77 and Nor-1 expression by dopaminergic drugs. J Neurochem 106, 875-888. Uusi-Oukari M, Korpi ER (2010) Regulation of GABA(A) receptor subunit expression by pharmacological agents. Pharmacol Rev 62, 97-135. Southwell AL, Patterson PH (2011) Gene therapy in mouse models of huntington disease. Neuroscientist 17, 153162. 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 hepatocyte growth factor to treat peripheral arterial disease. Hypertension 44, 203-209. Makino H, Aoki M, Hashiya N, Yamasaki K, Azuma J, Sawa Y, Kaneda Y, Ogihara T, Morishita R (2012) Longterm follow-up evaluation of results from clinical trial using hepatocyte growth factor gene to treat severe peripheral arterial disease. Arterioscler Thromb Vasc Biol 32, 2503-2509. Anonymous (2011) Rvx 208. Drugs R D 11, 207-213. Nicholls SJ, Gordon A, Johannson J, Ballantyne CM, Barter PJ, Brewer HB, Kastelein JJ, Wong NC, Borgman MR, Nissen SE (2012) ApoA-I induction as a potential cardioprotective strategy: Rationale for the SUSTAIN and ASSURE studies. Cardiovasc Drugs Ther 26, 181-187. Bailey D, Jahagirdar R, Gordon A, Hafiane A, Campbell S, Chatur S, Wagner GS, Hansen HC, Chiacchia FS, Johansson J, Krimbou L, Wong NC, Genest J (2010) RVX-208: [53] [54] [55] [56] [57] [58] [59] [60] [61] [62] [63] [64] [65] [66] A small molecule that increases apolipoprotein A-I and high-density lipoprotein cholesterol in vitro and in vivo. J Am Coll Cardiol 55, 2580-2589. McNeill E (2010) RVX-208, a stimulator of apolipoprotein AI gene expression for the treatment of cardiovascular diseases. Curr Opin Investig Drugs 11, 357-364. Davidson MH (2011) Apolipoprotein A-I therapy promise, challenges, and disappointment. J Am Coll Cardiol 57, 1120-1121. Nicholls SJ, Gordon A, Johansson J, Wolski K, Ballantyne CM, Kastelein JJ, Taylor A, Borgman M, Nissen SE (2011) Efficacy and safety of a novel oral inducer of apolipoprotein a-I synthesis in statin-treated patients with stable coronary artery disease a randomized controlled trial. J Am Coll Cardiol 57, 1111-1119. Shah PK (2011) Atherosclerosis: Targeting endogenous apo A-I –a new approach for raising HDL. Nat Rev Cardiol 8, 187-188. Hudry E, Van DD, Kulik W, De Deyn PP, Stet FS, Ahouansou O, Benraiss A, Delacourte A, Bougneres P, Aubourg P, Cartier N (2010) Adeno-associated virus gene therapy with cholesterol 24-hydroxylase reduces the amyloid pathology before or after the onset of amyloid plaques in mouse models of Alzheimer’s disease. Mol Ther 18, 44-53. Yang SY, He XY, Miller D (2007) HSD17B10: A gene involved in cognitive function through metabolism of isoleucine and neuroactive steroids. Mol Genet Metab 92, 36-42. Hafez DM, Huang JY, Richardson JC, Masliah E, Peterson DA, Marr RA (2012) F-spondin gene transfer improves memory performance and reduces amyloid-beta levels in mice. Neuroscience 223, 465-472. Chiba T, Yamada M, Hashimoto Y, Sato M, Sasabe J, Kita Y, Terashita K, Aiso S, Nishimoto I, Matsuoka M (2005) Development of a femtomolar-acting humanin derivative named colivelin by attaching activity-dependent neurotrophic factor to its N terminus: Characterization of colivelin-mediated neuroprotection against Alzheimer’s disease-relevant insults in vitro and in vivo. J Neurosci 25, 10252-10261. Chiba T, Nishimoto I, Aiso S, Matsuoka M (2007) Neuroprotection against neurodegenerative diseases: Development of a novel hybrid neuroprotective peptide Colivelin. Mol Neurobiol 35, 55-84. Matsuoka M, Hashimoto Y, Aiso S, Nishimoto I (2006) Humanin and colivelin: Neuronal-death-suppressing peptides for Alzheimer’s disease and amyotrophic lateral sclerosis. CNS Drug Rev 12, 113-122. Yamada M, Chiba T, Sasabe J, Terashita K, Aiso S, Matsuoka M (2008) Nasal Colivelin treatment ameliorates memory impairment related to Alzheimer’s disease. Neuropsychopharmacology 33, 2020-2032. Arakawa T, Niikura T, Arisaka F, Kita Y (2008) Activitydependent neurotrophic factor, ADNF, determines the structure characteristics of Colivelin, a fusion protein of ADNF9 and Humanin analog. J Pept Sci 14, 631-636. Hashimoto Y, Ito Y, Niikura T, Shao Z, Hata M, Oyama F, Nishimoto I (2001) Mechanisms of neuroprotection by a novel rescue factor humanin from Swedish mutant amyloid precursor protein. Biochem Biophys Res Commun 283, 460-468. Maximov V, Martynenko A, Hunsmann G, Tarantul V (2002) Mitochondrial 16S rRNA gene encodes a functional peptide, a potential drug for Alzheimer’s disease and target for cancer therapy. Med Hypotheses 59, 670-673. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [67] [68] [69] [70] [71] [72] [73] [74] [75] [76] [77] [78] [79] [80] Tortosa E, Santa-Maria I, Moreno F, Lim F, Perez M, Avila J (2009) Binding of Hsp90 to tau promotes a conformational change and aggregation of tau protein. J Alzheimers Dis 17, 319-325. Salminen A, Ojala J, Kaarniranta K, Hiltunen M, Soininen H (2011) Hsp90 regulates tau pathology through co-chaperone complexes in Alzheimer’s disease. Prog Neurobiol 93, 99-110. Vilenchik M, Solit D, Basso A, Huezo H, Lucas B, He H, Rosen N, Spampinato C, Modrich P, Chiosis G (2004) Targeting wide-range oncogenic transformation via PU24FCl, a specific inhibitor of tumor Hsp90. Chem Biol 11, 787797. He H, Zatorska D, Kim J, Aguirre J, Llauger L, She Y, Wu N, Immormino RM, Gewirth DT, Chiosis G (2006) Identification of potent water soluble purine-scaffold inhibitors of the heat shock protein 90. J Med Chem 49, 381-390. Caldas-Lopes E, Cerchietti L, Ahn JH, Clement CC, Robles AI, Rodina A, Moulick K, Taldone T, Gozman A, Guo Y, Wu N, de SE, White J, Gross SS, Ma Y, Varticovski L, Melnick A, Chiosis G (2009) Hsp90 inhibitor PU-H71, a multimodal inhibitor of malignancy, induces complete responses in triple-negative breast cancer models. Proc Natl Acad Sci U S A 106, 8368-8373. Luo W, Dou F, Rodina A, Chip S, Kim J, Zhao Q, Moulick K, Aguirre J, Wu N, Greengard P, Chiosis G (2007) Roles of heat-shock protein 90 in maintaining and facilitating the neurodegenerative phenotype in tauopathies. Proc Natl Acad Sci U S A 104, 9511-9516. Cole P, Vasiliou S, Mormeno D, Rosa E (2009) Medicinal chemistry selections from the 237th American Chemical Society National Meeting & Exposition. Drugs Future 34, 509-520. Taldone T, Chiosis G (2009) Purine-scaffold Hsp90 inhibitors. Curr Top Med Chem 9, 1436-1446. Taldone T, Zatorska D, Patel PD, Zong H, Rodina A, Ahn JH, Moulick K, Guzman ML, Chiosis G (2011) Design, synthesis, and evaluation of small molecule Hsp90 probes. Bioorg Med Chem 19, 2603-2614. Biamonte MA, Shi J, Hong K, Hurst DC, Zhang L, Fan J, Busch DJ, Karjian PL, Maldonado AA, Sensintaffar JL, Yang YC, Kamal A, Lough RE, Lundgren K, Burrows FJ, Timony GA, Boehm MF, Kasibhatla SR (2006) Orally active purine-based inhibitors of the heat shock protein 90. J Med Chem 49, 817-828. Dickey CA, Dunmore J, Lu B, Wang JW, Lee WC, Kamal A, Burrows F, Eckman C, Hutton M, Petrucelli L (2006) HSP induction mediates selective clearance of tau phosphorylated at proline-directed Ser/Thr sites but not KXGS (MARK) sites. FASEB J 20, 753-755. 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, Burrows F, Petrucelli L (2007) The high-affinity HSP90-CHIP complex recognizes and selectively degrades phosphorylated tau client proteins. J Clin Invest 117, 648-658. Oddo S, Caccamo A, Tseng B, Cheng D, Vasilevko V, Cribbs DH, LaFerla FM (2008) Blocking Abeta42 accumulation delays the onset and progression of tau pathology via the C terminus of heat shock protein70-interacting protein: A mechanistic link between Abeta and tau pathology. J Neurosci 28, 12163-12175. Patterson KR, Ward SM, Combs B, Voss K, Kanaan NM, Morfini G, Brady ST, Gamblin TC, Binder LI (2011) Heat shock protein 70 prevents both tau aggregation and the [81] [82] [83] [84] [85] [86] [87] [88] [89] [90] [91] [92] [93] [94] [95] 57 inhibitory effects of preexisting tau aggregates on fast axonal transport. Biochemistry 50, 10300-10310. Wilhelmus MM, de Waal RM, Verbeek MM (2007) Heat shock proteins and amateur chaperones in amyloid-Beta accumulation and clearance in Alzheimer’s disease. Mol Neurobiol 35, 203-216. Evans CG, Chang L, Gestwicki JE (2010) Heat shock protein 70 (hsp70) as an emerging drug target. J Med Chem 53, 4585-4602. Yarbrough GG (1983) Minireview. Thyrotropin releasing hormone and CNS cholinergic neurons. Life Sci 33, 111118. Mellow AM, Sunderland T, Cohen RM, Lawlor BA, Hill JL, Newhouse PA, Cohen MR, Murphy DL (1989) Acute effects of high-dose thyrotropin releasing hormone infusions in Alzheimer’s disease. Psychopharmacology (Berl) 98, 403-407. Molchan SE, Mellow AM, Lawlor BA, Weingartner HJ, Cohen RM, Cohen MR, Sunderland T (1990) TRH attenuates scopolamine-induced memory impairment in humans. Psychopharmacology (Berl) 100, 84-89. Khan A, Mirolo MH, Claypoole K, Bhang J, Cox G, Horita A, Tucker G (1994) Effects of low-dose TRH on cognitive deficits in the ECT postictal state. Am J Psychiatry 151, 1694-1696. Urayama A, Yamada S, Hirano K, Deguchi Y, Kimura R (2001) Brain receptor binding characteristics and pharmacokinetic-pharmacodynamic analysis of thyrotropin-releasing hormone analogues. Life Sci 70, 647657. Urayama A, Yamada S, Kimura R, Zhang J, Watanabe Y (2002) Neuroprotective effect and brain receptor binding of taltirelin, a novel thyrotropin-releasing hormone (TRH) analogue, in transient forebrain ischemia of C57BL/6J mice. Life Sci 72, 601-607. Tanabe M, Tokuda Y, Takasu K, Ono K, Honda M, Ono H (2007) The synthetic TRH analogue taltirelin exerts modality-specific antinociceptive effects via distinct descending monoaminergic systems. Br J Pharmacol 150, 403-414. Grigorova M, Sherwin BB, Tulandi T (2006) Effects of treatment with leuprolide acetate depot on working memory and executive functions in young premenopausal women. Psychoneuroendocrinology 31, 935-947. Bowen RL, Verdile G, Liu T, Parlow AF, Perry G, Smith MA, Martins RN, Atwood CS (2004) Luteinizing hormone, a reproductive regulator that modulates the processing of amyloid-beta precursor protein and amyloidbeta deposition. J Biol Chem 279, 20539-20545. Casadesus G, Webber KM, Atwood CS, Pappolla MA, Perry G, Bowen RL, Smith MA (2006) Luteinizing hormone modulates cognition and amyloid-beta deposition in Alzheimer APP transgenic mice. Biochim Biophys Acta 1762, 447-452. Casadesus G, Zhu X, Atwood CS, Webber KM, Perry G, Bowen RL, Smith MA (2004) Beyond estrogen: Targeting gonadotropin hormones in the treatment of Alzheimer’s disease. Curr Drug Targets CNS Neurol Disord 3, 281-285. Casadesus G, Atwood CS, Zhu X, Hartzler AW, Webber KM, Perry G, Bowen RL, Smith MA (2005) Evidence for the role of gonadotropin hormones in the development of Alzheimer disease. Cell Mol Life Sci 62, 293-298. Meethal SV, Smith MA, Bowen RL, Atwood CS (2005) The gonadotropin connection in Alzheimer’s disease. Endocrine 26, 317-326. 58 W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [96] [97] [98] [99] [100] [101] [102] [103] [104] [105] [106] [107] [108] Wilson AC, Meethal SV, Bowen RL, Atwood CS (2007) Leuprolide acetate: A drug of diverse clinical applications. Expert Opin Investig Drugs 16, 1851-1863. Baker LD, Barsness SM, Borson S, Merriam GR, Friedman SD, Craft S, Vitiello MV (2012) Effects of growth hormone-releasing hormone on cognitive function in adults with mild cognitive impairment and healthy older adults: Results of a controlled trial. Arch Neurol 69, 14201429. Mori M, Iriuchijima T, Yamada M, Murakami M, Kobayashi S (1991) A novel TRH analog, YM14673, stimulates intracellular signaling systems in the brain more potently than predicted by its pituitary actions. Res Commun Chem Pathol Pharmacol 71, 17-26. Yamamoto M, Ozawa Y, Takeuchi H (1993) Effects of YM14673, a new thyrotropin-releasing hormone analogue, on impaired learning of passive avoidance in mice. Arch Int Pharmacodyn Ther 321, 5-13. Matsushita M, Yonemori F, Hamada A, Toide K, Iwata K (1995) Effect of JTP-2942, a novel thyrotropin-releasing hormone analogue, on pentobarbital-induced anesthesia in rats. Eur J Pharmacol 276, 177-182. Katsumata T, Katayama Y, Yonemori H, Muramatsu H, Otori T, Nishiyama Y, Yamada H, Nakamura H, Terashi A (2001) Delayed administration of JTP-2942, a novel thyrotropin-releasing hormone analogue, improves cerebral blood flow and metabolism in rat postischaemic brain. Clin Exp Pharmacol Physiol 28, 48-54. 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 novel thyrotropin-releasing hormone analogue, on neurological outcome, local cerebral blood flow and glucose utilization in a rat focal cerebral ischemia. Brain Res 901, 62-70. Itoh Y, Ogasawara T, Mushiroi T, Yamazaki A, Ukai Y, Kimura K (1994) Effect of NS-3, a thyrotropin-releasing hormone analog, on in vivo acetylcholine release in rat brain: Regional differences and its sites of action. J Pharmacol Exp Ther 271, 884-890. Bristow LJ, Bennett GW (1989) Effect of chronic intraaccumbens administration of the TRH analogue CG3509 on histamine-induced behaviour in the rat. Br J Pharmacol 97, 745-752. Fone KC, Johnson JV, Bennett GW, Marsden CA (1989) Involvement of 5-HT2 receptors in the behaviours produced by intrathecal administration of selected 5-HT agonists and the TRH analogue (CG 3509) to rats. Br J Pharmacol 96, 599-608. Fone KC, Johnson JV, Marsden CA, Bennett GW (1989) Comparative behavioural and biochemical effects of repeated intrathecal administration of thyrotrophinreleasing hormone (TRH) or two analogues of TRH in adult rats. Neuropharmacology 28, 867-875. Parnetti L, Ambrosoli L, Abate G, Azzini C, Balestreri R, Bartorelli L, Bordin A, Crepaldi G, Cristianini G, Cucinotta D, et al. (1995) Posatirelin for the treatment of late-onset Alzheimer’s disease: A double-blind multicentre study vs citicoline and ascorbic acid. Acta Neurol Scand 92, 135-140. Parnetti L, Ambrosoli L, Agliati G, Caratozzolo P, Fossati L, Frattola L, Martucci N, Murri L, Nappi G, Puca FM, Poli A, Girardello R, Senin U (1996) Posatirelin in the treatment of vascular dementia: A double-blind multicentre study vs placebo. Acta Neurol Scand 93, 456-463. [109] [110] [111] [112] [113] [114] [115] [116] [117] [118] [119] [120] [121] [122] Gasbarrini G, Stefanini G, Addolorato G, Foschi F, Ricci C, Bertolotti P, Voltolini G, Bonavita E, Bertoncelli R, Renzi G, Bianchini G, Bonaiuto S, Giannandrea E, Cavassini G, Mazzini V, Chioma V, Marzara G, D’Addetta G, Totaro G, Dalmonte E, Tassini D, Giungi F, De NC, Di FG, Tessitore A, Guadagnino M, Tessitore E, Spina P, Luppi M, Bignamini A, Peracino L, Fiorentino M, Beun-Garbe D, Poli A, Ambrosoli L, Girardello R (1998) Posatirelin for the treatment of degenerative and vascular dementia: Results of explanatory and pragmatic efficacy analyses. Arch Gerontol Geriatr 26, 33-47. Suzuki T, Fujimoto K, Oohata H, Kawashima K (1989) Effects of TRH and DN-1417 on high potassium-evoked acetylcholine release from rat basal forebrain slices determined directly by radioimmunoassay. Gen Pharmacol 20, 239-242. Ballard TM, Hunter AJ, Bennett GW (1996) Effect of a thyrotrophin-releasing hormone analogue, RX77368, on AMPA-induced septal-hippocampal lesioned rats in an operant delayed non-matching to position test. Psychopharmacology (Berl) 127, 265-275. Itkin A, Dupres V, Dufrene YF, Bechinger B, Ruysschaert JM, Raussens V (2011) Calcium ions promote formation of amyloid beta-peptide (1-40) oligomers causally implicated in neuronal toxicity of Alzheimer’s disease. PLoS One 6, e18250. Kawahara M, Ohtsuka I, Yokoyama S, Kato-Negishi M, Sadakane Y (2011) Membrane incorporation, channel formation, and disruption of calcium homeostasis by Alzheimer’s beta-amyloid protein. Int J Alzheimers Dis 2011, 304583. Mezler M, Barghorn S, Schoemaker H, Gross G, Nimmrich V (2012) A beta-amyloid oligomer directly modulates P/Q-type calcium currents in Xenopus oocytes. Br J Pharmacol 165, 1572-1583. Chakroborty S, Kim J, Schneider C, Jacobson C, Molgo J, Stutzmann GE (2012) Early presynaptic and postsynaptic calcium signaling abnormalities mask underlying synaptic depression in presymptomatic Alzheimer’s disease mice. J Neurosci 32, 8341-8353. Woods NK, Padmanabhan J (2012) Neuronal calcium signaling and Alzheimer’s disease. Adv Exp Med Biol 740, 1193-1217. Lim YA, Giese M, Shepherd C, Halliday G, Kobayashi M, Takamatsu K, Staufenbiel M, Eckert A, Gotz J (2012) Role of hippocalcin in mediating Abeta toxicity. Biochim Biophys Acta 1822, 1247-1257. Chakroborty S, Stutzmann GE (2011) Early calcium dysregulation in Alzheimer’s disease: Setting the stage for synaptic dysfunction. Sci China Life Sci 54, 752-762. Corona C, Pensalfini A, Frazzini V, Sensi SL (2011) New therapeutic targets in Alzheimer’s disease: Brain deregulation of calcium and zinc. Cell Death Dis 2, e176. Fedrizzi L, Carafoli E (2011) Ca2+ dysfunction in neurodegenerative disorders: Alzheimer’s disease. Biofactors 37, 189-196. Yagami T, Kohma H, Yamamoto Y (2012) L-type voltagedependent calcium channels as therapeutic targets for neurodegenerative diseases. Curr Med Chem 19, 48164827. Bachmeier C, Beaulieu-Abdelahad D, Mullan M, Paris D (2011) Selective dihydropyiridine compounds facilitate the clearance of beta-amyloid across the blood-brain barrier. Eur J Pharmacol 659, 124-129. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [123] [124] [125] [126] [127] [128] [129] [130] [131] [132] [133] [134] 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 antihypertensive dihydropyridines lower Abeta accumulation by targeting both the production and the clearance of Abeta across the blood-brain barrier. Mol Med 17, 149-162. Iwasaki K, Mishima K, Egashira N, Al-Khatib IH, Ishibashi D, Irie K, Kobayashi H, Egawa T, Fujiwara M (2003) Effect of nilvadipine on the cerebral ischemiainduced impairment of spatial memory and hippocampal apoptosis in rats. J Pharmacol Sci 93, 188-196. Iwasaki K, Egashira N, Takagaki Y, Yoshimitsu Y, HatipAl-Khatib I, Mishima K, Fujiwara M (2007) Nilvadipine prevents the impairment of spatial memory induced by cerebral ischemia combined with beta-amyloid in rats. Biol Pharm Bull 30, 698-701. Paris D, Quadros A, Humphrey J, Patel N, Crescentini R, Crawford F, Mullan M (2004) Nilvadipine antagonizes both Abeta vasoactivity in isolated arteries, and the reduced cerebral blood flow in APPsw transgenic mice. Brain Res 999, 53-61. Kennelly SP, Abdullah L, Paris D, Parish J, Mathura V, Mullan M, Crawford F, Lawlor BA, Kenny RA (2011) Demonstration of safety in Alzheimer’s patients for intervention with an anti-hypertensive drug Nilvadipine: Results from a 6-week open label study. Int J Geriatr Psychiatry 26, 1038-1045. Kennelly S, Abdullah L, Kenny RA, Mathura V, Luis CA, Mouzon B, Crawford F, Mullan M, Lawlor B (2012) Apolipoprotein E genotype-specific short-term cognitive benefits of treatment with the antihypertensive nilvadipine in Alzheimer’s patients–an open-label trial. Int J Geriatr Psychiatry 27, 415-422. Hanyu H, Hirao K, Shimizu S, Iwamoto T, Koizumi K, Abe K (2007) Favourable effects of nilvadipine on cognitive function and regional cerebral blood flow on SPECT in hypertensive patients with mild cognitive impairment. Nucl Med Commun 28, 281-287. Hanyu H, Hirao K, Shimizu S, Sato T, Kiuchi A, Iwamoto T (2007) Nilvadipine prevents cognitive decline of patients with mild cognitive impairment. Int J Geriatr Psychiatry 22, 1264-1266. Matsuda H, Araki N, Kuji I, Ohkubo T, Imabayashi E, Shimazu K (2008) Effect of nilvadipine on regional cerebral blood flow in a patient with early Alzheimer disease. Clin Nucl Med 33, 34-35. Moriguchi S, Shioda N, Yamamoto Y, Tagashira H, Fukunaga K (2012) The T-type voltage-gated calcium channel as a molecular target of the novel cognitive enhancer ST101: Enhancement of long-term potentiation and CaMKII autophosphorylation in rat cortical slices. J Neurochem 121, 44-53. Yamaguchi Y, Miyashita H, Tsunekawa H, Mouri A, Kim HC, Saito K, Matsuno T, Kawashima S, Nabeshima T (2006) Effects of a novel cognitive enhancer, spiro[imidazo-[1,2-a]pyridine-3,2-indan]2(3H)-one (ZSET1446), on learning impairments induced by amyloid-beta1-40 in the rat. J Pharmacol Exp Ther 317, 1079-1087. Shioda N, Yamamoto Y, Han F, Moriguchi S, Yamaguchi Y, Hino M, Fukunaga K (2010) A novel cognitive enhancer, ZSET1446/ST101, promotes hippocampal neurogenesis and ameliorates depressive behavior in olfactory bulbectomized mice. J Pharmacol Exp Ther 333, 43-50. [135] [136] [137] [138] [139] [140] [141] [142] [143] [144] [145] [146] [147] [148] [149] [150] 59 Anekonda TS, Quinn JF, Harris C, Frahler K, Wadsworth TL, Woltjer RL (2011) L-type voltage-gated calcium channel blockade with isradipine as a therapeutic strategy for Alzheimer’s disease. Neurobiol Dis 41, 62-70. Anekonda TS, Quinn JF (2011) Calcium channel blocking as a therapeutic strategy for Alzheimer’s disease: The case for isradipine. Biochim Biophys Acta 1812, 15841590. Copenhaver PF, Anekonda TS, Musashe D, Robinson KM, Ramaker JM, Swanson TL, Wadsworth TL, Kretzschmar D, Woltjer RL, Quinn JF (2011) A translational continuum of model systems for evaluating treatment strategies in Alzheimer’s disease: Isradipine as a candidate drug. Dis Model Mech 4, 634-648. Saletu B, Darragh A, Salmon P, Coen R (1989) EEG brain mapping in evaluating the time-course of the central action of DUP 996–a new acetylcholine releasing drug. Br J Clin Pharmacol 28, 1-16. van Dyck CH, Lin CH, Robinson R, Cellar J, Smith EO, Nelson JC, Arnsten AF, Hoffer PB (1997) The acetylcholine releaser linopirdine increases parietal regional cerebral blood flow in Alzheimer’s disease. Psychopharmacology (Berl) 132, 217-226. Borjesson A, Karlsson T, Adolfsson R, Ronnlund M, Nilsson L (1999) Linopirdine (DUP 996): Cholinergic treatment of older adults using successive and nonsuccessive tests. Neuropsychobiology 40, 78-85. Rose GM, Ong VS, Woodruff-Pak DS (2007) Efficacy of MEM 1003, a novel calcium channel blocker, in delay and trace eyeblink conditioning in older rabbits. Neurobiol Aging 28, 766-773. 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 functions and electroencephalography in hypertensive patients. Fundam Clin Pharmacol 16, 527-535. Schwartz BL, Fay-McCarthy M, Kendrick K, Rosse RB, Deutsch SI (1997) Effects of nifedipine, a calcium channel antagonist, on cognitive function in schizophrenic patients with tardive dyskinesia. Clin Neuropharmacol 20, 364370. 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 age dementias. Prog Neuropsychopharmacol Biol Psychiatry 14, 525-551. Izquierdo I (1990) Nimodipine and the recovery of memory. Trends Pharmacol Sci 11, 309-310. Tedeschi D (1991) Calcium regulation in brain aging by nimodipine: A multicenterctrial in Italy. Curr Ther Res 50, 553-563. de Jonge MC, Traber J (1993) Nimodipine: Cognition, aging, and degeneration. Clin Neuropharmacol 16(Suppl 1), S25-S30. Fischhof PK (1993) Divergent neuroprotective effects of nimodipine in PDD and MID provide indirect evidence of disturbances in Ca2+ homeostasis in dementia. Methods Find Exp Clin Pharmacol 15, 549-555. Parnetti L, Senin U, Carosi M, Baasch H (1993) Mental deterioration in old age: Results of two multicenter, clinical trials with nimodipine. The Nimodipine Study Group. Clin Ther 15, 394-406. Bernhardt T, Kuebler J, Erzigkeit H (1995) Impairment of cerebral function in old age: Nimodipine in general practice. Eur J Clin Res 7, 205-215. 60 [151] [152] [153] [154] [155] [156] [157] [158] [159] [160] [161] [162] [163] [164] [165] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Fritze J, Walden J (1995) Clinical findings with nimodipine in dementia: Test of the calcium hypothesis. J Neural Transm Suppl 46, 439-453. Sze KH, Sim TC, Wong E, Cheng S, Woo J (1998) Effect of nimodipine on memory after cerebral infarction. Acta Neurol Scand 97, 386-392. Forette F, Seux ML, Staessen JA, Thijs L, Birkenhager WH, Babarskiene MR, Babeanu S, Bossini A, GilExtremera B, Girerd X, Laks T, Lilov E, Moisseyev V, TuomilehtoJ, Vanhanen H, Webster J, Yodfat Y, Fagard R (1998) Prevention of dementia in randomised doubleblind placebo-controlled Systolic Hypertension in Europe (Syst-Eur) trial. Lancet 352, 1347-1351. 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, Webster J, Yodfat Y, Birkenhager WH (2002) The prevention of dementia with antihypertensive treatment: New evidence from the Systolic Hypertension in Europe (Syst-Eur) study. Arch Intern Med 162, 2046-2052. Valero T, del BL, Egea J, Canas N, Martinez A, Garcia AG, Villarroya M, Lopez MG (2009) NP04634 prevents cell damage caused by calcium overload and mitochondrial disruption in bovine chromaffin cells. Eur J Pharmacol 607, 47-53. 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 cerebral ischaemia in vivo. Eur J Pharmacol 332, 121-131. Paroczai M, Kiss B, Karpati E (1998) Effect of RGH-2716 on learning and memory deficits of young and aged rats in water-labyrinth. Brain Res Bull 45, 475-488. Fukumoto H, Kakihana M, Kaisho Y, Suno M (1997) The novel compound TDN-345 induces synthesis/secretion of nerve growth factor in C6-10A glioma cells. Brain Res 774, 87-93. Nakayama T, Nagisa Y, Imamoto T, Nagai Y (1997) Beneficial effects of TDN-345, a novel Ca2+ antagonist, on ischemic brain injury and cerebral glucose metabolism in experimental animal models with cerebrovascular lesions. Brain Res 762, 203-210. Bourinet E, Stotz SC, Spaetgens RL, Dayanithi G, Lemos J, Nargeot J, Zamponi GW (2001) Interaction of SNX482 with domains III and IV inhibits activation gating of alpha(1E) (Ca(V)2.3) calcium channels. Biophys J 81, 79-88. Kohlmeier KA, Leonard CS (2006) Transmitter modulation of spike-evoked calcium transients in arousal related neurons: Muscarinic inhibition of SNX-482-sensitive calcium influx. Eur J Neurosci 23, 1151-1162. Matthews EA, Bee LA, Stephens GJ, Dickenson AH (2007) The Cav2.3 calcium channel antagonist SNX-482 reduces dorsal horn neuronal responses in a rat model of chronic neuropathic pain. Eur J Neurosci 25, 35613569. Miyake N, Wakamori M, Akaike N (1992) A new type of Ca2+ channel blocker, NC-1100, inhibits the low- and high-threshold Ca2+ currents in the rat CNS neurons. Brain Res 598, 215-220. Miyake N, Fujita R, Ishikawa M, Takayanagi M, Takayanagi Y, Sasaki K (2000) Reversal of multidrug resistance in human leukemia K562 by tamolarizine, a novel calcium antagonist. Jpn J Pharmacol 82, 265-268. Tamura R, Nakada Y, Nishijo H, Miyake N, Ono T (2000) Ameliorative effects of tamolarizine on place learning [166] [167] [168] [169] [170] [171] [172] [173] [174] [175] [176] [177] [178] [179] [180] [181] impairment induced by transient forebrain ischemia in rats. Brain Res 853, 81-92. Zaczek R, Chorvat RJ, Saye JA, Pierdomenico ME, Maciag CM, Logue AR, Fisher BN, Rominger DH, Earl RA (1998) Two new potent neurotransmitter release enhancers, 10,10-bis(4-pyridinylmethyl)-9(10H)anthracenone and 10,10-bis(2-fluoro-4-pyridinylmethyl)9(10H)-anthracenone: Comparison to linopirdine. J Pharmacol Exp Ther 285, 724-730. MacVinish LJ, Guo Y, Dixon AK, Murrell-Lagnado RD, Cuthbert AW (2001) Xe991 reveals differences in K(+) channels regulating chloride secretion in murine airway and colonic epithelium. Mol Pharmacol 60, 753760. Schindowski K, Belarbi K, Buee L (2008) Neurotrophic factors in Alzheimer’s disease: Role of axonal transport. Genes Brain Behav 7(Suppl 1), 43-56. Saragovi HU, Hamel E, Di PA (2009) A neurotrophic rationale for the therapy of neurodegenerative disorders. Curr Alzheimer Res 6, 419-423. Lu Y, Christian K, Lu B (2008) BDNF: A key regulator for protein synthesis-dependent LTP and long-term memory? Neurobiol Learn Mem 89, 312-323. Nagahara AH, Tuszynski MH (2011) Potential therapeutic uses of BDNF in neurological and psychiatric disorders. Nat Rev Drug Discov 10, 209-219. Fumagalli F, Racagni G, Riva MA (2006) The expanding role of BDNF: A therapeutic target for Alzheimer’s disease? Pharmacogenomics J 6, 8-15. Zhang F, Kang Z, Li W, Xiao Z, Zhou X (2012) Roles of brain-derived neurotrophic factor/tropomyosin-related kinase B (BDNF/TrkB) signalling in Alzheimer’s disease. J Clin Neurosci 19, 946-949. Castello NA, Green KN, Laferla FM (2012) Genetic knockdown of brain-derived neurotrophic factor in 3xTgAD mice does not alter Abeta or tau pathology. PLoS One 7, e39566. Capsoni S, Cattaneo A (2006) On the molecular basis linking nerve growth factor (NGF) to Alzheimer’s disease. Cell Mol Neurobiol 26, 619-633. Capsoni S, Tiveron C, Amato G, Vignone D, Cattaneo A (2010) Peripheral neutralization of nerve growth factor induces immunosympathectomy and central neurodegeneration in transgenic mice. J Alzheimers Dis 20, 527546. Capsoni S, Brandi R, Arisi I, D’Onofrio M, Cattaneo A (2011) A dual mechanism linking NGF/proNGF imbalance and early inflammation to Alzheimer’s disease neurodegeneration in the AD11 anti-NGF mouse model. CNS Neurol Disord Drug Targets 10, 635-647. Cuello AC (2012) Gangliosides, NGF, brain aging and disease: A mini-review with personal reflections. Neurochem Res 37, 1256-1260. Cuello AC, Bruno MA, Bell KF (2007) NGF-cholinergic dependency in brain aging, MCI and Alzheimer’s disease. Curr Alzheimer Res 4, 351-358. Cuello AC, Bruno MA, Allard S, Leon W, Iulita MF (2010) Cholinergic involvement in Alzheimer’s disease. A link with NGF maturation and degradation. J Mol Neurosci 40, 230-235. Cuello AC, Ferretti MT, Iulita MF (2012) Preplaque (’preclinical’) Abeta-induced inflammation and nerve growth factor deregulation in transgenic models of Alzheimer’s disease-like amyloid pathology. Neurodegener Dis 10, 104-107. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [182] [183] [184] [185] [186] [187] [188] [189] [190] [191] [192] [193] [194] Jonhagen ME (2000) Nerve growth factor treatment in dementia. Alzheimer Dis Assoc Disord 14(Suppl 1), S31-S38. Cattaneo A, Calissano P (2012) Nerve growth factor and Alzheimer’s disease: New facts for an old hypothesis. Mol Neurobiol 46, 588-604. Wang SH, Liao XM, Liu D, Hu J, Yin YY, Wang JZ, Zhu LQ (2012) NGF promotes long-term memory formation by activating poly(ADP-ribose)polymerase-1. Neuropharmacology 63, 1085-1092. Bishop KM, Hofer EK, Mehta A, Ramirez A, Sun L, Tuszynski M, Bartus RT (2008) Therapeutic potential of CERE-110 (AAV2-NGF): Targeted, stable, and sustained NGF delivery and trophic activity on rodent basal forebrain cholinergic neurons. Exp Neurol 211, 574-584. Mandel RJ (2010) CERE-110, an adeno-associated virusbased gene delivery vector expressing human nerve growth factor for the treatment of Alzheimer’s disease. Curr Opin Mol Ther 12, 240-247. Jain P, Li R, Lama T, Saragovi HU, Cumberlidge G, Meerovitch K (2011) An NGF mimetic, MIM-D3, stimulates conjunctival cell glycoconjugate secretion and demonstrates therapeutic efficacy in a rat model of dry eye. Exp Eye Res 93, 503-512. Visanji NP, Orsi A, Johnston TH, Howson PA, Dixon K, Callizot N, Brotchie JM, Rees DD (2008) PYM50028, a novel, orally active, nonpeptide neurotrophic factor inducer, prevents and reverses neuronal damage induced by MPP+ in mesencephalic neurons and by MPTP in a mouse model of Parkinson’s disease. FASEB J 22, 24882497. Fukushima T, Nakamura A, Iwakami N, Nakada Y, Hattori H, Hoki S, Yamaguchi H, Nakagawa M, Terashima N, Narita H (2011) T-817MA, a neuroprotective agent, attenuates the motor and cognitive impairments associated with neuronal degeneration in P301L tau transgenic mice. Biochem Biophys Res Commun 407, 730-734. Nguyen PT, Kimura T, Ho SA, Tran AH, Ono T, Nishijo H (2007) Ameliorative effects of a neuroprotective agent, T817MA, on place learning deficits induced by continuous infusion of amyloid-beta peptide (1-40) in rats. Hippocampus 17, 443-455. Kimura T, Hong Nguyen PT, Ho SA, Tran AH, Ono T, Nishijo H (2009) T-817MA, a neurotrophic agent, ameliorates the deficits in adult neurogenesis and spatial memory in rats infused i.c.v. with amyloid-beta peptide. Br J Pharmacol 157, 451-463. Hirata K, Yamaguchi H, Takamura Y, Takagi A, Fukushima T, Iwakami N, Saitoh A, Nakagawa M, Yamada T (2005) A novel neurotrophic agent, T-817MA [1-{3[2-(1-benzothiophen-5-yl) ethoxy] propyl}-3-azetidinol maleate], attenuates amyloid-beta-induced neurotoxicity and promotes neurite outgrowth in rat cultured central nervous system neurons. J Pharmacol Exp Ther 314, 252-259. Wahlberg LU, Lind G, Almqvist PM, Kusk P, Tornoe J, Juliusson B, Soderman M, Sellden E, Seiger A, Eriksdotter-Jonhagen M, Linderoth B (2012) Targeted delivery of nerve growth factor via encapsulated cell biodelivery in Alzheimer disease: A technology platform for restorative neurosurgery. J Neurosurg 117, 340-347. Voutilainen MH, Back S, Porsti E, Toppinen L, Lindgren L, Lindholm P, Peranen J, Saarma M, Tuominen RK (2009) Mesencephalic astrocyte-derived neurotrophic factor is neurorestorative in rat model of Parkinson’s disease. J Neurosci 29, 9651-9659. [195] [196] [197] [198] [199] [200] [201] [202] [203] [204] [205] [206] [207] [208] [209] 61 Glembotski CC, Thuerauf DJ, Huang C, Vekich JA, Gottlieb RA, Doroudgar S (2012) Mesencephalic astrocyte-derived neurotrophic factor protects the heart from ischemic damage and is selectively secreted upon sarco/endoplasmic reticulum calcium depletion. J Biol Chem 287, 25893-25904. Devi L, Ohno M (2012) 7,8-dihydroxyflavone, a smallmolecule TrkB agonist, reverses memory deficits and BACE1 elevation in a mouse model of Alzheimer’s disease. Neuropsychopharmacology 37, 434-444. Jang SW, Liu X, Yepes M, Shepherd KR, Miller GW, Liu Y, Wilson WD, Xiao G, Blanchi B, Sun YE, Ye K (2010) A selective TrkB agonist with potent neurotrophic activities by 7,8-dihydroxyflavone. Proc Natl Acad Sci U S A 107, 2687-2692. Johnson RA, Lam M, Punzo AM, Li H, Lin BR, Ye K, Mitchell GS, Chang Q (2012) 7,8-dihydroxyflavone exhibits therapeutic efficacy in a mouse model of Rett syndrome. J Appl Physiol 112, 704-710. 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) A synthetic 7,8-dihydroxyflavone derivative promotes neurogenesis and exhibits potent antidepressant effect. J Med Chem 53, 8274-8286. Jang SW, Okada M, Sayeed I, Xiao G, Stein D, Jin P, Ye K (2007) Gambogic amide, a selective agonist for TrkA receptor that possesses robust neurotrophic activity, prevents neuronal cell death. Proc Natl Acad Sci U S A 104, 16329-16334. Jang SW, Liu X, Chan CB, France SA, Sayeed I, Tang W, Lin X, Xiao G, Andero R, Chang Q, Ressler KJ, Ye K (2010) Deoxygedunin, a natural product with potent neurotrophic activity in mice. PLoS One 5, e11528. Nitta A, Ito M, Fukumitsu H, Ohmiya M, Ito H, Sometani A, Nomoto H, Furukawa Y, Furukawa S (1999) 4-methylcatechol increases brain-derived neurotrophic factor content and mRNA expression in cultured brain cells and in rat brain in vivo. J Pharmacol Exp Ther 291, 1276-1283. Sun MK, Alkon DL (2008) Effects of 4-methylcatechol on spatial memory and depression. Neuroreport 19, 355-359. Labie C, Canolle B, Chatelin S, Lafon C, Fournier J (2006) Effects of paliroden (SR57667B) and xaliproden on adult brain neurogenesis. Curr Alzheimer Res 3, 35-36. Douillet P, Orgogozo JM (2009) What we have learned from the Xaliproden Sanofi-aventis trials. J Nutr Health Aging 13, 365-366. Porzner M, Muller T, Seufferlein T (2009) SR 57746A/ xaliproden, a non-peptide neurotrophic compound: Prospects and constraints for the treatment of nervous system diseases. Expert Opin Investig Drugs 18, 17651772. Mori T, Town T, TanJ, Yada N, Horikoshi Y, Yamamoto J, Shimoda T, Kamanaka Y, Tateishi N, Asano T (2006) Arundic Acid ameliorates cerebral amyloidosis and gliosis in Alzheimer transgenic mice. J Pharmacol Exp Ther 318, 571-578. Pettigrew LC, Kasner SE, Albers GW, Gorman M, Grotta JC, Sherman DG, Funakoshi Y, Ishibashi H (2006) Safety and tolerability of arundic acid in acute ischemic stroke. J Neurol Sci 251, 50-56. Pettigrew LC, Kasner SE, Gorman M, Atkinson RP, Funakoshi Y, Ishibashi H (2006) Effect of arundic acid on serum S-100beta in ischemic stroke. J Neurol Sci 251, 57-61. 62 [210] [211] [212] [213] [214] [215] [216] [217] [218] [219] [220] [221] [222] [223] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes 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 of some physicochemical properties of arundic acid, (R)-(-)2-propyloctanonic acid, by complexation with hydrophilic cyclodextrins. Int J Pharm 413, 63-72. Glasky AJ, Melchior CL, Pirzadeh B, Heydari N, Ritzmann RF (1994) Effect of AIT-082, a purine analog, on working memory in normal and aged mice. Pharmacol Biochem Behav 47, 325-329. Glasky AJ, Glasky MS, Ritzmann RF, Rathbone MP (1997) AIT-082, a novel purine derivative with neuroregenerative properties. Expert Opin Investig Drugs 6, 1413-1417. Middlemiss PJ, Glasky AJ, Rathbone MP, Werstuik E, Hindley S, Gysbers J (1995) AIT-082, a unique purine derivative, enhances nerve growth factor mediated neurite outgrowth from PC12 cells. Neurosci Lett 199, 131-134. 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 regenerative agent in stroke and central nervous system injury. Expert Opin Investig Drugs 8, 1255-1262. Lahiri DK, Ge YW, Farlow MR (2000) Effect of a memoryenhancing drug, AIT-082, on the level of synaptophysin. Ann N Y Acad Sci 903, 387-393. Taylor EM, Yan R, Hauptmann N, Maher TJ, Djahandideh D, Glasky AJ (2000) AIT-082, a cognitive enhancer, is transported into brain by a nonsaturable influx mechanism and out of brain by a saturable efflux mechanism. J Pharmacol Exp Ther 293, 813-821. 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-carnitine arginyl amide on the human neuronal line HCN-1A. Int J Dev Neurosci 10, 361-373. 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-Lcarnitine arginine amide. Neurochem Res 20, 1-9. Scorziello A, Meucci O, Calvani M, Schettini G (1997) Acetyl-L-carnitine arginine amide prevents beta 25–35-induced neurotoxicity in cerebellar granule cells. Neurochem Res 22, 257-265. Ono S, Kitamura K, Maekawa M, Hirata K, Ano M, Ukai W, Yamafuji T, Narita H (1993) Protective effect of R(-)-1-(benzo[b]thiophen-5-yl)- 2-[2-(N,Ndiethylamino)ethoxy]ethanol hydrochloride (T-588), a novel cerebral activator, against experimental cerebral anoxia. Jpn J Pharmacol 62, 81-86. Miyazaki H, Murayama T, Ono S, Narita H, Nomura Y (1997) Effects of R(-)-1-(benzo[b]thiophen-5-yl)-2-[2N,N-diethylamino)ethoxy]ethan ol hydrochloride (T-588), a novel cognitive enhancer, on noradrenaline release in rat cerebral cortical slices. Biochem Pharmacol 53, 12631269. 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 cortical slices. Jpn J Pharmacol 77, 155-160. Iwasaki Y, Ikeda K, Ichikawa Y, Igarashi O, Kinoshita M, Marubuchi S, Ono S (2002) T-588 enhances neurite outgrowth and choline acetyltransferase in cultured rat spinal ventral horn neurons. Neurochem Res 27, 225-228. [224] [225] [226] [227] [228] [229] [230] [231] [232] [233] [234] [235] [236] [237] [238] [239] [240] Iwasaki Y, Ichikawa Y, Igarasi O, Aoyagi J, Konno S, Ikeda K, Iguchi H, Kawabe S, Marubuchi S, Ono S (2003) T-588 protects motor neuron death against glutamate-induced neurotoxicity. Neurochem Res 28, 1829-1832. 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 following axotomy. Neurochem Res 29, 403-406. Yamamuro A, Ago Y, Maeda S, Sakai Y, Baba A, Matsuda T (2003) Protective effect of T-588 on toxic damage by serum deprivation and amyloid-beta protein in cultured neurons. J Pharmacol Sci 92, 153-156. Phuagphong P, Fukushima T, Hatanaka R, Tanaka K, Baba A, Matsuda T (2004) T-588, a cognitive enhancer, protects against sodium nitroprusside-induced toxicity in cultured astrocytes. J Pharmacol Sci 95, 135-138. Advokat C (2010) What are the cognitive effects of stimulant medications? Emphasis on adults with attention-deficit/hyperactivity disorder (ADHD). Neurosci Biobehav Rev 34, 1256-1266. Berridge CW, Devilbiss DM (2011) Psychostimulants as cognitive enhancers: The prefrontal cortex, catecholamines, and attention-deficit/hyperactivity disorder. Biol Psychiatry 69, e101-e111. Berridge CW, Shumsky JS, Andrzejewski ME, McGaughy JA, Spencer RC, Devilbiss DM, Waterhouse BD (2012) Differential sensitivity to psychostimulants across prefrontal cognitive tasks: Differential involvement of noradrenergic alpha(1)- and alpha(2)-receptors. Biol Psychiatry 71, 467-473. Dolder CR, Davis LN, McKinsey J (2010) Use of psychostimulants in patients with dementia. Ann Pharmacother 44, 1624-1632. Arnsten AF, Pliszka SR (2011) Catecholamine influences on prefrontal cortical function: Relevance to treatment of attention deficit/hyperactivity disorder and related disorders. Pharmacol Biochem Behav 99, 211-216. Bidwell LC, McClernon FJ, Kollins SH (2011) Cognitive enhancers for the treatment of ADHD. Pharmacol Biochem Behav 99, 262-274. Husain M, Mehta MA (2011) Cognitive enhancement by drugs in health and disease. Trends Cogn Sci 15, 28-36. Levin ED, Bushnell PJ, Rezvani AH (2011) Attentionmodulating effects of cognitive enhancers. Pharmacol Biochem Behav 99, 146-154. Sahakian BJ, Morein-Zamir S (2011) Neuroethical issues in cognitive enhancement. J Psychopharmacol 25, 197204. Ragan CI, Bard I, Singh I (2012) What should we do about student use of cognitive enhancers? An analysis of current evidence. Neuropharmacology 64, 588-595. Wolkenberg SE, Sur C (2010) Recent progress in the discovery of non-sarcosine based GlyT1 inhibitors. Curr Top Med Chem 10, 170-186. Volkow ND, Wang GJ, Fowler JS, Ding YS (2005) Imaging the effects of methylphenidate on brain dopamine: New model on its therapeutic actions for attention-deficit/hyperactivity disorder. Biol Psychiatry 57, 1410-1415. Berridge CW, Devilbiss DM, Andrzejewski ME, Arnsten AF, Kelley AE, Schmeichel B, Hamilton C, Spencer RC (2006) Methylphenidate preferentially increases catecholamine neurotransmission within the prefrontal cortex at low doses that enhance cognitive function. Biol Psychiatry 60, 1111-1120. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [241] [242] [243] [244] [245] [246] [247] [248] [249] [250] [251] [252] [253] [254] [255] Devilbiss DM, Berridge CW (2008) Cognition-enhancing doses of methylphenidate preferentially increase prefrontal cortex neuronal responsiveness. Biol Psychiatry 64, 626-635. Volkow ND, Fowler JS, Wang GJ, Telang F, Logan J, Wong C, Ma J, Pradhan K, Benveniste H, Swanson JM (2008) Methylphenidate decreased the amount of glucose needed by the brain to perform a cognitive task. PLoS One 3, e2017. Repantis D, Schlattmann P, Laisney O, Heuser I (2010) Modafinil and methylphenidate for neuroenhancement in healthy individuals: A systematic review. Pharmacol Res 62, 187-206. Prashad M, Kim HY, Lu Y, Liu Y, Har D, Repic O, Blacklock TJ, Giannousis P (1999) The First Enantioselective Synthesis of (2R,2’R)-threo-(+)-Methylphenidate Hydrochloride. J Org Chem 64, 1750-1753. Spencer TJ, Bonab AA, Dougherty DD, Mirto T, Martin J, Clarke A, Fischman AJ (2012) Understanding the central pharmacokinetics of spheroidal oral drug absorption system (SODAS) dexmethylphenidate: A positron emission tomography study of dopamine transporter receptor occupancy measured with C-11 altropane. J Clin Psychiatry 73, 346-352. Schmitt KC, Reith ME (2011) The atypical stimulant and nootropic modafinil interacts with the dopamine transporter in a different manner than classical cocaine-like inhibitors. PLoS One 6, e25790. Turner DC, Clark L, Dowson J, Robbins TW, Sahakian BJ (2004) Modafinil improves cognition and response inhibition in adult attention-deficit/hyperactivity disorder. Biol Psychiatry 55, 1031-1040. Turner DC, Clark L, Pomarol-Clotet E, McKenna P, Robbins TW, Sahakian BJ (2004) Modafinil improves cognition and attentional set shifting in patients with chronic schizophrenia. Neuropsychopharmacology 29, 1363-1373. Scoriels L, Barnett JH, Murray GK, Cherukuru S, Fielding M, Cheng F, Lennox BR, Sahakian BJ, Jones PB (2011) Effects of modafinil on emotional processing in first episode psychosis. Biol Psychiatry 69, 457-464. Scoriels L, Barnett JH, Soma PK, Sahakian BJ, Jones PB (2012) Effects of modafinil on cognitive functions in first episode psychosis. Psychopharmacology (Berl) 220, 249258. Scoriels L, Jones PB, Sahakian BJ (2013) Modafinil effects on cognition and emotion in schizophrenia and its neurochemical modulation in the brain. Neuropharmacology 64, 168-184. Rasetti R, Mattay VS, Stankevich B, Skjei K, Blasi G, Sambataro F, rrillaga-Romany IC, Goldberg TE, Callicott JH, Apud JA, Weinberger DR (2010) Modulatory effects of modafinil on neural circuits regulating emotion and cognition. Neuropsychopharmacology 35, 21012109. Muller U, Rowe JB, Rittman T, Lewis C, Robbins TW, Sahakian BJ (2013) Effects of modafinil on non-verbal cognition, task enjoyment and creative thinking in healthy volunteers. Neuropharmacology 64, 490-495. Brady KT, Gray KM, Tolliver BK (2011) Cognitive enhancers in the treatment of substance use disorders: Clinical evidence. Pharmacol Biochem Behav 99, 285-294. Kalechstein AD, Mahoney JJ III, Yoon JH, Bennett R, De La GR (2013) Modafinil, but not escitalopram, improves working memory and sustained attention in long-term, [256] [257] [258] [259] [260] [261] [262] [263] [264] [265] [266] [267] 63 high-dose cocaine users. Neuropharmacology 64, 472478. Sutherland SM, Adler LA, Chen C, Smith MD, Feltner DE (2012) An 8-week, randomized controlled trial of atomoxetine, atomoxetine plus buspirone, or placebo in adults with ADHD. J Clin Psychiatry 73, 445-450. Gehlert DR, Schober DA, Hemrick-Luecke SK, Krushinski J, Howbert JJ, Robertson DW, Fuller RW, Wong DT (1995) Novel halogenated analogs of tomoxetine that are potent and selective inhibitors of norepinephrine uptake in brain. Neurochem Int 26, 47-52. Chamberlain SR, Del CN, Dowson J, Muller U, Clark L, Robbins TW, Sahakian BJ (2007) Atomoxetine improved response inhibition in adults with attention deficit/hyperactivity disorder. Biol Psychiatry 62, 977-984. Chamberlain SR, Hampshire A, Muller U, Rubia K, Del CN, Craig K, Regenthal R, Suckling J, Roiser JP, Grant JE, Bullmore ET, Robbins TW, Sahakian BJ (2009) Atomoxetine modulates right inferior frontal activation during inhibitory control: A pharmacological functional magnetic resonance imaging study. Biol Psychiatry 65, 550-555. Wigal SB, Wong AA, Jun A, Stehli A, SteinbergEpstein R, Lerner MA (2012) Adverse events in medication treatment-naive children with attentiondeficit/hyperactivity disorder: Results from a small, controlled trial of lisdexamfetamine dimesylate. J Child Adolesc Psychopharmacol 22, 149-156. Rothman RB, Baumann MH, Dersch CM, Romero DV, Rice KC, Carroll FI, Partilla JS (2001) Amphetamine-type central nervous system stimulants release norepinephrine more potently than they release dopamine and serotonin. Synapse 39, 32-41. Kojima T, Niigata K, Fujikura T, Tachikawa S, Nozaki Y, Kagami S, Takahashi K (1985) Syntheses of (+/-)-2-[(inden-7-yloxy)methyl]morpholine hydrochloride (YM-08054, indeloxazine hydrochloride) and its derivatives with potential cerebral-activating and antidepressive properties. Chem Pharm Bull (Tokyo) 33, 3766-3774. Yamamoto M, Ooyama M, Ozawa Y, Okada M, Tada S, Yamaguchi T, Endo H (1993) Effects of indeloxazine hydrochloride, a cerebral activator, on passive avoidance learning impaired by disruption of cholinergic transmission in rats. Neuropharmacology 32, 695-701. Yamamoto M, Takahashi K, Ohyama M, Sasamata M, Yatsugi S, Okada M, Endoh H (1994) Possible involvement of central cholinergic system in ameliorating effects of indeloxazine, a cerebral activator, on disturbance of learning behavior in rats. Prog Neuropsychopharmacol Biol Psychiatry 18, 603-613. Yamaguchi T, Ozawa Y, Suzuki M, Yamamoto M, Nakamura T, Yamaura A (1996) Indeloxazine hydrochloride improves impairment of passive avoidance performance after fluid percussion brain injury in rats. Neuropharmacology 35, 329-336. Yamaguchi T, Suzuki M, Yamamoto M (1997) Facilitation of acetylcholine release in rat frontal cortex by indeloxazine hydrochloride: Involvement of endogenous serotonin and 5-HT4 receptors. Naunyn Schmiedebergs Arch Pharmacol 356, 712-720. Yamaguchi T, Ohyama M, Suzuki M, Ozawa Y, Hatanaka K, Hidaka K, Yamamoto M (1998) Neurochemical and behavioral characterization of potential antidepressant properties of indeloxazine hydrochloride. Neuropharmacology 37, 1169-1176. 64 [268] [269] [270] [271] [272] [273] [274] [275] [276] [277] [278] [279] [280] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Kurosawa Y, Degrauw TJ, Lindquist DM, Blanco VM, Pyne-Geithman GJ, Daikoku T, Chambers JB, Benoit SC, Clark JF (2012) Cyclocreatine treatment improves cognition in mice with creatine transporter deficiency. J Clin Invest 122, 2837-2846. Sugane T, Tobe T, Hamaguchi W, Shimada I, Maeno K, Miyata J, Suzuki T, Kimizuka T, Kohara A, Morita T, Doihara H, Saita K, Aota M, Furutani M, Shimada Y, Hamada N, Sakamoto S, Tsukamoto S (2011) Synthesis and biological evaluation of 3-biphenyl-4-yl-4phenyl-4H-1,2,4-triazoles as novel glycine transporter 1 inhibitors. J Med Chem 54, 387-391. 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 evaluation of (4H-1,2,4-triazol-4-yl)isoquinoline derivatives as selective glycine transporter 1 inhibitors. Bioorg Med Chem 20, 34-41. Erdo SL, Kiss B, Rosdy B (1981) Inhibition of dopamine uptake by a new psychostimulant mesocarb (Sydnocarb). Pol J Pharmacol Pharm 33, 141-147. Ganiev MM, Kharlamov AN, Raevskii KS, Guseinov DI (1987) Effect of sidnocarb on learning and memory. Biull Eksp Biol Med 104, 453-454. Schmeichel BE, Zemlan FP, Berridge CW (2013) A selective dopamine reuptake inhibitor improves prefrontal cortex-dependent cognitive function: Potential relevance to attention deficit hyperactivity disorder. Neuropharmacology 64, 321-328. Nic Dhonnchadha BA, Kantak KM (2011) Cognitive enhancers for facilitating drug cue extinction: Insights from animal models. Pharmacol Biochem Behav 99, 229-244. Nic Dhonnchadha BA, Pinard E, Alberati D, Wettstein JG, Spealman RD, Kantak KM (2012) Inhibiting glycine transporter-1 facilitates cocaine-cue extinction and attenuates reacquisition of cocaine-seeking behavior. Drug Alcohol Depend 122, 119-126. Chow TW, Pollock BG, Milgram NW (2007) Potential cognitive enhancing and disease modification effects of SSRIs for Alzheimer’s disease. Neuropsychiatr Dis Treat 3, 627-636. Cassano GB, Puca F, Scapicchio PL, Trabucchi M (2002) Paroxetine and fluoxetine effects on mood and cognitive functions in depressed nondemented elderly patients. J Clin Psychiatry 63, 396-402. Pollock BG, Mulsant BH, Rosen J, Sweet RA, Mazumdar S, Bharucha A, Marin R, Jacob NJ, Huber KA, Kastango KB, Chew ML (2002) Comparison of citalopram, perphenazine, and placebo for the acute treatment of psychosis and behavioral disturbances in hospitalized, demented patients. Am J Psychiatry 159, 460465. 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 proteostasis is regulated by the membrane transport protein ABCC1 in mice. J Clin Invest 121, 3924-3931. Harada K, Nakato K, Yarimizu J, Yamazaki M, Morita M, Takahashi S, Aota M, Saita K, Doihara H, Sato Y, Yamaji T, Ni K, Matsuoka N (2012) A novel glycine transporter-1 (GlyT1) inhibitor, ASP2535 (4-[3-isopropyl-5-(6-phenyl3-pyridyl)-4H-1,2,4-triazol-4-yl]-2,1,3-benzoxadiazol e), improves cognition in animal models of cognitive impair- [281] [282] [283] [284] [285] [286] [287] [288] [289] [290] [291] [292] ment in schizophrenia and Alzheimer’s disease. Eur J Pharmacol 685, 59-69. Murai S, Saito H, Abe E, Masuda Y, Odashima J, Itoh T (1994) MKC-231, a choline uptake enhancer, ameliorates working memory deficits and decreased hippocampal acetylcholine induced by ethylcholine aziridinium ion in mice. J Neural Transm Gen Sect 98, 1-13. Bessho T, Takashina K, Tabata R, Ohshima C, Chaki H, Yamabe H, Egawa M, Tobe A, Saito K (1996) Effect of the novel high affinity choline uptake enhancer 2-(2oxopyrrolidin-1-yl)-N-(2,3-dimethyl-5,6,7,8- tetrahydrofuro [2,3-b] quinolin-4-yl)acetoamide on deficits of water maze learning in rats. Arzneimittelforschung 46, 369373. Bessho T, Takashina K, Eguchi J, Komatsu T, Saito K (2008) MKC-231, a choline-uptake enhancer: (1) long-lasting cognitive improvement after repeated administration in AF64A-treated rats. J Neural Transm 115, 1019-1025. Takashina K, Bessho T, Mori R, Eguchi J, Saito K (2008) MKC-231, a choline uptake enhancer: (2) Effect on synthesis and release of acetylcholine in AF64A-treated rats. J Neural Transm 115, 1027-1035. Takashina K, Bessho T, Mori R, Kawai K, Eguchi J, Saito K (2008) MKC-231, a choline uptake enhancer: (3) Mode of action of MKC-231 in the enhancement of high-affinity choline uptake. J Neural Transm 115, 1037-1046. Akaike A, Maeda T, Kaneko S, Tamura Y (1998) Protective effect of MKC-231, a novel high affinity choline uptake enhancer, on glutamate cytotoxicity in cultured cortical neurons. Jpn J Pharmacol 76, 219-222. Uemura K, Yoshioka S, Surina-Baumgartner DM, Tamagawa T, Miura H, Ueda M, Tamaya N, Iguchi A, Hotta N (1999) Central nervous system-mediated hyperglycemic effects of NIK-247, a cholinesterase inhibitor, and MKC231, a choline uptake enhancer, in rats. Jpn J Pharmacol 79, 113-115. Shirayama Y, Yamamoto A, Nishimura T, Katayama S, Kawahara R (2007) Subsequent exposure to the choline uptake enhancer MKC-231 antagonizes phencyclidineinduced behavioral deficits and reduction in septal cholinergic neurons in rats. Eur Neuropsychopharmacol 17, 616-626. Shimazaki T, Kaku A, Chaki S (2010) D-Serine and a glycine transporter-1 inhibitor enhance social memory in rats. Psychopharmacology (Berl) 209, 263-270. Boulay D, Pichat P, Dargazanli G, Estenne-Bouhtou G, Terranova JP, Rogacki N, Stemmelin J, Coste A, Lanneau C, Desvignes C, Cohen C, Alonso R, Vige X, Biton B, Steinberg R, Sevrin M, Oury-Donat F, George P, Bergis O, Griebel G, Avenet P, Scatton B (2008) Characterization of SSR103800, a selective inhibitor of the glycine transporter-1 in models predictive of therapeutic activity in schizophrenia. Pharmacol Biochem Behav 91, 47-58. Boulay D, Bergis O, Avenet P, Griebel G (2010) The glycine transporter-1 inhibitor SSR103800 displays a selective and specific antipsychotic-like profile in normal and transgenic mice. Neuropsychopharmacology 35, 416427. Black MD, Varty GB, Arad M, Barak S, De LA, Boulay D, Pichat P, Griebel G, Weiner I (2009) Procognitive and antipsychotic efficacy of glycine transport 1 inhibitors (GlyT1) in acute and neurodevelopmental models of schizophrenia: Latent inhibition studies in the rat. Psychopharmacology (Berl) 202, 385-396. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [293] [294] [295] [296] [297] [298] [299] [300] [301] [302] [303] [304] [305] [306] [307] Depoortere R, Dargazanli G, Estenne-Bouhtou G, Coste A, 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, Sevrin M, Vige X, Biton B, Steinberg R, Francon D, Alonso R, Avenet P, Oury-Donat F, Perrault G, Griebel G, George P, Soubrie P, Scatton B (2005) Neurochemical, electrophysiological and pharmacological profiles of the selective inhibitor of the glycine transporter-1 SSR504734, a potential new type of antipsychotic. Neuropsychopharmacology 30, 1963-1985. Singer P, Feldon J, Yee BK (2009) The glycine transporter 1 inhibitor SSR504734 enhances working memory performance in a continuous delayed alternation task in C57BL/6 mice. Psychopharmacology (Berl) 202, 371-384. Singer P, Feldon J, Yee BK (2009) Interactions between the glycine transporter 1(GlyT1) inhibitor SSR504734 and psychoactive drugs in mouse motor behaviour. Eur Neuropsychopharmacol 19, 571-580. Nishikawa H, Inoue T, Izumi T, Nakagawa S, Koyama T (2010) SSR504734, a glycine transporter-1 inhibitor, attenuates acquisition and expression of contextual conditioned fear in rats. Behav Pharmacol 21, 576-579. Nikiforuk A, Kos T, Rafa D, Behl B, Bespalov A, Popik P (2011) Blockade of glycine transporter 1 by SSR-504734 promotes cognitive flexibility in glycine/NMDA receptordependent manner. Neuropharmacology 61, 262-267. (1999) Sufoxazine. Lucelan, Metatone, teniloxazine, Y 8894. Drugs R D 2, 66-67. Anami K, Yamamoto Y, Setoguchi M (1985) Pharmacological studies on sufoxazine (Y-8894). (I) Effects on experimental amnesia in mice. Nihon Yakurigaku Zasshi 85, 71-77. Anami K, Yamamoto Y, Setoguchi M, Maruyama Y (1987) Pharmacological studies on Y-8894. (V) Effect on learning and memory in intact and experimentally amnesic rats. Nihon Yakurigaku Zasshi 89, 145-153. Anami K, Setoguchi M, Senoh H (1988) Pharmacological studies on Y-8894. (VIII). Effects on learning and memory in the radial maze task in mice. Nihon Yakurigaku Zasshi 92, 113-118. Lehr T, Staab A, Trommeshauser D, Schaefer HG, Kloft C (2010) Quantitative pharmacology approach in Alzheimer’s disease: Efficacy modeling of early clinical data to predict clinical outcome of tesofensine. AAPS J 12, 117-129. van de Giessen E, de Bruin K, la Fleur SE, van den Brink W, Booij J (2012) Triple monoamine inhibitor tesofensine decreases food intake, body weight, and striatal dopamine D2/D3 receptor availability in diet-induced obese rats. Eur Neuropsychopharmacol 22, 290-299. Adam Y, Edwards RH, Schuldiner S (2008) Expression and function of the rat vesicular monoamine transporter. Am J Physiol Cell Physiol 294, C1004-C1011. Koeppe RA, Gilman S, Joshi A, Liu S, Little R, Junck L, Heumann M, Frey KA, Albin RL (2005) 11C-DTBZ and 18F-FDG PET measures in differentiating dementias. J Nucl Med 46, 936-944. Gilman S, Koeppe RA, Little R, An H, Junck L, Giordani B, Persad C, Heumann M, Wernette K (2004) Striatal monoamine terminals in Lewy body dementia and Alzheimer’s disease. Ann Neurol 55, 774-780. Koeppe RA, Gilman S, Junck L, Wernette K, Frey KA (2008) Differentiating Alzheimer’s disease from dementia with Lewy bodies and Parkinson’s disease with [308] [309] [310] [311] [312] [313] [314] [315] [316] [317] [318] [319] [320] 65 (+)-[11C]dihydrotetrabenazine positron emission tomography. Alzheimers Dement 4, S67-S76. Nagren K, Halldin C, Rinne JO (2010) Radiopharmaceuticals for positron emission tomography investigations of Alzheimer’s disease. Eur J Nucl Med Mol Imaging 37, 1575-1593. Zhu L, Liu Y, Plossl K, Lieberman B, Liu J, Kung HF (2010) An improved radiosynthesis of [18F]AV-133: A PET imaging agent for vesicular monoamine transporter 2. Nucl Med Biol 37, 133-141. Tsao HH, Lin KJ, Juang JH, Skovronsky DM, Yen TC, Wey SP, Kung MP (2010) Binding characteristics of 9fluoropropyl-(+)-dihydrotetrabenzazine (AV-133) to the vesicular monoamine transporter type 2 in rats. Nucl Med Biol 37, 413-419. Wang JL, Oya S, Parhi AK, Lieberman BP, Ploessl K, Hou C, Kung HF (2010) In vivo studies of the SERTselective [18F]FPBM and VMAT2-selective [18F]AV-133 radiotracers in a rat model of Parkinson’s disease. Nucl Med Biol 37, 479-486. Lin KJ, Weng YH, Wey SP, Hsiao IT, Lu CS, Skovronsky D, Chang HP, Kung MP, Yen TC (2010) Whole-body biodistribution and radiation dosimetry of 18F-FP-(+)DTBZ (18F-AV-133): A novel vesicular monoamine transporter 2 imaging agent. J Nucl Med 51, 14801485. Okamura N, Villemagne VL, Drago J, Pejoska S, Dhamija RK, Mulligan RS, Ellis JR, Ackermann U, O’Keefe G, Jones G, Kung HF, Pontecorvo MJ, Skovronsky D, Rowe CC (2010) In vivo measurement of vesicular monoamine transporter type 2 density in Parkinson disease with (18)FAV-133. J Nucl Med 51, 223-228. Villemagne VL, Okamura N, Pejoska S, Drago J, Mulligan RS, Chetelat G, Ackermann U, O’Keefe G, Jones G, Gong S, Tochon-Danguy H, Kung HF, Masters CL, Skovronsky DM, Rowe CC (2011) In vivo assessment of vesicular monoamine transporter type 2 in dementia with lewy bodies and Alzheimer disease. Arch Neurol 68, 905-912. 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. Nucl Med Biol 38, 1149-1155. Toomey JS, Bhatia S, Moon LT, Orchard EA, Tainter KH, Lokitz SJ, Terry T, Mathis JM, Penman AD (2012) PET imaging a MPTP-induced mouse model of Parkinson’s disease using the fluoropropyl-dihydrotetrabenazine analog [(18)F]-DTBZ (AV-133). PLoS One 7, e39041. Rinne JO, Sahlberg N, Ruottinen H, Nagren K, Lehikoinen P (1998) Striatal uptake of the dopamine reuptake ligand [11C]beta-CFT is reduced in Alzheimer’s disease assessed by positron emission tomography. Neurology 50, 152-156. Rinne JO, Bergman J, Ruottinen H, Haaparanta M, Eronen E, Oikonen V, Sonninen P, Solin O (1999) Striatal uptake of a novel PET ligand, [18F]beta-CFT, is reduced in early Parkinson’s disease. Synapse 31, 119-124. Rinne JO, Nagren K (2010) Positron emission tomography in at risk patients and in the progression of mild cognitive impairment to Alzheimer’s disease. J Alzheimers Dis 19, 291-300. McKeith I, O’Brien J, Walker Z, Tatsch K, Booij J, Darcourt J, Padovani A, Giubbini R, Bonuccelli U, Volterrani D, Holmes C, Kemp P, Tabet N, Meyer I, Reininger C (2007) Sensitivity and specificity of dopamine transporter imaging with 123I-FP-CIT SPECT in dementia with Lewy 66 [321] [322] [323] [324] [325] [326] [327] [328] [329] [330] [331] [332] [333] [334] 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. Walker Z, Jaros E, Walker RW, Lee L, Costa DC, Livingston G, Ince PG, Perry R, McKeith I, Katona CL (2007) Dementia with Lewy bodies: A comparison of clinical diagnosis, FP-CIT single photon emission computed tomography imaging and autopsy. J Neurol Neurosurg Psychiatry 78, 1176-1181. Vlaar AM, de NT, Kessels AG, Vreeling FW, Winogrodzka A, Mess WH, Tromp SC, van Kroonenburgh MJ, Weber WE (2008) Diagnostic value of 123I-ioflupane and 123I-iodobenzamide SPECT scans in 248 patients with parkinsonian syndromes. Eur Neurol 59, 258-266. Roselli F, Pisciotta NM, Perneczky R, Pennelli M, Aniello MS, De Caro MF, Ferrannini E, Tartaglione B, Defazio G, Rubini G, Livrea P (2009) Severity of neuropsychiatric symptoms and dopamine transporter levels in dementia with Lewy bodies: A 123I-FP-CIT SPECT study. Mov Disord 24, 2097-2103. Roselli F, Pisciotta NM, Pennelli M, Aniello MS, Gigante A, De Caro MF, Ferrannini E, Tartaglione B, NiccoliAsabella A, Defazio G, Livrea P, Rubini G (2010) Midbrain SERT in degenerative parkinsonisms: A 123IFP-CIT SPECT study. Mov Disord 25, 1853-1859. Gerasimou G, Costa DC, Papanastasiou E, Bostanjiopoulou S, Arnaoutoglou M, Moralidis E, Aggelopoulou T, Gotzamani-Psarrakou A (2012) SPECT study with I-123-Ioflupane (DaTSCAN) in patients with essential tremor. Is there any correlation with Parkinson’s disease? Ann Nucl Med 26, 337-344. Scott R, Bourtchuladze R, Gossweiler S, Dubnau J, Tully T (2002) CREB and the discovery of cognitive enhancers. J Mol Neurosci 19, 171-177. Saura CA, Valero J (2011) The role of CREB signaling in Alzheimer’s disease and other cognitive disorders. Rev Neurosci 22, 153-169. Saura CA (2012) CREB-regulated transcription coactivator 1-dependent transcription in Alzheimer’s disease mice. Neurodegener Dis 10, 250-252. Scott Bitner R (2012) Cyclic AMP response elementbinding protein (CREB) phosphorylation: A mechanistic marker in the development of memory enhancing Alzheimer’s disease therapeutics. Biochem Pharmacol 83, 705-714. Oliveira AM, Bading H (2011) Calcium signaling in cognition and aging-dependent cognitive decline. Biofactors 37, 168-174. Espana J, Valero J, Minano-Molina AJ, Masgrau R, Martin E, Guardia-Laguarta C, Lleo A, Gimenez-Llort L, Rodriguez-Alvarez J, Saura CA (2010) beta-Amyloid disrupts activity-dependent gene transcription required for memory through the CREB coactivator CRTC1. J Neurosci 30, 9402-9410. Helm KA, Haberman RP, Dean SL, Hoyt EC, Melcher T, Lund PK, Gallagher M (2005) GABAB receptor antagonist SGS742 improves spatial memory and reduces protein binding to the cAMP response element (CRE) in the hippocampus. Neuropharmacology 48, 956-964. Florian C, Mons N, Roullet P (2006) CREB antisense oligodeoxynucleotide administration into the dorsal hippocampal CA3 region impairs long- but not short-term spatial memory in mice. Learn Mem 13, 465-472. Fontan-Lozano A, Romero-Granados R, del-Pozo-Martin Y, Suarez-Pereira I, gado-Garcia JM, Penninger JM, Carrion AM (2009) Lack of DREAM protein enhances [335] [336] [337] [338] [339] [340] [341] [342] [343] [344] [345] [346] [347] [348] [349] [350] [351] learning and memory and slows brain aging. Curr Biol 19, 54-60. Chow N, Bell RD, Deane R, Streb JW, Chen J, Brooks A, Van NW, Miano JM, Zlokovic BV (2007) Serum response factor and myocardin mediate arterial hypercontractility and cerebral blood flow dysregulation in Alzheimer’s phenotype. Proc Natl Acad Sci U S A 104, 823828. Bell RD (2012) The imbalance of vascular molecules in Alzheimer’s disease. J Alzheimers Dis 32, 699-709. McPhee I, Gibson LC, Kewney J, Darroch C, Stevens PA, Spinks D, Cooreman A, Mackenzie SJ (2005) Cyclic nucleotide signalling: A molecular approach to drug discovery for Alzheimer’s disease. Biochem Soc Trans 33, 1330-1332. Reddy PH, Beal MF (2008) Amyloid beta, mitochondrial dysfunction and synaptic damage: Implications for cognitive decline in aging and Alzheimer’s disease. Trends Mol Med 14, 45-53. Di Bona D, Scapagnini G, Candore G, Castiglia L, Colonna-Romano G, Duro G, Nuzzo D, Iemolo F, Lio D, Pellicano M, Scafidi V, Caruso C, Vasto S (2010) Immune-inflammatory responses and oxidative stress in Alzheimer’s disease: Therapeutic implications. Curr Pharm Des 16, 684-691. Obrenovich ME, Li Y, Parvathaneni K, Yendluri BB, Palacios HH, Leszek J, Aliev G (2011) Antioxidants in health, disease and aging. CNS Neurol Disord Drug Targets 10, 192-207. Pratico D (2008) Oxidative stress hypothesis in Alzheimer’s disease: A reappraisal. Trends Pharmacol Sci 29, 609-615. Pratico D (2008) Evidence of oxidative stress in Alzheimer’s disease brain and antioxidant therapy: Lights and shadows. Ann N Y Acad Sci 1147, 70-78. Young KJ, Bennett JP (2010) The mitochondrial secret(ase) of Alzheimer’s disease. J Alzheimers Dis 20(Suppl 2), S381-S400. Halliwell B (1999) Antioxidant defence mechanisms: From the beginning to the end (of the beginning). Free Radic Res 31, 261-272. Halliwell B (2001) Role of free radicals in the neurodegenerative diseases: Therapeutic implications for antioxidant treatment. Drugs Aging 18, 685-716. Halliwell B (2006) Oxidative stress and neurodegeneration: Where are we now? J Neurochem 97, 1634-1658. Halliwell B (2011) Free radicals and antioxidants - quo vadis? Trends Pharmacol Sci 32, 125-130. Lee HP, Zhu X, Casadesus G, Castellani RJ, Nunomura A, Smith MA, Lee HG, Perry G (2010) Antioxidant approaches for the treatment of Alzheimer’s disease. Expert Rev Neurother 10, 1201-1208. Vina J, Lloret A, Giraldo E, Badia MC, Alonso MD (2011) Antioxidant pathways in Alzheimer’s disease: Possibilities of intervention. Curr Pharm Des 17, 3861-3864. Baum L, Lam CW, Cheung SK, Kwok T, Lui V, Tsoh J, Lam L, Leung V, Hui E, Ng C, Woo J, Chiu HF, Goggins WB, Zee BC, Cheng KF, Fong CY, Wong A, Mok H, Chow MS, Ho PC, Ip SP, Ho CS, Yu XW, Lai CY, Chan MH, Szeto S, Chan IH, Mok V (2008) Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J Clin Psychopharmacol 28, 110-113. Halliwell B (1999) Vitamin C: Poison, prophylactic or panacea? Trends Biochem Sci 24, 255-259. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [352] [353] [354] [355] [356] [357] [358] [359] [360] [361] [362] [363] [364] [365] [366] [367] [368] [369] 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 be detrimental. J Alzheimers Dis 17, 143-149. Brewer GJ (2010) Why vitamin E therapy fails for treatment of Alzheimer’s disease. J Alzheimers Dis 19, 27-30. Usoro OB, Mousa SA (2010) Vitamin E forms in Alzheimer’s disease: A review of controversial and clinical experiences. Crit Rev Food Sci Nutr 50, 414-419. Guan JZ, Guan WP, Maeda T, Makino N (2012) Effect of vitamin E administration on the elevated oxygen stress and the telomeric and subtelomeric status in Alzheimer’s disease. Gerontology 58, 62-69. Behl C (1999) Alzheimer’s disease and oxidative stress: Implications for novel therapeutic approaches. Prog Neurobiol 57, 301-323. Behl C (2005) Oxidative stress in Alzheimer’s disease: Implications for prevention and therapy. Subcell Biochem 38, 65-78. Aliev G, Smith MA, Seyidov D, Neal ML, Lamb BT, Nunomura A, Gasimov EK, Vinters HV, Perry G, Lamanna JC, Friedland RP (2002) The role of oxidative stress in the pathophysiology of cerebrovascular lesions in Alzheimer’s disease. Brain Pathol 12, 21-35. Aliev G, Obrenovich ME, Reddy VP, Shenk JC, Moreira PI, Nunomura A, Zhu X, Smith MA, Perry G (2008) Antioxidant therapy in Alzheimer’s disease: Theory and practice. Mini Rev Med Chem 8, 1395-1406. Aliev G, Palacios HH, Walrafen B, Lipsitt AE, Obrenovich ME, Morales L (2009) Brain mitochondria as a primary target in the development of treatment strategies for Alzheimer disease. Int J Biochem Cell Biol 41, 19892004. Moreira PI, Honda K, Liu Q, Aliev G, Oliveira CR, Santos MS, Zhu X, Smith MA, Perry G (2005) Alzheimer’s disease and oxidative stress: The old problem remains unsolved. Curr Med Chem Central Nervous System Agents 5, 51-62. Moreira PI, Santos MS, Oliveira CR (2007) Alzheimer’s disease: A lesson from mitochondrial dysfunction. Antioxid Redox Signal 9, 1621-1630. Chauhan V, Chauhan A (2006) Oxidative stress in Alzheimer’s disease. Pathophysiology 13, 195-208. Nunomura A, Castellani RJ, Zhu X, Moreira PI, Perry G, Smith MA (2006) Involvement of oxidative stress in Alzheimer disease. J Neuropathol Exp Neurol 65, 631641. Nunomura A, Moreira PI, Lee HG, Zhu X, Castellani RJ, Smith MA, Perry G (2007) Neuronal death and survival under oxidative stress in Alzheimer and Parkinson diseases. CNS Neurol Disord Drug Targets 6, 411-423. Onyango IG, Khan SM (2006) Oxidative stress, mitochondrial dysfunction, and stress signaling in Alzheimer’s disease. Curr Alzheimer Res 3, 339-349. Liu Q, Xie F, Rolston R, Moreira PI, Nunomura A, Zhu X, Smith MA, Perry G (2007) Prevention and treatment of Alzheimer disease and aging: Antioxidants. Mini Rev Med Chem 7, 171-180. Kamat CD, Gadal S, Mhatre M, Williamson KS, Pye QN, Hensley K (2008) Antioxidants in central nervous system diseases: Preclinical promise and translational challenges. J Alzheimers Dis 15, 473-493. Toogood PL (2008) Mitochondrial drugs. Curr Opin Chem Biol 12, 457-463. [370] [371] [372] [373] [374] [375] [376] [377] [378] [379] [380] [381] [382] [383] [384] [385] [386] 67 Shen L, Ji HF (2010) Insights into the disappointing clinical trials of antioxidants in neurodegenerative diseases. J Alzheimers Dis 19, 1141-1142. Mecocci P, Polidori MC (2012) Antioxidant clinical trials in mild cognitive impairment and Alzheimer’s disease. Biochim Biophys Acta 1822, 631-638. Pocernich CB, Butterfield DA (2012) Elevation of glutathione as a therapeutic strategy in Alzheimer disease. Biochim Biophys Acta 1822, 625-630. Galasko DR, Peskind E, Clark CM, Quinn JF, Ringman JM, Jicha GA, Cotman C, Cottrell B, Montine TJ, Thomas RG, Aisen P (2012) Antioxidants for Alzheimer disease: A randomized clinical trial with cerebrospinal fluid biomarker measures. Arch Neurol 69, 836841. Kitagawa Y (2006) Edaravone in acute ischemic stroke. Intern Med 45, 225-226. Tsujita K, Shimomura H, Kaikita K, Kawano H, Hokamaki J, Nagayoshi Y, Yamashita T, Fukuda M, Nakamura Y, Sakamoto T, Yoshimura M, Ogawa H (2006) Long-term efficacy of edaravone in patients with acute myocardial infarction. Circ J 70, 832-837. Lapchak PA (2010) A critical assessment of edaravone acute ischemic stroke efficacy trials: Is edaravone an effective neuroprotective therapy? Expert Opin Pharmacother 11, 1753-1763. Kikuchi K, Kawahara K, Miyagi N, Uchikado H, Kuramoto T, Morimoto Y, Tancharoen S, Miura N, Takenouchi K, Oyama Y, Shrestha B, Matsuda F, Yoshida Y, Arimura S, Mera K, Tada K, Yoshinaga N, Maenosono R, Ohno Y, Hashiguchi T, Maruyama I, Shigemori M (2010) Edaravone: A new therapeutic approach for the treatment of acute stroke. Med Hypotheses 75, 583585. Gillis JC, Benefield P, McTavish D (1994) Idebenone. A review of its pharmacodynamic and pharmacokinetic properties, and therapeutic use in age-related cognitive disorders. Drugs Aging 5, 133-152. Weyer G, Babej-Dolle RM, Hadler D, Hofmann S, Herrmann WM (1997) A controlled study of 2 doses of idebenone in the treatment of Alzheimer’s disease. Neuropsychobiology 36, 73-82. Voronkova KV, Meleshkov MN (2009) Use of Noben (idebenone) in the treatment of dementia and memory impairments without dementia. Neurosci Behav Physiol 39, 501-506. Haley EC Jr (1998) High-dose tirilazad for acute stroke (RANTTAS II). RANTTAS II Investigators. Stroke 29, 1256-1257. Dorsch NW, Kassell NF, Sinkula MS (2001) Metaanalysis of trials of tirilazad mesylate in aneurysmal SAH. Acta Neurochir Suppl 77, 233-235. Kavanagh RJ, Kam PC (2001) Lazaroids: Efficacy and mechanism of action of the 21-aminosteroids in neuroprotection. Br J Anaesth 86, 110-119. Jang YG, Ilodigwe D, Macdonald RL (2009) Metaanalysis of tirilazad mesylate in patients with aneurysmal subarachnoid hemorrhage. Neurocrit Care 10, 141-147. Kelso GF, Porteous CM, Hughes G, Ledgerwood EC, Gane AM, Smith RA, Murphy MP (2002) Prevention of mitochondrial oxidative damage using targeted antioxidants. Ann N Y Acad Sci 959, 263-274. Murphy MP, Smith RA (2007) Targeting antioxidants to mitochondria by conjugation to lipophilic cations. Annu Rev Pharmacol Toxicol 47, 629-656. 68 [387] [388] [389] [390] [391] [392] [393] [394] [395] [396] [397] [398] [399] [400] [401] [402] [403] [404] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Smith RA, Murphy MP (2010) Animal and human studies with the mitochondria-targeted antioxidant MitoQ. Ann N Y Acad Sci 1201, 96-103. Smith RA, Murphy MP (2011) Mitochondria-targeted antioxidants as therapies. Discov Med 11, 106-114. Reddy PH (2008) Mitochondrial medicine for aging and neurodegenerative diseases. Neuromolecular Med 10, 291315. Reddy PH, Reddy TP (2011) Mitochondria as a therapeutic target for aging and neurodegenerative diseases. Curr Alzheimer Res 8, 393-409. Reddy PH, Tripathi R, Troung Q, Tirumala K, Reddy TP, Anekonda V, Shirendeb UP, Calkins MJ, Reddy AP, Mao P, Manczak M (2011) Abnormal mitochondrial dynamics and synaptic degeneration as early events in Alzheimer’s disease: Implications to mitochondria-targeted antioxidant therapeutics. Biochim Biophys Acta 1822, 639-649. Bonda DJ, Wang X, Gustaw-Rothenberg KA, Perry G, Smith MA, Zhu X (2009) Mitochondrial drugs for Alzheimer disease. Pharmaceuticals (Basel) 2, 287-298. Bonda DJ, Wang X, Perry G, Nunomura A, Tabaton M, Zhu X, Smith MA (2010) Oxidative stress in Alzheimer disease: A possibility for prevention. Neuropharmacology 59, 290-294. Manczak M, Mao P, Calkins MJ, Cornea A, Reddy AP, Murphy MP, Szeto HH, Park B, Reddy PH (2010) Mitochondria-targeted antioxidants protect against amyloid-beta toxicity in Alzheimer’s disease neurons. J Alzheimers Dis 20(Suppl 2), S609-S631. Smith RA, Porteous CM, Coulter CV, Murphy MP (1999) Selective targeting of an antioxidant to mitochondria. Eur J Biochem 263, 709-716. Reddy PH (2006) Mitochondrial oxidative damage in aging and Alzheimer’s disease: Implications for mitochondrially targeted antioxidant therapeutics. J Biomed Biotechnol 2006, 31372. Kotake Y (1999) Pharmacologic properties of phenyl Ntert-butylnitrone. Antioxid Redox Signal 1, 481-499. Lin S, Rhodes PG, Lei M, Zhang F, Cai Z (2004) alphaPhenyl-n-tert-butyl-nitrone attenuates hypoxic-ischemic white matter injury in the neonatal rat brain. Brain Res 1007, 132-141. Robertson L, Hartley RC (2009) Synthesis of Narylpyridinium salts bearing a nitrone spin trap as potential mitochondria-targeted antioxidants. Tetrahedron 65, 5284-5292. Zhao K, Zhao GM, Wu D, Soong Y, Birk AV, Schiller PW, Szeto HH (2004) Cell-permeable peptide antioxidants targeted to inner mitochondrial membrane inhibit mitochondrial swelling, oxidative cell death, and reperfusion injury. J Biol Chem 279, 34682-34690. Reddy PH (2007) Mitochondrial dysfunction in aging and Alzheimer’s disease: Strategies to protect neurons. Antioxid Redox Signal 9, 1647-1658. Rocha M, Hernandez-Mijares A, Garcia-Malpartida K, Banuls C, Bellod L, Victor VM (2010) Mitochondriatargeted antioxidant peptides. Curr Pharm Des 16, 3124-3131. Szeto HH, Schiller PW (2011) Novel therapies targeting inner mitochondrial membrane–from discovery to clinical development. Pharm Res 28, 2669-2679. Chyan YJ, Poeggeler B, Omar RA, Chain DG, Frangione B, Ghiso J, Pappolla MA (1999) Potent neuroprotective properties against the Alzheimer beta-amyloid by an endogenous melatonin-related indole structure, [405] [406] [407] [408] [409] [410] [411] [412] [413] [414] [415] [416] [417] [418] indole-3-propionic acid. J Biol Chem 274, 2193721942. Liang LP, Huang J, Fulton R, Day BJ, Patel M (2007) An orally active catalytic metalloporphyrin protects against 1methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity in vivo. J Neurosci 27, 4326-4333. Liang LP, Waldbaum S, Rowley S, Huang TT, Day BJ, Patel M (2012) Mitochondrial oxidative stress and epilepsy in SOD2 deficient mice: Attenuation by a lipophilic metalloporphyrin. Neurobiol Dis 45, 1068-1076. Day BJ, Patel M (2008) Metalloporphyrin catalytic antioxidants for the potential treatment of neurodegenerative diseases. Drugs Future 33, 1025-1032. Batinic-Haberle I, Reboucas JS, Spasojevic I (2010) Superoxide dismutase mimics: Chemistry, pharmacology, and therapeutic potential. Antioxid Redox Signal 13, 877918. 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 mimics. Free Radic Biol Med 51, 1035-1053. Sheng H, Batinc-Haberle I, Warner DS (2002) Catalytic antioxidants as novel pharmacologic approaches to treatment of ischemic brain injury. Drug News Perspect 15, 654-665. Sheng H, Yang W, Fukuda S, Tse HM, Paschen W, Johnson K, Batinic-Haberle I, Crapo JD, Pearlstein RD, Piganelli J, Warner DS (2009) Long-term neuroprotection from a potent redox-modulating metalloporphyrin in the rat. Free Radic Biol Med 47, 917-923. Sheng H, Spasojevic I, Tse HM, Jung JY, Hong J, Zhang Z, Piganelli JD, Batinic-Haberle I, Warner DS (2011) Neuroprotective efficacy from a lipophilic redox-modulating Mn(III) N-Hexylpyridylporphyrin, MnTnHex-2-PyP: Rodent models of ischemic stroke and subarachnoid hemorrhage. J Pharmacol Exp Ther 338, 906-916. Li L, Zhang B, Tao Y, Wang Y, Wei H, Zhao J, Huang R, Pei Z (2009) DL-3-n-butylphthalide protects endothelial cells against oxidative/nitrosative stress, mitochondrial damage and subsequent cell death after oxygen glucose deprivation in vitro. Brain Res 1290, 91-101. Xiong N, Huang J, Chen C, Zhao Y, Zhang Z, Jia M, Zhang Z, Hou L, Yang H, Cao X, Liang Z, Zhang Y, Sun S, Lin Z, Wang T (2012) Dl-3-n-butylphthalide, a natural antioxidant, protects dopamine neurons in rotenone models for Parkinson’s disease. Neurobiol Aging 33, 1777-1791. Valgimigli L, Valgimigli M, Gaiani S, Pedulli GF, Bolondi L (2000) Measurement of oxidative stress in human liver by EPR spin-probe technique. Free Radic Res 33, 167-178. D’Aleo V, Del GS, Martano M, Bonamassa B, Canistro D, Soleti A, Valgimigli L, Paolini M, Filipponi F, Boggi U, Del PS, Lupi R (2009) The non-peptidyl low molecular weight radical scavenger IAC protects human pancreatic islets from lipotoxicity. Mol Cell Endocrinol 309, 63-66. Vasina V, Broccoli M, Ursino MG, Bellot SF, Soleti A, Paolini M, De PF (2009) Effects of the non-peptidyl low molecular weight radical scavenger IAC in DNBS-induced colitis in rats. Eur J Pharmacol 614, 137-145. Vasina V, Broccoli M, Ursino MG, Canistro D, Valgimigli L, Soleti A, Paolini M, De PF (2010) Non-peptidyl low molecular weight radical scavenger IAC attenuates DSSinduced colitis in rats. World J Gastroenterol 16, 36423650. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [419] [420] [421] [422] [423] [424] [425] [426] [427] [428] [429] [430] [431] Puolivali J, Nurmi A, Miettinen TK, Soleti A, Riccardino F, Kalesnykas G, Heikkinen T, Vartiainen N, Pussinen R, Tahtivaara L, Lehtimaki K, Yrjanheikki J, Canistro D, Sapone A, Spisni E, Paolini M (2011) The radical scavenger IAC (bis(1-hydroxy-2,2,6,6-tetramethyl-4piperidinyl) decantionate) decreases mortality, enhances cognitive functions in water maze and reduces amyloid plaque burden in hAbetaPP transgenic mice. J Alzheimers Dis 27, 499-510. Giles FJ, Fracasso PM, Kantarjian HM, Cortes JE, Brown RA, Verstovsek S, Alvarado Y, Thomas DA, Faderl S, Garcia-Manero G, Wright LP, Samson T, Cahill A, Lambert P, Plunkett W, Sznol M, DiPersio JF, Gandhi V (2003) Phase I and pharmacodynamic study of Triapine, a novel ribonucleotide reductase inhibitor, in patients with advanced leukemia. Leuk Res 27, 1077-1083. 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 thiosemicarbazone), a novel neuroprotectant, elicits its function in primary neuronal cultures by up-regulating Bcl-2 expression. Neuroscience 135, 191-201. Gao X, Zheng CY, Qin GW, Tang XC, Zhang HY (2012) S-52, a novel nootropic compound, protects against beta-amyloid induced neuronal injury by attenuating mitochondrial dysfunction. J Neurosci Res 90, 19811988. Zhong SZ, Ge QH, Li Q, Qu R, Ma SP (2009) Peoniflorin attentuates Abeta((1-42))-mediated neurotoxicity by regulating calcium homeostasis and ameliorating oxidative stress in hippocampus of rats. J Neurol Sci 280, 71-78. Hatanaka M, Nishizawa C, Kakinoki T, Takahashi K, Nakamura S, Mashino T (2008) 2,2 -Pyridoin derivatives protect HL-60 cells against oxidative stress. Bioorg Med Chem Lett 18, 5290-5293. 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 of quetiapine: Implications for the treatment of Alzheimer’s disease. FEBS J 275, 3718-3728. Han M, Liu Y, Tan Q, Zhang B, Wang W, Liu J, Zhang XJ, Wang YY, Zhang JM (2011) Therapeutic efficacy of stemazole in a beta-amyloid injection rat model of Alzheimer’s disease. Eur J Pharmacol 657, 104-110. Choi SJ, Jeong CH, Choi SG, Chun JY, Kim YJ, Lee J, Shin DH, Heo HJ (2009) Zeatin prevents amyloid betainduced neurotoxicity and scopolamine-induced cognitive deficits. J Med Food 12, 271-277. Flaherty DP, Kiyota T, Dong Y, Ikezu T, Vennerstrom JL (2010) Phenolic bis-styrylbenzenes as beta-amyloid binding ligands and free radical scavengers. J Med Chem 53, 7992-7999. Lenhart JA, Ling X, Gandhi R, Guo TL, Gerk PM, Brunzell DH, Zhang S (2010) “Clicked” bivalent ligands containing curcumin and cholesterol as multifunctional abeta oligomerization inhibitors: Design, synthesis, and biological characterization. J Med Chem 53, 6198-6209. Jung YS, Kang TS, Yoon JH, Joe BY, Lim HJ, Seong CM, Park WK, Kong JY, Cho J, Park NS (2002) Synthesis and evaluation of 4-hydroxyphenylacetic acid amides and 4hydroxycinnamamides as antioxidants. Bioorg Med Chem Lett 12, 2599-2602. Weston RM, Jarrott B, Ishizuka Y, Callaway JK (2006) AM-36 modulates the neutrophil inflammatory response and reduces breakdown of the blood brain barrier after [432] [433] [434] [435] [436] [437] [438] [439] [440] [441] [442] [443] [444] [445] [446] 69 endothelin-1 induced focal brain ischaemia. Br J Pharmacol 149, 712-723. 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 neuroprotective agent, following intravenous administration to rats. J Pharm Pharmacol 60, 171-178. Sorbera LA, Leeson PA, Castaner J (2001) CPI-1189. Drugs Future 26, 647-650. Clifford DB, McArthur JC, Schifitto G, Kieburtz K, McDermott MP, Letendre S, Cohen BA, Marder K, Ellis RJ, Marra CM (2002) A randomized clinical trial of CPI1189 for HIV-associated cognitive-motor impairment. Neurology 59, 1568-1573. Strid S, Borga O, Edenius C, Jostell KG, Odergren T, Weil A (2002) Pharmacokinetics in renally impaired subjects of NXY-059, a nitrone-based, free-radical trapping agent developed for the treatment of acute stroke. Eur J Clin Pharmacol 58, 409-415. Green AR, Shuaib A (2006) Therapeutic strategies for the treatment of stroke. Drug Discov Today 11, 681-693. Lees KR, Zivin JA, Ashwood T, Davalos A, Davis SM, Diener HC, Grotta J, Lyden P, Shuaib A, Hardemark HG, Wasiewski WW (2006) NXY-059 for acute ischemic stroke. N Engl J Med 354, 588-600. Lees KR, Davalos A, Davis SM, Diener HC, Grotta J, Lyden P, Shuaib A, Ashwood T, Hardemark HG, Wasiewski W, Emeribe U, Zivin JA (2006) Additional outcomes and subgroup analyses of NXY-059 for acute ischemic stroke in the SAINT I trial. Stroke 37, 2970-2978. Ginsberg MD (2007) Life after cerovive: A personal perspective on ischemic neuroprotection in the post-NXY-059 era. Stroke 38, 1967-1972. Lyden PD, Shuaib A, Lees KR, Davalos A, Davis SM, Diener HC, Grotta JC, Ashwood TJ, Hardemark HG, Svensson HH, Rodichok L, Wasiewski WW, Ahlberg G (2007) Safety and tolerability of NXY-059 for acute intracerebral hemorrhage: The CHANT Trial. Stroke 38, 2262-2269. Saver JL (2007) Clinical impact of NXY-059 demonstrated in the SAINT I trial: Derivation of number needed to treat for benefit over entire range of functional disability. Stroke 38, 1515-1518. Shuaib A, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, Diener HC, Ashwood T, Wasiewski WW, Emeribe U (2007) NXY-059 for the treatment of acute ischemic stroke. N Engl J Med 357, 562-571. Cheng YF, Jiang J, Hu P, Reinholdsson I, Guo W, Asenblad N, Nilsson D (2008) Pharmacokinetics of 8-hour intravenous infusion of NXY-059: A phase I, randomized, double-blind (within dose panels), placebo-controlled study in healthy Chinese volunteers. Clin Ther 30, 23422353. Diener HC, Lees KR, Lyden P, Grotta J, Davalos A, Davis SM, Shuaib A, Ashwood T, Wasiewski W, Alderfer V, Hardemark HG, Rodichok L (2008) NXY-059 for the treatment of acute stroke: Pooled analysis of the SAINT I and II Trials. Stroke 39, 1751-1758. Savitz SI, Schabitz WR (2008) A Critique of SAINT II: Wishful thinking, dashed hopes, and the future of neuroprotection for acute stroke. Stroke 39, 1389-1391. Iwashita A, Maemoto T, Nakada H, Shima I, Matsuoka N, Hisajima H (2003) A novel potent radical scavenger, 8-(4-fluorophenyl)-2-((2E)-3-phenyl-2propenoyl)-1,2,3,4-tetrahydropyrazol o[5,1-c] [1,2,4]tri- 70 [447] [448] [449] [450] [451] [452] [453] [454] [455] [456] [457] [458] [459] [460] [461] 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 cultured primary neurons and attenuates brain injury after focal ischemia in rats. J Pharmacol Exp Ther 307, 961-968. Dage JL, Ackermann BL, Barbuch RJ, Bernotas RC, Ohlweiler DF, Haegele KD, Thomas CE (1997) Evidence for a novel pentyl radical adduct of the cyclic nitrone spin trap MDL 101,002. Free Radic Biol Med 22, 807-812. 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 efficacious in permanent and transient focal ischemia models. Life Sci 63, 241-253. Galeano M, Torre V, Deodato B, Campo GM, Colonna M, Sturiale A, Squadrito F, Cavallari V, Cucinotta D, Buemi M, Altavilla D (2001) Raxofelast, a hydrophilic vitamin Elike antioxidant, stimulates wound healing in genetically diabetic mice. Surgery 129, 467-477. Altavilla D, Galeano M, Bitto A, Minutoli L, Squadrito G, Seminara P, Venuti FS, Torre V, Calo M, Colonna M, Lo CP, Giugliano G, Scuderi N, Mioni C, Leone S, Squadrito F (2005) Lipid peroxidation inhibition by raxofelast improves angiogenesis and wound healing in experimental burn wounds. Shock 24, 85-91. Altavilla D, Marini H, Seminara P, Squadrito G, Minutoli L, Passaniti M, Bitto A, Calapai G, Calo M, Caputi AP, Squadrito F (2005) Protective effects of antioxidant raxofelast in alcohol-induced liver disease in mice. Pharmacology 74, 6-14. Bitto A, Minutoli L, Squadrito F, Polito F, Altavilla D (2007) Raxofelast, (+/–)5-(acetyloxy)-2,3-dihydro-4,6,7trimethyl-2-benzofuranacetic acid: A new antioxidant to modulate the inflammatory response during ischemiareperfusion injury and impaired wound healing. Mini Rev Med Chem 7, 339-343. Dorrell S (2001) Alzheimer’s disease - a metallic problem. Drug Discov Today 6, 61-62. Perry G, Cash AD, Srinivas R, Smith MA (2002) Metals and oxidative homeostasis in Alzheimer’s disease. Drug Dev Res 56, 293-299. Bush AI (2003) The metallobiology of Alzheimer’s disease. Trends Neurosci 26, 207-214. Crouch PJ, Barnham KJ, Bush AI, White AR (2006) Therapeutic treatments for Alzheimer’s disease based on metal bioavailability. Drug News Perspect 19, 469-474. Schugar H, Green DE, Bowen ML, Scott LE, Storr T, Bohmerle K, Thomas F, Allen DD, Lockman PR, Merkel M, Thompson KH, Orvig C (2007) Combating Alzheimer’s disease with multifunctional molecules designed for metal passivation. Angew Chem Int Ed Engl 46, 1716-1718. Shcherbatykh I, Carpenter DO (2007) The role of metals in the etiology of Alzheimer’s disease. J Alzheimers Dis 11, 191-205. 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 metal coordinating ability of multifunctional carbohydratecontaining compounds for Alzheimer’s therapy. J Am Chem Soc 129, 7453-7463. Barnham KJ, Bush AI (2008) Metals in Alzheimer’s and Parkinson’s diseases. Curr Opin Chem Biol 12, 222-228. Bush AI (2008) Drug development based on the metals hypothesis of Alzheimer’s disease. J Alzheimers Dis 15, 223-240. [462] [463] [464] [465] [466] [467] [468] [469] [470] [471] [472] [473] [474] [475] [476] [477] Bush AI, Tanzi RE (2008) Therapeutics for Alzheimer’s disease based on the metal hypothesis. Neurotherapeutics 5, 421-432. Bolognin S, Messori L, Zatta P (2009) Metal ion physiopathology in neurodegenerative disorders. Neuromolecular Med 11, 223-238. Bolognin S, Drago D, Messori L, Zatta P (2009) Chelation therapy for neurodegenerative diseases. Med Res Rev 29, 547-570. 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 intercalation compounds with metal chelating properties for application in Alzheimer’s disease. J Am Chem Soc 131, 1436-1451. Storr T, Scott LE, Bowen ML, Green DE, Thompson KH, Schugar HJ, Orvig C (2009) Glycosylated tetrahydrosalens as multifunctional molecules for Alzheimer’s therapy. Dalton Trans, 3034-3043. Zatta P, Drago D, Bolognin S, Sensi SL (2009) Alzheimer’s disease, metal ions and metal homeostatic therapy. Trends Pharmacol Sci 30, 346-355. Bandyopadhyay S, Huang X, Lahiri DK, Rogers JT (2010) Novel drug targets based on metallobiology of Alzheimer’s disease. Expert Opin Ther Targets 14, 1177-1197. Choi JS, Braymer JJ, Nanga RP, Ramamoorthy A, Lim MH (2010) Design of small molecules that target metalA{beta} species and regulate metal-induced A{beta} aggregation and neurotoxicity. Proc Natl Acad Sci U S A 107, 21990-21995. Braymer JJ, DeToma AS, Choi JS, Ko KS, Lim MH (2010) Recent development of bifunctional small molecules to study metal-amyloid-beta species in Alzheimer’s disease. Int J Alzheimers Dis 2011, 623051. Duce JA, Bush AI (2010) Biological metals and Alzheimer’s disease: Implications for therapeutics and diagnostics. Prog Neurobiol 92, 1-18. Rodriguez-Rodriguez C, Rimola A, Rodriguez-Santiago L, Ugliengo P, Avarez-Larena A, Gutierrez-de-Teran H, Sodupe M, Gonzalez-Duarte P (2010) Crystal structure of thioflavin-T and its binding to amyloid fibrils: Insights at the molecular level. Chem Commun (Camb) 46, 11561158. Braymer JJ, Choi JS, DeToma AS, Wang C, Nam K, Kampf JW, Ramamoorthy A, Lim MH (2011) Development of bifunctional stilbene derivatives for targeting and modulating metal-amyloid-beta species. Inorg Chem 50, 10724-10734. Choi JS, Braymer JJ, Park SK, Mustafa S, Chae J, Lim MH (2011) Synthesis and characterization of IMPY derivatives that regulate metal-induced amyloid-beta aggregation. Metallomics 3, 284-291. Rimola A, Ali-Torres J, Rodriguez-Rodriguez C, Poater J, Matito E, Sola M, Sodupe M (2011) Ab initio design of chelating ligands relevant to Alzheimer’s disease: Influence of metalloaromaticity. J Phys Chem A 115, 12659-12666. Kenche VB, Barnham KJ (2011) Alzheimer’s disease & metals: Therapeutic opportunities. Br J Pharmacol 163, 211-219. Tomljenovic L (2011) Aluminum and Alzheimer’s disease: After a century of controversy, is there a plausible link? J Alzheimers Dis 23, 567-598. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [478] [479] [480] [481] [482] [483] [484] [485] [486] [487] [488] [489] [490] [491] [492] [493] [494] 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. Crouch PJ, Barnham KJ (2012) Therapeutic redistribution of metal ions to treat Alzheimer’s disease. Acc Chem Res 45, 1604-1611. Grasso G, Giuffrida ML, Rizzarelli E (2012) Metallostasis and amyloid beta-degrading enzymes. Metallomics 4, 937949. Hung LW, Barnham KJ (2012) Modulating metals as a therapeutic strategy for Alzheimer’s disease. Future Med Chem 4, 955-969. Squitti R (2012) Metals in Alzheimer’s disease: A systemic perspective. Front Biosci 17, 451-472. Watt AD, Villemagne VL, Barnham KJ (2012) Metals, membranes, and amyloid-beta oligomers: Key pieces in the Alzheimer’s disease puzzle? J Alzheimers Dis, doi: 10.3233/JAD-2012-129017 [Epub ahead of print]. Craddock TJ, Tuszynski JA, Chopra D, Casey N, Goldstein LE, Hameroff SR, Tanzi RE (2012) The zinc dyshomeostasis hypoothesis of Alzheimer’s disease. PloS ONE, 7, 3. Wise-Scira O, Xu L, Perry G, Coskuner O (2012) Structures and free energy landscapes of aqueous zinc(II)-bound amyloid-beta(1-40) and zinc(II)-bound amyloid-beta(142) with dynamics. J Biol Inorg Chem 17, 927-938. Rosenberg G, Angel I, Kozak A (2005) Clinical pharmacology of DP-b99 in healthy volunteers: First administration to humans. Br J Clin Pharmacol 60, 7-16. Diener HC, Schneider D, Lampl Y, Bornstein NM, Kozak A, Rosenberg G (2008) DP-b99, a membrane-activated metal ion chelator, as neuroprotective therapy in ischemic stroke. Stroke 39, 1774-1778. Rosenberg G, Bornstein N, Diener HC, Gorelick PB, Shuaib A, Lees K (2011) The Membrane-Activated Chelator Stroke Intervention (MACSI) Trial of DP-b99 in acute ischemic stroke: A randomized, double-blind, placebocontrolled, multinational pivotal phase III study. Int J Stroke 6, 362-367. Angel I, Bar A, Horovitz T, Taler G, Krakovsky M, Resnitsky D, Rosenberg G, Striem S, Friedman JE, Kozak A (2002) Metal ion chelation in neurodegenerative disorders. Drug Dev Res 56, 300-309. Yeung PK (2004) DP-b99 (D-Pharm). Curr Opin Investig Drugs 5, 90-94. Barkalifa R, Hershfinkel M, Friedman JE, Kozak A, Sekler I (2009) The lipophilic zinc chelator DP-b99 prevents zinc induced neuronal death. Eur J Pharmacol 618, 15-21. Duce JA, Tsatsanis A, Cater MA, James SA, Robb E, Wikhe K, Leong SL, Perez K, Johanssen T, Greenough MA, Cho HH, Galatis D, Moir RD, Masters CL, McLean C, Tanzi RE, Cappai R, Barnham KJ, Ciccotosto GD, Rogers JT, Bush AI (2010) Iron-export ferroxidase activity of beta-amyloid precursor protein is inhibited by zinc in Alzheimer’s disease. Cell 142, 857-867. Lannfelt L, Blennow K, Zetterberg H, Batsman S, Ames D, Harrison J, Masters CL, Targum S, Bush AI, Murdoch R, Wilson J, Ritchie CW (2008) Safety, efficacy, and biomarker findings of PBT2 in targeting Abeta as a modifying therapy for Alzheimer’s disease: A phase IIa, double-blind, randomised, placebo-controlled trial. Lancet Neurol 7, 779-786. Faux NG, Ritchie CW, Gunn A, Rembach A, Tsatsanis A, Bedo J, Harrison J, Lannfelt L, Blennow K, Zetterberg H, Ingelsson M, Masters CL, Tanzi RE, Cummings JL, [495] [496] [497] [498] [499] [500] [501] [502] [503] [504] [505] [506] [507] 71 Herd CM, Bush AI (2010) PBT2 rapidly improves cognition in Alzheimer’s Disease: Additional phase II analyses. J Alzheimers Dis 20, 509-516. Adlard PA, James SA, Bush AI, Masters CL (2009) betaAmyloid as a molecular therapeutic target in Alzheimer’s disease. Drugs Today (Barc) 45, 293-304. Adlard PA, Bica L, White AR, Nurjono M, Filiz G, Crouch PJ, Donnelly PS, Cappai R, Finkelstein DI, Bush AI (2011) Metal ionophore treatment restores dendritic spine density and synaptic protein levels in a mouse model of Alzheimer’s disease. PLoS One 6, e17669. Crouch PJ, Savva MS, Hung LW, Donnelly PS, Mot AI, Parker SJ, Greenough MA, Volitakis I, Adlard PA, Cherny RA, Masters CL, Bush AI, Barnham KJ, White AR (2011) The Alzheimer’s therapeutic PBT2 promotes amyloid-beta degradation and GSK3 phosphorylation via a metal chaperone activity. J Neurochem 119, 220-230. Lee JY, Friedman JE, Angel I, Kozak A, Koh JY (2004) The lipophilic metal chelator DP-109 reduces amyloid pathology in brains of human beta-amyloid precursor protein transgenic mice. Neurobiol Aging 25, 1315-1321. Petri S, Calingasan NY, Alsaied OA, Wille E, Kiaei M, Friedman JE, Baranova O, Chavez JC, Beal MF (2007) The lipophilic metal chelators DP-109 and DP-460 are neuroprotective in a transgenic mouse model of amyotrophic lateral sclerosis. J Neurochem 102, 991-1000. 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 deferoxamine improves performance in radial arm water maze, stabilizes HIF-1alpha, and phosphorylates GSK3beta in P301L tau transgenic mice. Exp Brain Res 219, 381-390. Zheng H, Weiner LM, Bar-Am O, Epsztejn S, Cabantchik ZI, Warshawsky A, Youdim MB, Fridkin M (2005) Design, synthesis, and evaluation of novel bifunctional iron-chelators as potential agents for neuroprotection in Alzheimer’s, Parkinson’s, and other neurodegenerative diseases. Bioorg Med Chem 13, 773-783. Zheng H, Youdim MB, Fridkin M (2009) Site-activated multifunctional chelator with acetylcholinesterase and neuroprotective-neurorestorative moieties for Alzheimer’s therapy. J Med Chem 52, 4095-4098. Jones MR, Service EL, Thompson JR, Wang MC, Kimsey IJ, Detoma AS, Ramamoorthy A, Lim MH, Storr T (2012) Dual-function triazole-pyridine derivatives as inhibitors of metal-induced amyloid-beta aggregation. Metallomics 4, 910-920. Youdim MB, Fridkin M, Zheng H (2005) Bifunctional drug derivatives of MAO-B inhibitor rasagiline and iron chelator VK-28 as a more effective approach to treatment of brain ageing and ageing neurodegenerative diseases. Mech Ageing Dev 126, 317-326. Youdim MB (2006) The path from anti Parkinson drug selegiline and rasagiline to multifunctional neuroprotective anti Alzheimer drugs ladostigil and m30. Curr Alzheimer Res 3, 541-550. Gal S, Fridkin M, Amit T, Zheng H, Youdim MB (2006) M30, a novel multifunctional neuroprotective drug with potent iron chelating and brain selective monoamine oxidase-ab inhibitory activity for Parkinson’s disease. J Neural Transm Suppl 70, 447-456. Avramovich-Tirosh Y, Amit T, Bar-Am O, Zheng H, Fridkin M, Youdim MB (2007) Therapeutic targets and potential of the novel brain- permeable multifunctional iron chelator-monoamine oxidase inhibitor drug, M-30, 72 [508] [509] [510] [511] [512] [513] [514] [515] [516] [517] [518] [519] [520] [521] 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. Avramovich-Tirosh Y, Reznichenko L, Mit T, Zheng H, Fridkin M, Weinreb O, Mandel S, Youdim MB (2007) Neurorescue activity, APP regulation and amyloid-beta peptide reduction by novel multi-functional brain permeable ironchelating- antioxidants, M-30 and green tea polyphenol, EGCG. Curr Alzheimer Res 4, 403-411. Weinreb O, Mandel S, Bar-Am O, Yogev-Falach M, Avramovich-Tirosh Y, Amit T, Youdim MB (2009) Multifunctional neuroprotective derivatives of rasagiline as anti-Alzheimer’s disease drugs. Neurotherapeutics 6, 163174. Weinreb O, Mandel S, Bar-Am O, Amit T (2011) Iron-chelating backbone coupled with monoamine oxidase inhibitory moiety as novel pluripotential therapeutic agents for Alzheimer’s disease: A tribute to Moussa Youdim. J Neural Transm 118, 479-492. Bar-Am O, Amit T, Weinreb O, Youdim MB, Mandel S (2010) Propargylamine containing compounds as modulators of proteolytic cleavage of amyloid-beta protein precursor: Involvement of MAPK and PKC activation. J Alzheimers Dis 21, 361-371. Johnson S, Tazik S, Lu D, Johnson C, Youdim MB, Wang J, Rajkowska G, Ou XM (2010) The new inhibitor of monoamine oxidase, M30, has a neuroprotective effect against dexamethasone-induced brain cell apoptosis. Front Neurosci 4, 180. Zheng H, Fridkin M, Youdim MB (2010) Site-activated chelators derived from anti-Parkinson drug rasagiline as a potential safer and more effective approach to the treatment of Alzheimer’s disease. Neurochem Res 35, 2117-2123. Kupershmidt L, Amit T, Bar-Am O, Weinreb O, Youdim MB (2012) Multi-target, neuroprotective and neurorestorative M30 improves cognitive impairment and reduces Alzheimer’s-like neuropathology and age-related alterations in mice. Mol Neurobiol 46, 217-220. Kupershmidt L, Weinreb O, Amit T, Mandel S, Bar-Am O, Youdim MB (2011) Novel molecular targets of the neuroprotective/neurorescue multimodal iron chelating drug M30 in the mouse brain. Neuroscience 189, 345-358. Kupershmidt L, Amit T, Bar-Am O, Youdim MB, Weinreb O (2012) Neuroprotection by the multitarget iron chelator M30 on age-related alterations in mice. Mech Ageing Dev 133, 267-274. Kupershmidt L, Amit T, Bar-Am O, Youdim MB, Weinreb O (2012) The novel multi-target iron chelating-radical scavenging compound M30 possesses beneficial effects on major hallmarks of Alzheimer’s disease. Antioxid Redox Signal 17, 860-877. Crapper McLachlan DR, Dalton AJ, Kruck TP, Bell MY, Smith WL, Kalow W, Andrews DF (1991) Intramuscular desferrioxamine in patients with Alzheimer’s disease. Lancet 337, 1304-1308. Cuajungco MP, Faget KY, Huang X, Tanzi RE, Bush AI (2000) Metal chelation as a potential therapy for Alzheimer’s disease. Ann N Y Acad Sci 920, 292-304. de Lima MN, Presti-Torres J, Caldana F, Grazziotin MM, Scalco FS, Guimaraes MR, Bromberg E, Franke SI, Henriques JA, Schroder N (2007) Desferoxamine reverses neonatal iron-induced recognition memory impairment in rats. Eur J Pharmacol 570, 111-114. Cherny RA, Atwood CS, Xilinas ME, Gray DN, Jones WD, McLean CA, Barnham KJ, Volitakis I, Fraser FW, Kim Y, Huang X, Goldstein LE, Moir RD, Lim [522] [523] [524] [525] [526] [527] [528] [529] [530] [531] [532] [533] [534] [535] [536] JT, Beyreuther K, Zheng H, Tanzi RE, Masters CL, Bush AI (2001) Treatment with a copper-zinc chelator markedly and rapidly inhibits beta-amyloid accumulation in Alzheimer’s disease transgenic mice. Neuron 30, 665676. Ibach B, Haen E, Marienhagen J, Hajak G (2005) Clioquinol treatment in familiar early onset of Alzheimer’s disease: A case report. Pharmacopsychiatry 38, 178-179. Shachar DB, Kahana N, Kampel V, Warshawsky A, Youdim MB (2004) Neuroprotection by a novel brain permeable iron chelator, VK-28, against 6-hydroxydopamine lession in rats. Neuropharmacology 46, 254-263. Youdim MB, Stephenson G, Ben SD (2004) Ironing iron out in Parkinson’s disease and other neurodegenerative diseases with iron chelators: A lesson from 6-hydroxydopamine and iron chelators, desferal and VK28. Ann N Y Acad Sci 1012, 306-325. Bandyopadhyay S, Huang X, Cho H, Greig NH, Youdim MB, Rogers JT (2006) Metal specificity of an iron-responsive element in Alzheimer’s APP mRNA 5’untranslated region, tolerance of SH-SY5Y and H4 neural cells to desferrioxamine, clioquinol, VK-28, and a piperazine chelator. J Neural Transm Suppl 71, 237247. Weinreb O, Mandel S, Bar-Am O, Yogev-Falach M, Vramovich-Tirosh Y, Amit T, Youdim MB (2009) Multifunctional neuroprotective derivatives of rasagiline as anti-Alzheimer’s disease drugs. Neurotherapeutics 6, 163174. Akhondzadeh S, Abbasi SH (2006) Herbal medicine in the treatment of Alzheimer’s disease. Am J Alzheimers Dis Other Demen 21, 113-118. Man SC, Durairajan SS, Kum WF, Lu JH, Huang JD, Cheng CF, Chung V, Xu M, Li M (2008) Systematic review on the efficacy and safety of herbal medicines for Alzheimer’s disease. J Alzheimers Dis 14, 209-223. Kim J, Lee HJ, Lee KW (2010) Naturally occurring phytochemicals for the prevention of Alzheimer’s disease. J Neurochem 112, 1415-1430. Kennedy DO, Wightman EL (2011) Herbal extracts and phytochemicals: Plant secondary metabolites and the enhancement of human brain function. Adv Nutr (Bethesda) 2, 32-50. Perry E, Howes MJ (2011) Medicinal plants and dementia therapy: Herbal hopes for brain aging? CNS Neurosci Ther 17, 683-698. Kim HG, Oh MS (2012) Herbal medicines for the prevention and treatment of Alzheimer’s disease. Curr Pharm Des 18, 57-75. Mancuso C, Siciliano R, Barone E, Preziosi P (2012) Natural substances and Alzheimer’s disease: From preclinical studies to evidence based medicine. Biochim Biophys Acta 1822, 616-624. Yoshitake T, Yoshitake S, Kehr J (2010) The Ginkgo biloba extract EGb 761(R) and its main constituent flavonoids and ginkgolides increase extracellular dopamine levels in the rat prefrontal cortex. Br J Pharmacol 159, 659-668. Han CK, Park YH, Jin DQ, Hwang YK, Oh KB, Han JS (2007) SK-PC-B70M from Pulsatilla koreana improves scopolamine-induced impairments of memory consolidation and spatial working memory. Brain Res 1184, 254-259. Han CK, Choi WR, Oh KB (2007) Cognition-enhancing and neuroprotective effects of hederacolchiside-E from Pulsatilla koreana. Planta Med 73, 665-669. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [537] [538] [539] [540] [541] [542] [543] [544] [545] [546] [547] [548] [549] [550] [551] Seo JS, Kim TK, Leem YH, Lee KW, Park SK, Baek IS, Kim KS, Im GJ, Lee SM, Park YH, Han PL (2009) SKPC-B70M confers anti-oxidant activity and reduces Abeta levels in the brain of Tg2576 mice. Brain Res 1261, 100108. Seo JS, Baek IS, Leem YH, Kim TK, Cho Y, Lee SM, Park YH, Han PL (2011) SK-PC-B70M alleviates neurologic symptoms in G93A-SOD1 amyotrophic lateral sclerosis mice. Brain Res 1368, 299-307. Baur JA, Sinclair DA (2006) Therapeutic potential of resveratrol: The in vivo evidence. Nat Rev Drug Discov 5, 493-506. 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, Wang M, Ramaswamy S, Fishbein KW, Spencer RG, Lakatta EG, Le CD, Shaw RJ, Navas P, Puigserver P, Ingram DK, de CR, Sinclair DA (2006) Resveratrol improves health and survival of mice on a high-calorie diet. Nature 444, 337-342. Baur JA (2010) Resveratrol, sirtuins, and the promise of a DR mimetic. Mech Ageing Dev 131, 261-269. Baur JA, Chen D, Chini EN, Chua K, Cohen HY, de CR, Deng C, Dimmeler S, Gius D, Guarente LP, Helfand SL, Imai S, Itoh H, Kadowaki T, Koya D, Leeuwenburgh C, McBurney M, Nabeshima Y, Neri C, Oberdoerffer P, Pestell RG, Rogina B, Sadoshima J, Sartorelli V, Serrano M, Sinclair DA, Steegborn C, Tatar M, Tissenbaum HA, Tong Q, Tsubota K, Vaquero A, Verdin E (2010) Dietary restriction: Standing up for sirtuins. Science 329, 10121013. Kennedy DO, Wightman EL, Reay JL, Lietz G, Okello EJ, Wilde A, Haskell CF (2010) Effects of resveratrol on cerebral blood flow variables and cognitive performance in humans: A double-blind, placebo-controlled, crossover investigation. Am J Clin Nutr 91, 1590-1597. Quideau S, Deffieux D, Pouysegu L (2012) Resveratrol still has something to say about aging! Angew Chem Int Ed Engl 51, 6824-6826. Marambaud P, Zhao H, Davies P (2005) Resveratrol promotes clearance of Alzheimer’s disease amyloid-beta peptides. J Biol Chem 280, 37377-37382. Albani D, Polito L, Batelli S, De MS, Fracasso C, Martelli G, Colombo L, Manzoni C, Salmona M, Caccia S, Negro A, Forloni G (2009) The SIRT1 activator resveratrol protects SK-N-BE cells from oxidative stress and against toxicity caused by alpha-synuclein or amyloid-beta (1-42) peptide. J Neurochem 110, 1445-1456. Albani D, Polito L, Forloni G (2010) Sirtuins as novel targets for Alzheimer’s disease and other neurodegenerative disorders: Experimental and genetic evidence. J Alzheimers Dis 19, 11-26. Albani D, Polito L, Signorini A, Forloni G (2010) Neuroprotective properties of resveratrol in different neurodegenerative disorders. Biofactors 36, 370-376. Ladiwala AR, Lin JC, Bale SS, Marcelino-Cruz AM, Bhattacharya M, Dordick JS, Tessier PM (2010) Resveratrol selectively remodels soluble oligomers and fibrils of amyloid Abeta into off-pathway conformers. J Biol Chem 285, 24228-24237. Beher D, Wu J, Cumine S, Kim KW, Lu SC, Atangan L, Wang M (2009) Resveratrol is not a direct activator of SIRT1 enzyme activity. Chem Biol Drug Des 74, 619-624. Jeon BT, Jeong EA, Shin HJ, Lee Y, Lee DH, Kim HJ, Kang SS, Cho GJ, Choi WS, Roh GS (2012) Resvera- [552] [553] [554] [555] [556] [557] [558] [559] [560] [561] [562] [563] [564] [565] [566] 73 trol attenuates obesity-associated peripheral and central inflammation and improves memory deficit in mice fed a high-fat diet. Diabetes 61, 1444-1454. Johnson WD, Morrissey RL, Usborne AL, Kapetanovic I, Crowell JA, Muzzio M, McCormick DL (2011) Subchronic oral toxicity and cardiovascular safety pharmacology studies of resveratrol, a naturally occurring polyphenol with cancer preventive activity. Food Chem Toxicol 49, 3319-3327. Zhang Y, Li SZ, Li J, Pan X, Cahoon RE, Jaworski JG, Wang X, Jez JM, Chen F, Yu O (2006) Using unnatural protein fusions to engineer resveratrol biosynthesis in yeast and Mammalian cells. J Am Chem Soc 128, 13030-13031. Pasinetti GM, Wang J, Marambaud P, Ferruzzi M, Gregor P, Knable LA, Ho L (2011) Neuroprotective and metabolic effects of resveratrol: Therapeutic implications for Huntington’s disease and other neurodegenerative disorders. Exp Neurol 232, 1-6. Wang Y, Xia Z, Xu JR, Wang YX, Hou LN, Qiu Y, Chen HZ (2012) Alpha-mangostin, a polyphenolic xanthone derivative from mangosteen, attenuates betaamyloid oligomers-induced neurotoxicity by inhibiting amyloid aggregation. Neuropharmacology 62, 871-881. Paris D, Beaulieu-Abdelahad D, Bachmeier C, Reed J, itGhezala G, Bishop A, Chao J, Mathura V, Crawford F, Mullan M (2011) Anatabine lowers Alzheimer’s Abeta production in vitro and in vivo. Eur J Pharmacol 670, 384-391. Radhika P, Annapurna A, Rao SN (2012) Immunostimulant, cerebroprotective & nootropic activities of Andrographis paniculata leaves extract in normal & type 2 diabetic rats. Indian J Med Res 135, 636-641. Lashuel HA, Hartley DM, Balakhaneh D, Aggarwal A, Teichberg S, Callaway DJ (2002) New class of inhibitors of amyloid-beta fibril formation. Implications for the mechanism of pathogenesis in Alzheimer’s disease. J Biol Chem 277, 42881-42890. Himeno E, Ohyagi Y, Ma L, Nakamura N, Miyoshi K, Sakae N, Motomura K, Soejima N, Yamasaki R, Hashimoto T, Tabira T, LaFerla FM, Kira J (2011) Apomorphine treatment in Alzheimer mice promoting amyloid-beta degradation. Ann Neurol 69, 248-256. Steele JW, Gandy S (2011) Apomorphine and Alzheimer Abeta: Roles for regulated alpha cleavage, autophagy, and antioxidation? Ann Neurol 69, 221-225. Howland RH (2010) Drug therapies for cognitive impairment and dementia. J Psychosoc Nurs Ment Health Serv 48, 11-14. Roman MW (2010) Axona (Accera, Inc): A new medical food therapy for persons with Alzheimer’s disease. Issues Ment Health Nurs 31, 435-436. Reger MA, Henderson ST, Hale C, Cholerton B, Baker LD, Watson GS, Hyde K, Chapman D, Craft S (2004) Effects of beta-hydroxybutyrate on cognition in memory-impaired adults. Neurobiol Aging 25, 311-314. Costantini LC, Barr LJ, Vogel JL, Henderson ST (2008) Hypometabolism as a therapeutic target in Alzheimer’s disease. BMC Neurosci 9(Suppl 2), S16. Pocernich CB, Lange ML, Sultana R, Butterfield DA (2011) Nutritional approaches to modulate oxidative stress in Alzheimer’s disease. Curr Alzheimer Res 8, 452469. Pase MP, Kean J, Sarris J, Neale C, Scholey AB, Stough C (2012) The cognitive-enhancing effects of Bacopa monnieri: A systematic review of randomized, controlled 74 [567] [568] [569] [570] [571] [572] [573] [574] [575] [576] [577] [578] [579] [580] [581] 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, 647652. Saini N, Singh D, Sandhir R (2012) Neuroprotective effects of Bacopa monnieri in experimental model of dementia. Neurochem Res 37, 1928-1937. Lebeau A, Esclaire F, Rostene W, Pelaprat D (2001) Baicalein protects cortical neurons from beta-amyloid (2535) induced toxicity. Neuroreport 12, 2199-2202. 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. Yin F, Liu J, Ji X, Wang Y, Zidichouski J, Zhang J (2011) Baicalin prevents the production of hydrogen peroxide and oxidative stress induced by Abeta aggregation in SHSY5Y cells. Neurosci Lett 492, 76-79. Gauci AJ, Caruana M, Giese A, Scerri C, Vassallo N (2011) Identification of polyphenolic compounds and black tea extract as potent inhibitors of lipid membrane destabilization by abeta42 aggregates. J Alzheimers Dis 27, 767-779. Lu JH, Ardah MT, Durairajan SS, Liu LF, Xie LX, Fong WF, Hasan MY, Huang JD, El-Agnaf OM, Li M (2011) Baicalein inhibits formation of alpha-synuclein oligomers within living cells and prevents Abeta peptide fibrillation and oligomerisation. Chembiochem 12, 615-624. Geng Y, Li C, Liu J, Xing G, Zhou L, Dong M, Li X, Niu Y (2010) Beta-asarone improves cognitive function by suppressing neuronal apoptosis in the beta-amyloid hippocampus injection rats. Biol Pharm Bull 33, 836-843. Li C, Xing G, Dong M, Zhou L, Li J, Wang G, Zou D, Wang R, Liu J, Niu Y (2010) Beta-asarone protection against beta-amyloid-induced neurotoxicity in PC12 cells via JNK signaling and modulation of Bcl-2 family proteins. Eur J Pharmacol 635, 96-102. 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 attenuates neuronal apoptosis induced by Beta amyloid in rat hippocampus. Yakugaku Zasshi 130, 737-746. 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. 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 memory impairments through increment of glucose utilization and total neural cell adhesion molecule levels in rat brains. J Neurosci Res 87, 260-268. Shin KY, Lee JY, Won BY, Jung HY, Chang KA, Koppula S, Suh YH (2009) BT-11 is effective for enhancing cognitive functions in the elderly humans. Neurosci Lett 465, 157-159. Wang J, Ho L, Zhao Z, Seror I, Humala N, Dickstein DL, Thiyagarajan M, Percival SS, Talcott ST, Pasinetti GM (2006) Moderate consumption of Cabernet Sauvignon attenuates Abeta neuropathology in a mouse model of Alzheimer’s disease. FASEB J 20, 2313-2320. Azizi Z, Ebrahimi S, Saadatfar E, Kamalinejad M, Majlessi N (2012) Cognitive-enhancing activity of thymol and carvacrol in two rat models of dementia. Behav Pharmacol 23, 241-249. Ono K, Yoshiike Y, Takashima A, Hasegawa K, Naiki H, Yamada M (2003) Potent anti-amyloidogenic and fibrildestabilizing effects of polyphenols in vitro: Implications for the prevention and therapeutics of Alzheimer’s disease. J Neurochem 87, 172-181. [582] [583] [584] [585] [586] [587] [588] [589] [590] [591] [592] [593] [594] [595] [596] Mandel SA, Amit T, Kalfon L, Reznichenko L, Youdim MB (2008) Targeting multiple neurodegenerative diseases etiologies with multimodal-acting green tea catechins. J Nutr 138, 1578S-1583S. 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 grape seed polyphenol extract is improved by repeated dosing in rats: Implications for treatment in Alzheimer’s disease. J Alzheimers Dis 18, 113-124. Paris D, Ganey NJ, Laporte V, Patel NS, BeaulieuAbdelahad D, Bachmeier C, March A, it-Ghezala G, Mullan MJ (2010) Reduction of beta-amyloid pathology by celastrol in a transgenic mouse model of Alzheimer’s disease. J Neuroinflammation 7, 17. Bhanumathy M, Harish MS, Shivaprasad HN, Sushma G (2010) Nootropic activity of Celastrus paniculatus seed. Pharm Biol 48, 324-327. 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 cinnamon extract reduces beta-amyloid oligomerization and corrects cognitive impairment in Alzheimer’s disease animal models. PLoS One 6, e16564. Marumoto S, Miyazawa M (2012) Structure-activity relationships for naturally occurring coumarins as betasecretase inhibitor. Bioorg Med Chem 20, 784-788. Anand P, Singh B, Singh N (2012) A review on coumarins as acetylcholinesterase inhibitors for Alzheimer’s disease. Bioorg Med Chem 20, 1175-1180. Mei Z, Yan P, Situ B, Mou Y, Liu P (2012) Cryptotanshinione inhibits beta-amyloid aggregation and protects damage from beta-amyloid in SH-SY5Y cells. Neurochem Res 37, 622-628. Mei Z, Situ B, Tan X, Zheng S, Zhang F, Yan P, Liu P (2010) Cryptotanshinione upregulates alpha-secretase by activation PI3K pathway in cortical neurons. Brain Res 1348, 165-173. Jiang T, Zhi XL, Zhang YH, Pan LF, Zhou P (2012) Inhibitory effect of curcumin on the Al(III)-induced Abeta(42) aggregation and neurotoxicity in vitro. Biochim Biophys Acta 1822, 1207-1215. Lim GP, Chu T, Yang F, Beech W, Frautschy SA, Cole GM (2001) The curry spice curcumin reduces oxidative damage and amyloid pathology in an Alzheimer transgenic mouse. J Neurosci 21, 8370-8377. Ono K, Hasegawa K, Naiki H, Yamada M (2004) Curcumin has potent anti-amyloidogenic effects for Alzheimer’s beta-amyloid fibrils in vitro. J Neurosci Res 75, 742-750. Ringman JM, Frautschy SA, Cole GM, Masterman DL, Cummings JL (2005) A potential role of the curry spice curcumin in Alzheimer’s disease. Curr Alzheimer Res 2, 131-136. Yang F, Lim GP, Begum AN, Ubeda OJ, Simmons MR, Ambegaokar SS, Chen PP, Kayed R, Glabe CG, Frautschy SA, Cole GM (2005) Curcumin inhibits formation of amyloid beta oligomers and fibrils, binds plaques, and reduces amyloid in vivo. J Biol Chem 280, 58925901. Garcia-Alloza M, Borrelli LA, Rozkalne A, Hyman BT, Bacskai BJ (2007) Curcumin labels amyloid pathology in vivo, disrupts existing plaques, and partially restores distorted neurites in an Alzheimer mouse model. J Neurochem 102, 1095-1104. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [597] [598] [599] [600] [601] [602] [603] [604] [605] [606] [607] [608] [609] [610] [611] Begum AN, Jones MR, Lim GP, Morihara T, Kim P, Heath DD, Rock CL, Pruitt MA, Yang F, Hudspeth B, Hu S, Faull KF, Teter B, Cole GM, Frautschy SA (2008) Curcumin structure-function, bioavailability, and efficacy in models of neuroinflammation and Alzheimer’s disease. J Pharmacol Exp Ther 326, 196-208. Park SY, Kim HS, Cho EK, Kwon BY, Phark S, Hwang KW, Sul D (2008) Curcumin protected PC12 cells against beta-amyloid-induced toxicity through the inhibition of oxidative damage and tau hyperphosphorylation. Food Chem Toxicol 46, 2881-2887. Hamaguchi T, Ono K, Murase A, Yamada M (2009) Phenolic compounds prevent Alzheimer’s pathology through different effects on the amyloid-beta aggregation pathway. Am J Pathol 175, 2557-2565. Hamaguchi T, Ono K, Yamada M (2010) REVIEW: Curcumin and Alzheimer’s disease. CNS Neurosci Ther 16, 285-297. Ma QL, Yang F, Rosario ER, Ubeda OJ, Beech W, Gant DJ, Chen PP, Hudspeth B, Chen C, Zhao Y, Vinters HV, Frautschy SA, Cole GM (2009) Beta-amyloid oligomers induce phosphorylation of tau and inactivation of insulin receptor substrate via c-Jun N-terminal kinase signaling: Suppression by omega-3 fatty acids and curcumin. J Neurosci 29, 9078-9089. Ray B, Lahiri DK (2009) Neuroinflammation in Alzheimer’s disease: Different molecular targets and potential therapeutic agents including curcumin. Curr Opin Pharmacol 9, 434-444. Zhang C, Browne A, Child D, Tanzi RE (2010) Curcumin decreases amyloid-beta peptide levels by attenuating the maturation of amyloid-beta precursor protein. J Biol Chem 285, 28472-28480. Ferrari E, Pignedoli F, Imbriano C, Marverti G, Basile V, Venturi E, Saladini M (2011) Newly synthesized curcumin derivatives: Crosstalk between chemico-physical properties and biological activity. J Med Chem 54, 8066-8077. Gupta SC, Prasad S, Kim JH, Patchva S, Webb LJ, Priyadarsini IK, Aggarwal BB (2011) Multitargeting by curcumin as revealed by molecular interaction studies. Nat Prod Rep 28, 1937-1955. Mancuso C, Siciliano R, Barone E (2011) Curcumin and Alzheimer disease: This marriage is not to be performed. J Biol Chem 286, le3. Ray B, Bisht S, Maitra A, Maitra A, Lahiri DK (2011) Neuroprotective and neurorescue effects of a novel polymeric nanoparticle formulation of curcumin (NanoCurc) in the neuronal cell culture and animal model: Implications for Alzheimer’s disease. J Alzheimers Dis 23, 61-77. Mourtas S, Canovi M, Zona C, Aurilia D, Niarakis A, La FB, Salmona M, Nicotra F, Gobbi M, Antimisiaris SG (2011) Curcumin-decorated nanoliposomes with very high affinity for amyloid-beta1-42 peptide. Biomaterials 32, 1635-1645. Reinke AA, Gestwicki JE (2007) Structure-activity relationships of amyloid beta-aggregation inhibitors based on curcumin: Influence of linker length and flexibility. Chem Biol Drug Des 70, 206-215. Narlawar R, Pickhardt M, Leuchtenberger S, Baumann K, Krause S, Dyrks T, Weggen S, Mandelkow E, Schmidt B (2008) Curcumin-derived pyrazoles and isoxazoles: Swiss army knives or blunt tools for Alzheimer’s disease? ChemMedChem 3, 165-172. Orlando RA, Gonzales AM, Royer RE, Deck LM, Vander Jagt DL (2012) A chemical analog of curcumin as [612] [613] [614] [615] [616] [617] [618] [619] [620] [621] [622] [623] [624] 75 an improved inhibitor of amyloid Abeta oligomerization. PLoS One 7, e31869. Zhang Y, Saito H, Nishiyama N, Abe K (1994) Effects of DX-9386, a traditional Chinese medicinal prescription, on long-term potentiation in the dentate gyrus in rats. Biol Pharm Bull 17, 1337-1340 Yang SG, Zhang X, Sun XS, Ling TJ, Feng Y, Du XY, Zhao M, Yang Y, Xue D, Wang L, Liu RT (2010) Diverse ecdysterones show different effects on amyloid-beta42 aggregation but all uniformly inhibit amyloid-beta42induced cytotoxicity. J Alzheimers Dis 22, 107-117. Rezai-Zadeh K, Shytle D, Sun N, Mori T, Hou H, Jeanniton 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 cleavage and reduces cerebral amyloidosis in Alzheimer transgenic mice. J Neurosci 25, 8807-8814. Rezai-Zadeh K, Arendash GW, Hou H, Fernandez F, Jensen M, Runfeldt M, Shytle RD, Tan J (2008) Green tea epigallocatechin-3-gallate (EGCG) reduces beta-amyloid mediated cognitive impairment and modulates tau pathology in Alzheimer transgenic mice. Brain Res 1214, 177-187. 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 reduces Alzheimer’s disease beta-amyloid production. J Cell Mol Med 13, 574-588. Lee JW, Lee YK, Ban JO, Ha TY, Yun YP, Han SB, Oh KW, Hong JT (2009) Green tea (–)-epigallocatechin3-gallate inhibits beta-amyloid-induced cognitive dysfunction through modification of secretase activity via inhibition of ERK and NF-kappaB pathways in mice. J Nutr 139, 1987-1993. Bieschke J, Russ J, Friedrich RP, Ehrnhoefer DE, Wobst H, Neugebauer K, Wanker EE (2010) EGCG remodels mature alpha-synuclein and amyloid-beta fibrils and reduces cellular toxicity. Proc Natl Acad Sci U S A 107, 7710-7715. Mandel SA, Avramovich-Tirosh Y, Reznichenko L, Zheng H, Weinreb O, Amit T, Youdim MB (2005) Multifunctional activities of green tea catechins in neuroprotection. Modulation of cell survival genes, iron-dependent oxidative stress and PKC signaling pathway. Neurosignals 14, 46-60. Mandel SA, Amit T, Weinreb O, Reznichenko L, Youdim MB (2008) Simultaneous manipulation of multiple brain targets by green tea catechins: A potential neuroprotective strategy for Alzheimer and Parkinson diseases. CNS Neurosci Ther 14, 352-365. Mandel SA, Amit T, Kalfon L, Reznichenko L, Weinreb O, Youdim MB (2008) Cell signaling pathways and iron chelation in the neurorestorative activity of green tea polyphenols: Special reference to epigallocatechin gallate (EGCG). J Alzheimers Dis 15, 211-222. Mandel SA, Amit T, Weinreb O, Youdim MB (2011) Understanding the broad-spectrum neuroprotective action profile of green tea polyphenols in aging and neurodegenerative diseases. J Alzheimers Dis 25, 187-208. Mandel SA, Weinreb O, Amit T, Youdim MB (2012) The importance of the multiple target action of green tea polyphenols for neuroprotection. Front Biosci (Schol Ed) 4, 581-598. Kalfon L, Youdim MB, Mandel SA (2007) Green tea polyphenol (–) -epigallocatechin-3-gallate promotes the rapid protein kinase C- and proteasome-mediated 76 [625] [626] [627] [628] [629] [630] [631] [632] [633] [634] [635] [636] [637] [638] 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. Lopez Del Amo JM, Fink U, Dasari M, Grelle G, Wanker EE, Bieschke J, Reif B (2012) Structural properties of EGCG-induced, nontoxic Alzheimer’s disease Abeta oligomers. J Mol Biol 421, 517-524. Fernandez JW, Rezai-Zadeh K, Obregon D, Tan J (2010) EGCG functions through estrogen receptormediated activation of ADAM10 in the promotion of non-amyloidogenic processing of APP. FEBS Lett 584, 4259-4267. Obregon DF, Rezai-Zadeh K, Bai Y, Sun N, Hou H, Ehrhart J, Zeng J, Mori T, Arendash GW, Shytle D, Town T, Tan J (2006) ADAM10 activation is required for green tea (–)-epigallocatechin-3-gallate-induced alpha-secretase cleavage of amyloid precursor protein. J Biol Chem 281, 16419-16427. Smith A, Giunta B, Bickford PC, Fountain M, Tan J, Shytle RD (2010) Nanolipidic particles improve the bioavailability and alpha-secretase inducing ability of epigallocatechin-3-gallate (EGCG) for the treatment of Alzheimer’s disease. Int J Pharm 389, 207-212. Kang SY, Lee KY, Koo KA, Yoon JS, Lim SW, Kim YC, Sung SH (2005) ESP-102, a standardized combined extract of Angelica gigas, Saururus chinensis and Schizandra chinensis, significantly improved scopolamine-induced memory impairment in mice. Life Sci 76, 1691-1705. Kim DH, Jung WY, Park SJ, Kim JM, Lee S, Kim YC, Ryu JH (2010) Anti-amnesic effect of ESP-102 on Abeta(1-42)-induced memory impairment in mice. Pharmacol Biochem Behav 97, 239-248. 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. Bark on amyloid beta(25-35)-induced learning and memory impairments in mice. Neurosci Lett 487, 123-127. Williams RJ, Spencer JP (2012) Flavonoids, cognition, and dementia: Actions, mechanisms, and potential therapeutic utility for Alzheimer disease. Free Radic Biol Med 52, 35-45. Spencer JP, Vafeiadou K, Williams RJ, Vauzour D (2012) Neuroinflammation: Modulation by flavonoids and mechanisms of action. Mol Aspects Med 33, 83-97. Paris D, Mathura V, it-Ghezala G, Beaulieu-Abdelahad D, Patel N, Bachmeier C, Mullan M (2011) Flavonoids lower Alzheimer’s Abeta production via an NFkappaB dependent mechanism. Bioinformation 6, 229-236. Zhu JT, Choi RC, Chu GK, Cheung AW, Gao QT, Li J, Jiang ZY, Dong TT, Tsim KW (2007) Flavonoids possess neuroprotective effects on cultured pheochromocytoma PC12 cells: A comparison of different flavonoids in activating estrogenic effect and in preventing betaamyloid-induced cell death. J Agric Food Chem 55, 2438-2445. Uriarte-Pueyo I, Calvo MI (2011) Flavonoids as acetylcholinesterase inhibitors. Curr Med Chem 18, 5289-5302. Thapa A, Woo ER, Chi EY, Sharoar MG, Jin HG, Shin SY, Park IS (2011) Biflavonoids are superior to monoflavonoids in inhibiting amyloid-beta toxicity and fibrillogenesis via accumulation of nontoxic oligomer-like structures. Biochemistry 50, 2445-2455. Broersen LM, Kuipers AA, Balvers M, van WN, Savelkoul PJ, de Wilde MC, van der Beek EM, Sijben JW, Hageman RJ, Kamphuis PJ, Kiliaan AJ (2012) A specific multinutrient diet reduces Alzheimer-like pathology in young [639] [640] [641] [642] [643] [644] [645] [646] [647] [648] [649] [650] [651] [652] [653] [654] adult AbetaPPswe/PS1dE9 mice. J Alzheimers Dis, doi: 10.3233/JAD-2012-112039 [Epub ahead of print]. Cornejo A, Jimenez JM, Caballero L, Melo F, Maccioni RB (2011) Fulvic acid inhibits aggregation and promotes disassembly of tau fibrils associated with Alzheimer’s disease. J Alzheimers Dis 27, 143-153. Selassie M, Griffin B, Gwebu N, Gwebu ET (1999) Aged garlic extract attenuates the cytotoxicity of beta-amyloid on undifferentiated PC12 cells. In Vitro Cell Dev Biol Anim 35, 369-370. Griffin B, Selassie M, Gwebu ET (2000) Aged garlic extract suppresses lipid peroxidation induced by betaamyloid in PC12 cells. In Vitro Cell Dev Biol Anim 36, 279-280. Peng Q, Buz’Zard AR, Lau BH (2002) Neuroprotective effect of garlic compounds in amyloid-beta peptide-induced apoptosis in vitro. Med Sci Monit 8, BR328-BR337. Gupta VB, Indi SS, Rao KS (2009) Garlic extract exhibits antiamyloidogenic activity on amyloid-beta fibrillogenesis: Relevance to Alzheimer’s disease. Phytother Res 23, 111-115. Chauhan NB (2006) Effect of aged garlic extract on APP processing and tau phosphorylation in Alzheimer’s transgenic model Tg2576. J Ethnopharmacol 108, 385-394. Chauhan NB, Sandoval J (2007) Amelioration of early cognitive deficits by aged garlic extract in Alzheimer’s transgenic mice. Phytother Res 21, 629-640. Ray B, Chauhan NB, Lahiri DK (2011) The “aged garlic extract:” (AGE) and one of its active ingredients S-allylL-cysteine (SAC) as potential preventive and therapeutic agents for Alzheimer’s disease (AD). Curr Med Chem 18, 3306-3313. Stackman RW, Eckenstein F, Frei B, Kulhanek D, Nowlin J, Quinn JF (2003) Prevention of age-related spatial memory deficits in a transgenic mouse model of Alzheimer’s disease by chronic Ginkgo biloba treatment. Exp Neurol 184, 510-520. Tchantchou F, Xu Y, Wu Y, Christen Y, Luo Y (2007) EGb 761 enhances adult hippocampal neurogenesis and phosphorylation of CREB in transgenic mouse model of Alzheimer’s disease. FASEB J 21, 2400-2408. Defeudis FV, Drieu K (2000) Ginkgo biloba extract (EGb 761) and CNS functions: Basic studies and clinical applications. Curr Drug Targets 1, 25-58. Defeudis FV (2003) A brief history of EGb 761 and its therapeutic uses. Pharmacopsychiatry 36(Suppl 1), S2-S7. Allain H, Raoul P, Lieury A, LeCoz F, Gandon JM, D’Arbigny P (1993) Effect of two doses of ginkgo biloba extract (EGb 761) on the dual-coding test in elderly subjects. Clin Ther 15, 549-558. Le Bars PL, Katz MM, Berman N, Itil TM, Freedman AM, Schatzberg AF (1997) A placebo-controlled, doubleblind, randomized trial of an extract of Ginkgo biloba for dementia. North American EGb Study Group. JAMA 278, 1327-1332. Le Bars PL, Kieser M, Itil KZ (2000) A 26-week analysis of a double-blind, placebo-controlled trial of the ginkgo biloba extract EGb 761 in dementia. Dement Geriatr Cogn Disord 11, 230-237. Le Bars PL, Velasco FM, Ferguson JM, Dessain EC, Kieser M, Hoerr R (2002) Influence of the severity of cognitive impairment on the effect of the Gnkgo biloba extract EGb 761 in Alzheimer’s disease. Neuropsychobiology 45, 1926. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [655] [656] [657] [658] [659] [660] [661] [662] [663] [664] [665] [666] [667] [668] [669] Le Bars PL (2003) Magnitude of effect and special approach to Ginkgo biloba extract EGb 761 in cognitive disorders. Pharmacopsychiatry 36(Suppl 1), S44-S49. Le Bars PL (2003) Response patterns of EGb 761 in Alzheimer’s disease: Influence of neuropsychological profiles. Pharmacopsychiatry 36(Suppl 1), S50-S55. Oken BS, Storzbach DM, Kaye JA (1998) The efficacy of Ginkgo biloba on cognitive function in Alzheimer disease. Arch Neurol 55, 1409-1415. Mix JA, Crews WD Jr (2000) An examination of the efficacy of Ginkgo biloba extract EGb761 on the neuropsychologic functioning of cognitively intact older adults. J Altern Complement Med 6, 219-229. Mix JA, Crews WD Jr (2002) A double-blind, placebocontrolled, randomized trial of Ginkgo biloba extract EGb 761 in a sample of cognitively intact older adults: Neuropsychological findings. Hum Psychopharmacol 17, 267-277. Dekosky ST, Fitzpatrick A, Ives DG, Saxton J, Williamson J, Lopez OL, Burke G, Fried L, Kuller LH, Robbins J, 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 randomized trial of Ginkgo biloba extract in prevention of dementia. Contemp Clin Trials 27, 238-253. Dekosky ST, Furberg CD (2008) Turning over a new leaf: Ginkgo biloba in prevention of dementia? Neurology 70, 1730-1731. Mazza M, Capuano A, Bria P, Mazza S (2006) Ginkgo biloba and donepezil: A comparison in the treatment of Alzheimer’s dementia in a randomized placebo-controlled double-blind study. Eur J Neurol 13, 981-985. Snitz BE, O’Meara ES, Carlson MC, Arnold AM, Ives DG, Rapp SR, Saxton J, Lopez OL, Dunn LO, Sink KM, Dekosky ST (2009) Ginkgo biloba for preventing cognitive decline in older adults: A randomized trial. JAMA 302, 2663-2670. Yancheva S, Ihl R, Nikolova G, Panayotov P, Schlaefke S, Hoerr R (2009) Ginkgo biloba extract EGb 761(R), donepezil or both combined in the treatment of Alzheimer’s disease with neuropsychiatric features: A randomised, double-blind, exploratory trial. Aging Ment Health 13, 183-190. 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, 539546. Wang J, Ho L, Zhao W, Ono K, Rosensweig C, Chen L, Humala N, Teplow DB, Pasinetti GM (2008) Grapederived polyphenolics prevent Abeta oligomerization and attenuate cognitive deterioration in a mouse model of Alzheimer’s disease. J Neurosci 28, 6388-6392. Ksiezak-Reding H, Ho L, Santa-Maria I, az-Ruiz C, Wang J, Pasinetti GM (2012) Ultrastructural alterations of Alzheimer’s disease paired helical filaments by grape seedderived polyphenols. Neurobiol Aging 33, 1427-1439. Tarozzi A, Merlicco A, Morroni F, Bolondi C, Di IP, Ciccarelli R, Romano S, Giuliani P, Hrelia P (2010) Guanosine protects human neuroblastoma cells from oxidative stress and toxicity induced by Amyloid-beta peptide oligomers. J Biol Regul Homeost Agents 24, 297-306. 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] [671] [672] [673] [674] [675] [676] [677] [678] [679] [680] [681] [682] [683] [684] 77 Dong X, Geng M, Guan H (2003) Effects of the marine acidic oligose on dopamine release from striatum and amygdala in the rat model of Parkinson’s disease. Chin J Marine Drugs 22, 9-12. Zeng KW, Wang XM, Ko H, Kwon HC, Cha JW, Yang HO (2011) Hyperoside protects primary rat cortical neurons from neurotoxicity induced by amyloid beta-protein via the PI3K/Akt/Bad/Bcl(XL)-regulated mitochondrial apoptotic pathway. Eur J Pharmacol 672, 45-55. Nie J, Luo Y, Huang XN, Gong QH, Wu Q, Shi JS (2010) Icariin inhibits beta-amyloid peptide segment 25-35 induced expression of beta-secretase in rat hippocampus. Eur J Pharmacol 626, 213-218. Zeng KW, Ko H, Yang HO, Wang XM (2010) Icariin attenuates beta-amyloid-induced neurotoxicity by inhibition of tau protein hyperphosphorylation in PC12 cells. Neuropharmacology 59, 542-550. Zeng KW, Fu H, Liu GX, Wang XM (2010) Icariin attenuates lipopolysaccharide-induced microglial activation and resultant death of neurons by inhibiting TAK1/IKK/NF-kappaB and JNK/p38 MAPK pathways. Int Immunopharmacol 10, 668-678. Urano T, Tohda C (2010) Icariin improves memory impairment in Alzheimer’s disease model mice (5xFAD) and attenuates amyloid beta-induced neurite atrophy. Phytother Res 24, 1658-1663. Zheng M, Qu L, Lou Y (2008) Effects of icariin combined with Panax notoginseng saponins on ischemia reperfusioninduced cognitive impairments related with oxidative stress and CA1 of hippocampal neurons in rat. Phytother Res 22, 597-604. Carvajal FJ, Inestrosa NC (2011) Interactions of AChE with Abeta aggregates in Alzheimer’s brain: Therapeutic relevance of IDN 5706. Front Mol Neurosci 4, 19. Roth A, Schaffner W, Hertel C (1999) Phytoestrogen kaempferol (3,4 ,5,7-tetrahydroxyflavone) protects PC12 and T47D cells from beta-amyloid-induced toxicity. J Neurosci Res 57, 399-404. Kim JK, Choi SJ, Cho HY, Hwang HJ, Kim YJ, Lim ST, Kim CJ, Kim HK, Peterson S, Shin DH (2010) Protective effects of kaempferol (3,4 ,5,7-tetrahydroxyflavone) against amyloid beta peptide (Abeta)-induced neurotoxicity in ICR mice. Biosci Biotechnol Biochem 74, 397-401. Lee KY, Sung SH, Kim SH, Jang YP, Oh TH, Kim YC (2009) Cognitive-enhancing activity of loganin isolated from Cornus officinalis in scopolamine-induced amnesic mice. Arch Pharm Res 32, 677-683. Tsai FS, Peng WH, Wang WH, Wu CR, Hsieh CC, Lin YT, Feng IC, Hsieh MT (2007) Effects of luteolin on learning acquisition in rats: involvement of the central cholinergic system. Life Sci 80, 1692-1698. Tsai FS, Cheng HY, Hsieh MT, Wu CR, Lin YC, Peng WH (2010) The ameliorating effects of luteolin on betaamyloid-induced impairment of water maze performance and passive avoidance in rats. Am J Chin Med 38, 279291. Cheng HY, Hsieh MT, Tsai FS, Wu CR, Chiu CS, Lee MM, Xu HX, Zhao ZZ, Peng WH (2010) Neuroprotective effect of luteolin on amyloid beta protein (25-35)-induced toxicity in cultured rat cortical neurons. Phytother Res 24(Suppl 1), S102-S108. Xu B, Li XX, He GR, Hu JJ, Mu X, Tian S, Du GH (2010) Luteolin promotes long-term potentiation and improves cognitive functions in chronic cerebral hypoperfused rats. Eur J Pharmacol 627, 99-105. 78 [685] [686] [687] [688] [689] [690] [691] [692] [693] [694] [695] [696] [697] [698] [699] [700] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Parachikova A, Green KN, Hendrix C, LaFerla FM (2010) Formulation of a medical food cocktail for Alzheimer’s disease: Beneficial effects on cognition and neuropathology in a mouse model of the disease. PLoS One 5, e14015. Pappolla MA, Sos M, Omar RA, Bick RJ, Hickson-Bick DL, Reiter RJ, Efthimiopoulos S, Robakis NK (1997) Melatonin prevents death of neuroblastoma cells exposed to the Alzheimer amyloid peptide. J Neurosci 17, 16831690. Pappolla M, Bozner P, Soto C, Shao H, Robakis NK, Zagorski M, Frangione B, Ghiso J (1998) Inhibition of Alzheimer beta-fibrillogenesis by melatonin. J Biol Chem 273, 7185-7188. Pappolla MA, Chyan YJ, Poeggeler B, Frangione B, Wilson G, Ghiso J, Reiter RJ (2000) An assessment of the antioxidant and the antiamyloidogenic properties of melatonin: Implications for Alzheimer’s disease. J Neural Transm 107, 203-231. Pappolla MA, Simovich MJ, Bryant-Thomas T, Chyan YJ, Poeggeler B, Dubocovich M, Bick R, Perry G, Cruz-Sanchez F, Smith MA (2002) The neuroprotective activities of melatonin against the Alzheimer beta-protein are not mediated by melatonin membrane receptors. J Pineal Res 32, 135-142. Wang JZ, Wang ZF (2006) Role of melatonin in Alzheimer-like neurodegeneration. Acta Pharmacol Sin 27, 41-49. Cheng Y, Feng Z, Zhang QZ, Zhang JT (2006) Beneficial effects of melatonin in experimental models of Alzheimer disease. Acta Pharmacol Sin 27, 129-139. Srinivasan V, Kaur C, Pandi-Perumal S, Brown GM, Cardinali DP (2010) Melatonin and its agonist ramelteon in Alzheimer’s disease: Possible therapeutic value. Int J Alzheimers Dis 2011, 741974. Srinivasan V, Cardinali DP, Srinivasan US, Kaur C, Brown GM, Spence DW, Hardeland R, Pandi-Perumal SR (2011) Therapeutic potential of melatonin and its analogs in Parkinson’s disease: Focus on sleep and neuroprotection. Ther Adv Neurol Disord 4, 297-317. Argyriou A, Prast H, Philippu A (1998) Melatonin facilitates short-term memory. Eur J Pharmacol 349, 159-162. Bhadania M, Joshi H, Patel P, Kulkarni VH (2012) Protective effect of menthol on beta-amyloid peptide induced cognitive deficits in mice. Eur J Pharmacol 681, 50-54. Lemkul JA, Bevan DR (2010) Destabilizing Alzheimer’s Abeta(42) protofibrils with morin: Mechanistic insights from molecular dynamics simulations. Biochemistry 49, 3935-3946. Lemkul JA, Bevan DR (2012) Morin inhibits the early stages of amyloid beta-peptide aggregation by altering tertiary and quaternary interactions to produce “off-pathway” structures. Biochemistry 51, 5990-6009. Gong EJ, Park HR, Kim ME, Piao S, Lee E, Jo DG, Chung HY, Ha NC, Mattson MP, Lee J (2011) Morin attenuates tau hyperphosphorylation by inhibiting GSK3beta. Neurobiol Dis 44, 223-230. DeToma AS, Choi JS, Braymer JJ, Lim MH (2011) Myricetin: A naturally occurring regulator of metalinduced amyloid-beta aggregation and neurotoxicity. Chembiochem 12, 1198-1201. Jones JR, Lebar MD, Jinwal UK, Abisambra JF, Koren J III, Blair L, O’Leary JC, Davey Z, Trotter J, Johnson AG, Weeber E, Eckman CB, Baker BJ, Dickey CA [701] [702] [703] [704] [705] [706] [707] [708] [709] [710] [711] [712] (2011) The diarylheptanoid (+)-aR,11S-myricanol and two flavones from bayberry (Myrica cerifera) destabilize the microtubule-associated protein tau. J Nat Prod 74, 38-44. Maratha SR, Mahadevan N (2012) Memory enhancing activity of naringin in unstressed and stressed mice: Possible cholinergic and nitriergic modulation. Neurochem Res 37, 2206-2212. Ono K, Hasegawa K, Yoshiike Y, Takashima A, Yamada M, Naiki H (2002) Nordihydroguaiaretic acid potently breaks down pre-formed Alzheimer’s beta-amyloid fibrils in vitro. J Neurochem 81, 434-440. Moss MA, Varvel NH, Nichols MR, Reed DK, Rosenberry TL (2004) Nordihydroguaiaretic acid does not disaggregate beta-amyloid(1-40) protofibrils but does inhibit growth arising from direct protofibril association. Mol Pharmacol 66, 592-600. Bieschke J, Herbst M, Wiglenda T, Friedrich RP, Boeddrich 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 conversion of toxic oligomers to nontoxic beta-sheet-rich amyloid fibrils. Nat Chem Biol 8, 93-101. Choi DY, Lee YJ, Lee SY, Lee YM, Lee HH, Choi IS, Oh KW, Han SB, Nam SY, Hong JT (2012) Attenuation of scopolamine-induced cognitive dysfunction by obovatol. Arch Pharm Res 35, 1279-1286. Choi DY, Lee JW, Peng J, Lee YJ, Han JY, Lee YH, Choi IS, Han SB, Jung JK, Lee WS, Lee SH, Kwon BM, Oh KW, Hong JT (2012) Obovatol improves cognitive functions in animal models for Alzheimer’s disease. J Neurochem 120, 1048-1059. 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 memory impairments in mice via inhibition of NF-kappaB signaling pathway. Neurochem Int 60, 68-77. Choi DY, Lee YJ, Hong JT, Lee HJ (2012) Antioxidant properties of natural polyphenols and their therapeutic potentials for Alzheimer’s disease. Brain Res Bull 87, 144153. Daccache A, Lion C, Sibille N, Gerard M, Slomianny C, Lippens G, Cotelle P (2011) Oleuropein and derivatives from olives as Tau aggregation inhibitors. Neurochem Int 58, 700-707. Yu CJ, Zheng MF, Kuang CX, Huang WD, Yang Q (2010) Oren-gedoku-to and its constituents with therapeutic potential in Alzheimer’s disease inhibit indoleamine 2, 3-dioxygenase activity in vitro. J Alzheimers Dis 22, 257-266. Kim DH, Jeon SJ, Son KH, Jung JW, Lee S, Yoon BH, Choi JW, Cheong JH, Ko KH, Ryu JH (2006) Effect of the flavonoid, oroxylin A, on transient cerebral hypoperfusioninduced memory impairment in mice. Pharmacol Biochem Behav 85, 658-668. Fujiwara H, Tabuchi M, Yamaguchi T, Iwasaki K, Furukawa K, Sekiguchi K, Ikarashi Y, Kudo Y, Higuchi M, Saido TC, Maeda S, Takashima A, Hara M, Yaegashi N, Kase Y, Arai H (2009) A traditional medicinal herb Paeonia suffruticosa and its active constituent 1,2,3,4,6-penta-O-galloyl-beta-D-glucopyranose have potent anti-aggregation effects on Alzheimer’s amyloid beta proteins in vitro and in vivo. J Neurochem 109, 1648-1657. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [713] [714] [715] [716] [717] [718] [719] [720] [721] [722] [723] [724] [725] [726] [727] [728] Kim HJ, Lee KW, Lee HJ (2007) Protective effects of piceatannol against beta-amyloid-induced neuronal cell death. Ann N Y Acad Sci 1095, 473-482. Liu R, Wu CX, Zhou D, Yang F, Tian S, Zhang L, Zhang TT, Du GH (2012) Pinocembrin protects against beta-amyloid-induced toxicity in neurons through inhibiting receptor for advanced glycation end products (RAGE)-independent signaling pathways and regulating mitochondrion-mediated apoptosis. BMC Med 10, 105. Gazit E (2002) A possible role for pi-stacking in the selfassembly of amyloid fibrils. FASEB J 16, 77-83. Smid SD, Maag JL, Musgrave IF (2012) Dietary polyphenol-derived protection against neurotoxic betaamyloid protein: From molecular to clinical. Food Funct 3, 1242-1250. Xu J, Rong S, Xie B, Sun Z, Deng Q, Wu H, Bao W, Wang D, Yao P, Huang F, Liu L (2010) Memory impairment in cognitively impaired aged rats associated with decreased hippocampal CREB phosphorylation: Reversal by procyanidins extracted from the lotus seedpod. J Gerontol A Biol Sci Med Sci 65, 933-940. Xu J, Rong S, Xie B, Sun Z, Deng Q, Bao W, Wang D, Yao P, Huang F, Liu L (2011) Changes in the nitric oxide system contribute to effect of procyanidins extracted from the lotus seedpod ameliorating memory impairment in cognitively impaired aged rats. Rejuvenation Res 14, 33-43. Bithu BS, Reddy NR, Prasad SK, Sairam K, Hemalatha S (2012) Prosopis cineraria: A potential nootropic agent. Pharm Biol 50, 1241-1247. Li J, Wang G, Liu J, Zhou L, Dong M, Wang R, Li X, Li X, Lin C, Niu Y (2010) Puerarin attenuates amyloidbeta-induced cognitive impairment through suppression of apoptosis in rat hippocampus in vivo. Eur J Pharmacol 649, 195-201. Wang G, Zhou L, Zhang Y, Dong M, Li X, Liu J, Niu Y (2011) Implication of the c-Jun-NH2-terminal kinase pathway in the neuroprotective effect of puerarin against 1-methyl-4-phenylpyridinium (MPP+)-induced apoptosis in PC-12 cells. Neurosci Lett 487, 88-93. Xing G, Dong M, Li X, Zou Y, Fan L, Wang X, Cai D, Li C, Zhou L, Liu J, Niu Y (2011) Neuroprotective effects of puerarin against beta-amyloid-induced neurotoxicity in PC12 cells via a PI3K-dependent signaling pathway. Brain Res Bull 85, 212-218. Lin F, Xie B, Cai F, Wu G (2012) Protective effect of Puerarin on beta-amyloid-induced neurotoxicity in rat hippocampal neurons. Arzneimittelforschung 62, 187-193. Peng QL, Buz’Zard AR, Lau BH (2002) Pycnogenol protects neurons from amyloid-beta peptide-induced apoptosis. Brain Res Mol Brain Res 104, 55-65. Ansari MA, Abdul HM, Joshi G, Opii WO, Butterfield DA (2009) Protective effect of quercetin in primary neurons against Abeta(1-42): Relevance to Alzheimer’s disease. J Nutr Biochem 20, 269-275. Iuvone T, De FD, Esposito G, D’Amico A, Izzo AA (2006) The spice sage and its active ingredient rosmarinic acid protect PC12 cells from amyloid-beta peptide-induced neurotoxicity. J Pharmacol Exp Ther 317, 1143-1149. Park DH, Park SJ, Kim JM, Jung WY, Ryu JH (2010) Subchronic administration of rosmarinic acid, a natural prolyl oligopeptidase inhibitor, enhances cognitive performances. Fitoterapia 81, 644-648. Wang SW, Wang YJ, Su YJ, Zhou WW, Yang SG, Zhang R, Zhao M, Li YN, Zhang ZP, Zhan DW, Liu RT (2012) Rutin inhibits beta-amyloid aggregation and cytotoxicity, [729] [730] [731] [732] [733] [734] [735] [736] [737] [738] [739] [740] [741] 79 attenuates oxidative stress, and decreases the production of nitric oxide and proinflammatory cytokines. Neurotoxicology 33, 482-490. Papandreou MA, Kanakis CD, Polissiou MG, Efthimiopoulos S, Cordopatis P, Margarity M, Lamari FN (2006) Inhibitory activity on amyloid-beta aggregation and antioxidant properties of Crocus sativus stigmas extract and its crocin constituents. J Agric Food Chem 54, 8762-8768. Papandreou MA, Tsachaki M, Efthimiopoulos S, Cordopatis P, Lamari FN, Margarity M (2011) Memory enhancing effects of saffron in aged mice are correlated with antioxidant protection. Behav Brain Res 219, 197204. 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) Saffron in the treatment of patients with mild to moderate Alzheimer’s disease: A 16-week, randomized and placebocontrolled trial. J Clin Pharm Ther 35, 581-588. Akhondzadeh S, Shafiee SM, Harirchian MH, Togha M, Cheraghmakani H, Razeghi S, Hejazi SS, Yousefi MH, Alimardani R, Jamshidi A, Rezazadeh SA, Yousefi A, Zare F, Moradi A, Vossoughi A (2010) A 22-week, multicenter, randomized, double-blind controlled trial of Crocus sativus in the treatment of mild-to-moderate Alzheimer’s disease. Psychopharmacology (Berl) 207, 637-643. Geromichalos GD, Lamari FN, Papandreou MA, Trafalis DT, Margarity M, Papageorgiou A, Sinakos Z (2012) Saffron as a source of novel acetylcholinesterase inhibitors: Molecular docking and in vitro enzymatic studies. J Agric Food Chem 60, 6131-6138. Zhang L, Yu H, Zhao X, Lin X, Tan C, Cao G, Wang Z (2010) Neuroprotective effects of salidroside against betaamyloid-induced oxidative stress in SH-SY5Y human neuroblastoma cells. Neurochem Int 57, 547-555. 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 effect of S-allyl-L-cysteine, a garlic compound, on amyloid beta-protein-induced cell death in nerve growth factordifferentiated PC12 cells. Neurosci Res 46, 119-125. Ito Y, Ito M, Takagi N, Saito H, Ishige K (2003) Neurotoxicity induced by amyloid beta-peptide and ibotenic acid in organotypic hippocampal cultures: Protection by S-allylL-cysteine, a garlic compound. Brain Res 985, 98-107. Lu P, Mamiya T, Lu LL, Mouri A, Zou L, Nagai T, Hiramatsu M, Ikejima T, Nabeshima T (2009) Silibinin prevents amyloid beta peptide-induced memory impairment and oxidative stress in mice. Br J Pharmacol 157, 1270-1277. 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 mouse model of amyloid beta(1-42) protein-induced Alzheimer’s disease. Pharmacol Biochem Behav 103, 260-266. Scheltens P, Kamphuis PJ, Verhey FR, Olde Rikkert MG, Wurtman RJ, Wilkinson D, Twisk JW, Kurz A (2010) Efficacy of a medical food in mild Alzheimer’s disease: A randomized, controlled trial. Alzheimers Dement 6, 1-10. Alzheimer Research Forum (2011) Nutrient formulation appears to grease memory function, http://www. alzforum.org/new/detail.asp?id=2957. Cummings JL (2012) Food for thought: Souvenaid(R) in mild Alzheimer’s disease. J Alzheimers Dis 31, 237-238. 80 [742] [743] [744] [745] [746] [747] [748] [749] [750] [751] [752] [753] [754] [755] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Pieri M, Amadoro G, Carunchio I, Ciotti MT, Quaresima S, Florenzano F, Calissano P, Possenti R, Zona C, Severini C (2010) SP protects cerebellar granule cells against beta-amyloid-induced apoptosis by down-regulation and reduced activity of Kv4 potassium channels. Neuropharmacology 58, 268-276. Yang EJ, Kim SI, Ku HY, Lee DS, Lee JW, Kim YS, Seong YH, Song KS (2010) Syringin from stem bark of Fraxinus rhynchophylla protects Abeta(25-35)-induced toxicity in neuronal cells. Arch Pharm Res 33, 531-538. Ono K, Hasegawa K, Naiki H, Yamada M (2004) Antiamyloidogenic activity of tannic acid and its activity to destabilize Alzheimer’s beta-amyloid fibrils in vitro. Biochim Biophys Acta 1690, 193-202. Inbar P, Bautista MR, Takayama SA, Yang J (2008) Assay to screen for molecules that associate with Alzheimer’s related beta-amyloid fibrils. Anal Chem 80, 3502-3506. Mori T, Rezai-Zadeh K, Koyama N, Arendash GW, Yamaguchi H, Kakuda N, Horikoshi-Sakuraba Y, Tan J, Town T (2012) Tannic acid is a natural beta-secretase inhibitor that prevents cognitive impairment and mitigates Alzheimerlike pathology in transgenic mice. J Biol Chem 287, 6912-6927. Yang SG, Wang WY, Ling TJ, Feng Y, Du XT, Zhang X, Sun XX, Zhao M, Xue D, Yang Y, Liu RT (2010) alpha-Tocopherol quinone inhibits beta-amyloid aggregation and cytotoxicity, disaggregates preformed fibrils and decreases the production of reactive oxygen species, NO and inflammatory cytokines. Neurochem Int 57, 914-922. Yang ZQ, Yang SF, Yang JQ, Zhou QX, Li SL (2007) Protective effects and mechanism of total coptis alkaloids on a beta 25-35 induced learning and memory dysfunction in rats. Chin J Integr Med 13, 50-54. Hong SY, Jeong WS, Jun M (2012) Protective effects of the key compounds isolated from Corni fructus against betaamyloid-induced neurotoxicity in PC12 cells. Molecules 17, 10831-10845. Takasaki J, Ono K, Yoshiike Y, Hirohata M, Ikeda T, Morinaga A, Takashima A, Yamada M (2011) Vitamin A has anti-oligomerization effects on amyloid-beta in vitro. J Alzheimers Dis 27, 271-280. Kulkarni SK, Dhir A (2008) Withania somnifera: An Indian ginseng. Prog Neuropsychopharmacol Biol Psychiatry 32, 1093-1105. Sehgal N, Gupta A, Valli RK, Joshi SD, Mills JT, Hamel E, Khanna P, Jain SC, Thakur SS, Ravindranath V (2012) Withania somnifera reverses Alzheimer’s disease pathology by enhancing low-density lipoprotein receptor-related protein in liver. Proc Natl Acad Sci U S A 109, 3510-3515. Wang XM, Fu H, Liu GX, Zhu W, Li L, Yang JX (2007) Effect of modified wuzi yanzong granule on patients with mild cognitive impairment from oxidative damage aspect. Chin J Integr Med 13, 258-263. Hayashi Y, Ishida Y, Inoue T, Udagawa M, Takeuchi K, Yoshimuta H, Kiue K, Ninomiya Y, Kawano J, Sameshima T, Kawahara T, Goto I, Shudo K, Kurayama S, Nakamura J, Okahara K, Mitsuyama Y (2010) Treatment of behavioral and psychological symptoms of Alzheimertype dementia with Yokukansan in clinical practice. Prog Neuropsychopharmacol Biol Psychiatry 34, 541-545. Uchida N, Takasaki K, Sakata Y, Nogami A, Oishi H, Watanabe T, Shindo T, Egashira N, Kubota K, Katsurabayashi S, Mishima K, Fujiwara M, Nishimura R, Iwasaki K (2012) Cholinergic involvement and synaptic dynamin 1 expression in yokukansan-mediated improve- [756] [757] [758] [759] [760] [761] [762] [763] [764] [765] [766] [767] [768] [769] [770] [771] ment of spatial memory in a rat model of early Alzheimer’s disease. Phytother Res, doi: 10.1002/ptr.4818 [Epub ahead of print]. Tohda C, Tamura T, Komatsu K (2003) Repair of amyloid beta(25-35)-induced memory impairment and synaptic loss by a Kampo formula, Zokumei-to. Brain Res 990, 141-147. Takahashi K, Matsumura T, Ishihara J, Hatakeyama S (2007) A highly stereocontrolled total synthesis of dysiherbaine. Chem Commun (Camb) 4158-4160. Nakanishi K (2005) Terpene trilactones from Gingko biloba: From ancient times to the 21st century. Bioorg Med Chem 13, 4987-5000. Keowkase R, Aboukhatwa M, Adam BL, Beach JW, Terry AV Jr, Buccafussco JJ, Luo Y (2010) Neuroprotective effects and mechanism of cognitive-enhancing choline analogs JWB 1-84-1 and JAY 2-22-33 in neuronal culture and Caenorhabditis elegans. Mol Neurodegener 5, 59. Sood A, Warren BJ, Webster SJ, Terry AV, Buccafusco JJ (2007) The effects of JWB1-84-1 on memory-related task performance by amyloid Abeta transgenic mice and by young and aged monkeys. Neuropharmacology 53, 588600. 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, attenuates cognitive deficits and shows neuroprotective effects in vitro and in vivo dementia models. J Neurosci Res 85, 2500-2511. Guan J, Zhang R, le-Gandar L, Hodgkinson S, Vickers MH (2010) NNZ-2591, a novel diketopiperazine, prevented scopolamine-induced acute memory impairment in the adult rat. Behav Brain Res 210, 221-228. Wang HY, Bakshi K, Frankfurt M, Stucky A, Goberdhan M, Shah SM, Burns LH (2012) Reducing amyloid-related Alzheimer’s disease pathogenesis by a small molecule targeting filamin a. J Neurosci 32, 9773-9784. Zs-Nagy I, Ohta M, Kitani K (1989) Effect of centrophenoxine and BCE-001 treatment on lateral diffusion of proteins in the hepatocyte plasma membrane as revealed by fluorescence recovery after photobleaching in rat liver smears. Exp Gerontol 24, 317-330. Zs-Nagy I (1994) A survey of the available data on a new nootropic drug, BCE-001. Ann N Y Acad Sci 717, 102-114. Nagy K, Dajko G, Uray I, Zs-Nagy I (1994) Comparative studies on the free radical scavenger properties of two nootropic drugs, CPH and BCE-001. Ann N Y Acad Sci 717, 115-121. Scopes D, O’Hare E, Jeggo R, Whyment A, Spanswick D, Kim EM, Gannon J, Amijee H, Treherne J (2012) Abeta oligomer toxicity inhibitor protects memory in models of synaptic toxicity. Br J Pharmacol 167, 383-392. Pierce JE, Smith DH, Eison MS, McIntosh TK (1993) The nootropic compound BMY-21502 improves spatial learning ability in brain injured rats. Brain Res 624, 199-208. Saletu B, Schulz H, Herrmann WM, Anderer P, Shrotriya RC, Vanbrabant E (1994) BMS-181168 for protection of the human brain against hypoxia: Double-blind, placebocontrolled EEG mapping studies. Pharmacopsychiatry 27, 189-197. Shrotriya RC, Cutler NR, Sramek JJ, Veroff AE, Hironaka DY (1996) Efficacy and safety of BMY 21,502 in Alzheimer disease. Ann Pharmacother 30, 1376-1380. Lishko PV, Maximyuk OP, Chatterjee SS, Noldner M, Krishtal OA (1998) The putative cognitive enhancer KA- W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [772] [773] [774] [775] [776] [777] [778] [779] [780] [781] [782] [783] [784] [785] [786] [787] 672.HCl is an uncompetitive voltage-dependent NMDA receptor antagonist. Neuroreport 9, 4193-4197. Sourgens H, Hoerr R, Biber A, Steinbrede H, Derendorf H (1998) KA 672-HCl, a neuronal activator against dementia: Tolerability, safety, and preliminary pharmacokinetics after single and multiple oral doses in healthy male and female volunteers. J Clin Pharmacol 38, 373-381. Winter JC, Helsley SE, Rabin RA (1998) The discriminative stimulus effects of KA 672, a putative cognitive enhancer: Evidence for a 5-HT1A component. Pharmacol Biochem Behav 60, 703-707. Reisner E, Noldner M, Rossner S, Chatterjee SS, Bigl V, Schliebs R (1999) Acute effect of KA-672, a putative cognitive enhancer, on neurotransmitter receptor binding in mouse brain. Neurosci Lett 274, 187-190. Teismann P, Ferger B (2000) In vivo effects of the putative cognitive enhancer KA-672.HCl in comparison with 8hydroxy-2-(di-N-propylamino) tetralin and haloperidol on dopamine, 3,4-dihydroxyphenylacetic acid, serotonin and 5-hydroxyindoleacetic acid levels in striatal and cortical brain regions. Prog Neuropsychopharmacol Biol Psychiatry 24, 337-348. Teismann P, Ferger B (2000) Effects of ensaculin on dopamine metabolite levels and K(+)-induced glutamate release. Eur J Pharmacol 398, 247-250. Hoerr R, Noeldner M (2002) Ensaculin (KA-672 HCl): a multitransmitter approach to dementia treatment. CNS Drug Rev 8, 143-158. Sramek JJ, Cutler NR (2000) Ongoing trials in Alzheimer’s disease. Expert Opin Investig Drugs 9, 899-915. Inozemtsev AN, Garibova TL, Khromova IV, Alvarez R, Voronina TA, Tushmalova NA (1993) [The effect of nooglutil and piracetam on different forms of operant learning]. Eksp Klin Farmakol 56, 6-8. Mondadori C, Ducret T, Borkowski J (1991) How long does ‘memory consolidation’ take? New compounds can improve retention performance, even if administered up to 24 hours after the learning experience. Brain Res 555, 107-111. Mondadori C, Buerki H, Borkowski J, Radeke E, Ducret T, Glatt A (1992) CGS 5649 B, a new compound, reverses age-related cognitive dysfunctions in rats. Behav Neural Biol 57, 149-156. Nilsson E, Schneider T, Hoffmann E, Schmidt EK (1991) Urinary excretion of CGS 5,649 and its conjugated glucuronide and sulfate in man. Eur J Drug Metab Pharmacokinet Spec No 3, 201-204. Molloy DW, Guyatt GH, Standish T, Willan A, McIlroy W, D’Souza J, Brown G, Mondadori C (1993) Effect of a new nootropic agent, CGS 5649B, on cognition, function, and behavior in dementia. J Gen Intern Med 8, 444-447. 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. Psychopharmacology (Berl) 94, 52-57. Clincke GH, Tritsmans L (1988) Sabeluzole (R58 735) increases consistent retrieval during serial learning and relearning of nonsense syllables. Psychopharmacology (Berl) 96, 309-310. Tritsmans L, Clincke G, Amery WK (1988) The effect of sabeluzole (R 58735) on memory retrieval functions. Psychopharmacology (Berl) 94, 527-531. Geerts H, Nuydens R, Nuyens R, Cornelissen F (1992) Sabeluzole accelerates neurite outgrowth in different neuronal cell lines. Restor Neurol Neurosci 4, 21-32. [788] [789] [790] [791] [792] [793] [794] [795] [796] [797] [798] [799] [800] [801] [802] [803] 81 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 fast axonal transport in neuronal cell cultures. Exp Neurol 117, 36-43. Geerts H, Nuydens R, de JM, Cornelissen F, Nuyens R, Wouters L (1996) Sabeluzole stabilizes the neuronal cytoskeleton. Neurobiol Aging 17, 573-581. Wong YN, Quon CY, Holm KA, Burcham DL, Frey NL, Huang SM, Lam GN (1996) Pharmacokinetics and metabolism of EXP921, a novel cognitive enhancer, in rats. Drug Metab Dispos 24, 172-179. Saletu B, Semlitsch HV, Anderer P, Resch F, Presslich O, Schuster P (1989) Psychophysiological research in psychiatry and neuropsychopharmacology. II. The investigation of antihypoxidotic/nootropic drugs (tenilsetam and co-dergocrine-mesylate) in elderlies with the Viennese Psychophysiological Test-System (VPTS). Methods Find Exp Clin Pharmacol 11, 43-55. 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 tenilsetam protect against the neurotoxic effects of methylglyoxal. Neurotox Res 7, 95-101. Rainer M, Brunnbauer M, Dunky A, Ender F, Goldsteiner H, Holl O, Kotlan P, Paulitsch G, Reiner C, Stossl J, Zachhuber C, Mossler H (1997) Therapeutic results with Cerebrolysin in the treatment of dementia. Wien Med Wochenschr 147, 426-431. Xiong H, Baskys A, Wojtowicz JM (1996) Brain-derived peptides inhibit synaptic transmission via presynaptic GABAB receptors in CA1 area of rat hippocampal slices. Brain Res 737, 188-194. Kofler B, Erhart C, Erhart P, Harrer G (1990) A multidimensional approach in testing nootropic drug effects (Cerebrolysin). Arch Gerontol Geriatr 10, 129-140. Hutter-Paier B, Eggenreich U, Windisch M (1996) Effects of two protein-free peptide derivatives on passive avoidance behaviour of 24-month-old rats. Arzneimittelforschung 46, 237-241. Hutter-Paier B, Eggenreich U, Windisch M (1996) Dosedependent behavioural effects of two protein-free peptide derivatives on the passive avoidance reaction of rats. Arzneimittelforschung 46, 242-246. Gschanes A, Windisch M (1996) The influence of cerebrolysin and E021 on spatial navigation of young rats. J Neural Transm Suppl 47, 278. Gschanes A, Windisch M (1998) The influence of Cerebrolysin and E021 on spatial navigation of 24-month-old rats. J Neural Transm Suppl 53, 313-321. Hutter-Paier B, Fruhwirth M, Grygar E, Windisch M (1996) Cerebrolysin protects neurons from ischemiainduced loss of microtubule–associated protein 2. J Neural Transm Suppl 47, 276. Rockenstein E, Mallory M, Mante M, Alford M, Windisch M, Moessler H, Masliah E (2002) Effects of Cerebrolysin on amyloid-beta deposition in a transgenic model of Alzheimer’s disease. J Neural Transm Suppl 62, 327-336. Rockenstein E, Adame A, Mante M, Moessler H, Windisch M, Masliah E (2003) The neuroprotective effects of Cerebrolysin in a transgenic model of Alzheimer’s disease are associated with improved behavioral performance. J Neural Transm 110, 1313-1327. Rockenstein E, Adame A, Mante M, Larrea G, Crews L, Windisch M, Moessler H, Masliah E (2005) Ameliora- 82 [804] [805] [806] [807] [808] [809] [810] [811] [812] [813] [814] [815] 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 transgenic model of Alzheimer’s disease with the neurotrophic compound cerebrolysin. J Neural Transm 112, 269282. Rockenstein E, Torrance M, Mante M, Adame A, Paulino A, Rose JB, Crews L, Moessler H, Masliah E (2006) Cerebrolysin decreases amyloid-beta production by regulating amyloid protein precursor maturation in a transgenic model of Alzheimer’s disease. J Neurosci Res 83, 12521261. Rockenstein E, Mante M, Adame A, Crews L, Moessler H, Masliah E (2007) Effects of Cerebrolysin on neurogenesis in an APP transgenic model of Alzheimer’s disease. Acta Neuropathol 113, 265-275. 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 a neurotrophic peptidergic mixture persist for a prolonged period following treatment interruption in a transgenic model of Alzheimer’s disease. J Neurosci Res 89, 18121821. 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 comparison of the neurogenic effects of CNTF-derived peptides and cerebrolysin in AbetaPP transgenic mice. J Alzheimers Dis 27, 743-752. Masliah E, Diez-Tejedor E (2012) The pharmacology of neurotrophic treatment with Cerebrolysin: Brain protection and repair to counteract pathologies of acute and chronic neurological disorders. Drugs Today (Barc) 48(Suppl A), 3-24. Panisset M, Gauthier S, Moessler H, Windisch M (2002) Cerebrolysin in Alzheimer’s disease: A randomized, double-blind, placebo-controlled trial with a neurotrophic agent. J Neural Transm 109, 1089-1104. Molloy DW, Standish TI (2000) Clinical experience with Cerebrolysin. J Neural Transm Suppl 59, 293-300. Ruether E, Alvarez XA, Rainer M, Moessler H (2002) Sustained improvement of cognition and global function in patients with moderately severe Alzheimer’s disease: A double-blind, placebo-controlled study with the neurotrophic agent Cerebrolysin. J Neural Transm Suppl 62, 265-275. Ladurner G, Kalvach P, Moessler H (2005) Neuroprotective treatment with cerebrolysin in patients with acute stroke: A randomised controlled trial. J Neural Transm 112, 415-428. Alvarez XA, Cacabelos R, Laredo M, Couceiro V, Sampedro C, Varela M, Corzo L, Fernandez-Novoa L, Vargas M, Aleixandre M, Linares C, Granizo E, Muresanu D, Moessler H (2006) A 24-week, double-blind, placebocontrolled study of three dosages of Cerebrolysin in patients with mild to moderate Alzheimer’s disease. Eur J Neurol 13, 43-54. Alvarez XA, Sampedro C, Figueroa J, Tellado I, Gonzalez A, Garcia-Fantini M, Cacabelos R, Muresanu D, Moessler H (2008) Reductions in qEEG slowing over 1 year and after treatment with Cerebrolysin in patients with moderate-severe traumatic brain injury. J Neural Transm 115, 683-692. 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 the serum of Alzheimer’s disease patients treated with the neu- [816] [817] [818] [819] [820] [821] [822] [823] [824] [825] [826] [827] [828] [829] [830] rotrophic agent cerebrolysin. Int J Neuropsychopharmacol 12, 867-872. 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 moderately severe Alzheimer’s disease: Results of a randomized, double-blind, controlled trial investigating three dosages of Cerebrolysin. Eur J Neurol 18, 59-68. Muresanu DF, Alvarez XA, Moessler H, Buia M, Stan A, Pintea D, Moldovan F, Popescu BO (2008) A pilot study to evaluate the effects of Cerebrolysin on cognition and qEEG in vascular dementia: Cognitive improvement correlates with qEEG acceleration. J Neurol Sci 267, 112-119. Muresanu DF, Alvarez XA, Moessler H, Novak PH, Stan A, Buzoianu A, Bajenaru O, Popescu BO (2010) Persistence of the effects of Cerebrolysin on cognition and qEEG slowing in vascular dementia patients: Results of a 3-month extension study. J Neurol Sci 299, 179-183. Plosker GL, Gauthier S (2009) Cerebrolysin: A review of its use in dementia. Drugs Aging 26, 893-915. Plosker GL, Gauthier S (2010) Spotlight on cerebrolysin in dementia. CNS Drugs 24, 263-266. Bajenaru O, Tiu C, Moessler H, Antochi F, Muresanu D, Popescu BO, Novak P (2010) Efficacy and safety of Cerebrolysin in patients with hemorrhagic stroke. J Med Life 3, 137-143. Guekht AB, Moessler H, Novak PH, Gusev EI (2011) Cerebrolysin in vascular dementia: Improvement of clinical outcome in a randomized, double-blind, placebocontrolled multicenter trial. J Stroke Cerebrovasc Dis 20, 310-318. Allegri RF, Guekht A (2012) Cerebrolysin improves symptoms and delays progression in patients with Alzheimer’s disease and vascular dementia. Drugs Today (Barc) 48 Suppl A, 25-41. Alvarez XA, Cacabelos R, Sampedro C, Couceiro V, Aleixandre M, Vargas M, Linares C, Granizo E, GarciaFantini M, Baurecht W, Doppler E, Moessler H (2011) Combination treatment in Alzheimer’s disease: Results of a randomized, controlled trial with cerebrolysin and donepezil. Curr Alzheimer Res 8, 583-591. Anton Alvarez X, Fuentes P (2011) Cerebrolysin in Alzheimer’s disease. Drugs Today (Barc) 47, 487-513. Thome J, Doppler E (2012) Safety profile of Cerebrolysin: Clinical experience from dementia and stroke trials. Drugs Today (Barc)48 Suppl A, 63-69. Leker RR, Teichner A, Grigoriadis N, Ovadia H, Brenneman DE, Fridkin M, Giladi E, Romano J, Gozes I (2002) NAP, a femtomolar-acting peptide, protects the brain against ischemic injury by reducing apoptotic death. Stroke 33, 1085-1092. Ashur-Fabian O, Segal-Ruder Y, Skutelsky E, Brenneman DE, Steingart RA, Giladi E, Gozes I (2003) The neuroprotective peptide NAP inhibits the aggregation of the beta-amyloid peptide. Peptides 24, 1413-1423. Alcalay RN, Giladi E, Pick CG, Gozes I (2004) Intranasal administration of NAP, a neuroprotective peptide, decreases anxiety-like behavior in aging mice in the elevated plus maze. Neurosci Lett 361, 128-131. Gozes I, Zaltzman R, Hauser J, Brenneman DE, Shohami E, Hill JM (2005) The expression of activity-dependent neuroprotective protein (ADNP) is regulated by brain damage and treatment of mice with the ADNP derived peptide, NAP, reduces the severity of traumatic head injury. Curr Alzheimer Res 2, 149-153. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [831] [832] [833] [834] [835] [836] [837] [838] [839] [840] [841] [842] [843] [844] [845] [846] Matsuoka Y, Gray AJ, Hirata-Fukae C, Minami SS, Waterhouse EG, Mattson MP, LaFerla FM, Gozes I, Aisen PS (2007) Intranasal NAP administration reduces accumulation of amyloid peptide and tau hyperphosphorylation in a transgenic mouse model of Alzheimer’s disease at early pathological stage. J Mol Neurosci 31, 165-170. Matsuoka Y, Jouroukhin Y, Gray AJ, Ma L, Hirata-Fukae C, Li HF, Feng L, Lecanu L, Walker BR, Planel E, Arancio O, Gozes I, Aisen PS (2008) A neuronal microtubuleinteracting agent, NAPVSIPQ, reduces tau pathology and enhances cognitive function in a mouse model of Alzheimer’s disease. J Pharmacol Exp Ther 325, 146-153. Shiryaev N, Jouroukhin Y, Giladi E, Polyzoidou E, Grigoriadis NC, Rosenmann H, Gozes I (2009) NAP protects memory, increases soluble tau and reduces tau hyperphosphorylation in a tauopathy model. Neurobiol Dis 34, 381-388. Gozes I, Divinski I (2004) The femtomolar-acting NAP interacts with microtubules: Novel aspects of astrocyte protection. J Alzheimers Dis 6, S37-S41. Gozes I, Divinski I (2007) NAP, a neuroprotective drug candidate in clinical trials, stimulates microtubule assembly in the living cell. Curr Alzheimer Res 4, 507-509. Merenlender-Wagner A, Pikman R, Giladi E, Andrieux A, Gozes I (2010) NAP (davunetide) enhances cognitive behavior in the STOP heterozygous mouse–a microtubuledeficient model of schizophrenia. Peptides 31, 1368-1373. Fleming SM, Mulligan CK, Richter F, Mortazavi F, Lemesre V, Frias C, Zhu C, Stewart A, Gozes I, Morimoto B, Chesselet MF (2011) A pilot trial of the microtubule-interacting peptide (NAP) in mice overexpressing alpha-synuclein shows improvement in motor function and reduction of alpha-synuclein inclusions. Mol Cell Neurosci 46, 597-606. Gozes I (2007) Activity-dependent neuroprotective protein: From gene to drug candidate. Pharmacol Ther 114, 146-154. Gozes I (2011) NAP (davunetide) provides functional and structural neuroprotection. Curr Pharm Des 17, 10401044. Gozes I (2011) Microtubules, schizophrenia and cognitive behavior: Preclinical development of davunetide (NAP) as a peptide-drug candidate. Peptides 32, 428-431. Gozes I (2011) Microtubules (tau) as an emerging therapeutic target: NAP (davunetide). Curr Pharm Des 17, 3413-3417. Gozes I, Spivak-Pohis I (2006) Neurotrophic effects of the peptide NAP: A novel neuroprotective drug candidate. Curr Alzheimer Res 3, 197-199. Gozes I, Steingart RA, Spier AD (2004) NAP mechanisms of neuroprotection. J Mol Neurosci 24, 67-72. Gozes I, Morimoto BH, Tiong J, Fox A, Sutherland K, Dangoor D, Holser-Cochav M, Vered K, Newton P, Aisen PS, Matsuoka Y, van Dyck CH, Thal L (2005) NAP: Research and development of a peptide derived from activity-dependent neuroprotective protein (ADNP). CNS Drug Rev 11, 353-368. Gozes I, Stewart A, Morimoto B, Fox A, Sutherland K, Schmeche D (2009) Addressing Alzheimer’s disease tangles: From NAP to AL-108. Curr Alzheimer Res 6, 455-460. Geerts H (2008) AL-108 and AL-208, formulations of the neuroprotective NAP fragment of activity-dependent neuroprotective protein, for cognitive disorders. Curr Opin Investig Drugs 9, 800-811. [847] [848] [849] [850] [851] [852] [853] [854] [855] [856] [857] [858] [859] [860] [861] [862] 83 Shiryaev N, Pikman R, Giladi E, Gozes I (2011) Protection against tauopathy by the drug candidates NAP (davunetide) and D-SAL: Biochemical, cellular and behavioral aspects. Curr Pharm Des 17, 2603-2612. Javitt DC, Buchanan RW, Keefe RS, Kern R, McMahon RP, Green MF, Lieberman J, Goff DC, Csernansky JG, McEvoy JP, Jarskog F, Seidman LJ, Gold JM, Kimhy D, Nolan KS, Barch DS, Ball MP, Robinson J, Marder SR (2012) Effect of the neuroprotective peptide davunetide (AL-108) on cognition and functional capacity in schizophrenia. Schizophr Res 136, 25-31. Borsello T, Clarke PG, Hirt L, Vercelli A, Repici M, Schorderet DF, Bogousslavsky J, Bonny C (2003) A peptide inhibitor of c-Jun N-terminal kinase protects against excitotoxicity and cerebral ischemia. Nat Med 9, 11801186. Hirt L, Badaut J, Thevenet J, Granziera C, Regli L, Maurer F, Bonny C, Bogousslavsky J (2004) D-JNKI1, a cell-penetrating c-Jun-N-terminal kinase inhibitor, protects against cell death in severe cerebral ischemia. Stroke 35, 1738-1743. Suckfuell M, Canis M, Strieth S, Scherer H, Haisch A (2007) Intratympanic treatment of acute acoustic trauma with a cell-permeable JNK ligand: A prospective randomized phase I/II study. Acta Otolaryngol 127, 938-942. Wiegler K, Bonny C, Coquoz D, Hirt L (2008) The JNK inhibitor XG-102 protects from ischemic damage with delayed intravenous administration also in the presence of recombinant tissue plasminogen activator. Cerebrovasc Dis 26, 360-366. Tayal V, Kalra BS (2008) Cytokines and anti-cytokines as therapeutics–an update. Eur J Pharmacol 579, 1-12. Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP (2008) Tumor necrosis factor antagonist mechanisms of action: A comprehensive review. Pharmacol Ther 117, 244-279. Tobinick EL (2007) Perispinal etanercept for the treatment of Alzheimer’s disease. Curr Alzheimer Res 4, 550-552. Tobinick EL, Gross H (2008) Rapid improvement in verbal fluency and aphasia following perispinal etanercept in Alzheimer’s disease. BMC Neurol 8, 27. Tobinick EL, Gross H (2008) Rapid cognitive improvement in Alzheimer’s disease following perispinal etanercept administration. J Neuroinflammation 5, 2. Griffin WS (2008) Perispinal etanercept: Potential as an Alzheimer therapeutic. J Neuroinflammation 5, 3. Sara VR, Carlsson-Skwirut C, Bergman T, Jornvall H, Roberts PJ, Crawford M, Hakansson LN, Civalero I, Nordberg A (1989) Identification of Gly-Pro-Glu (GPE), the aminoterminal tripeptide of insulin-like growth factor 1 which is truncated in brain, as a novel neuroactive peptide. Biochem Biophys Res Commun 165, 766-771. Sara VR, Carlsson-Skwirut C, Drakenberg K, Giacobini MB, Hakansson L, Mirmiran M, Nordberg A, Olson L, Reinecke M, Stahlbom PA (1993) The biological role of truncated insulin-like growth factor-1 and the tripeptide GPE in the central nervous system. Ann N Y Acad Sci 692, 183-191. Saura J, Curatolo L, Williams CE, Gatti S, Benatti L, Peeters C, Guan J, Dragunow M, Post C, Faull RL, Gluckman PD, Skinner SJ (1999) Neuroprotective effects of Gly-Pro-Glu, the N-terminal tripeptide of IGF-1, in the hippocampus in vitro. Neuroreport 10, 161-164. Aguado-Llera D, Martin-Martinez M, Garcia-Lopez MT, rilla-Ferreiro E, Barrios V (2004) Gly-Pro-Glu protects 84 [863] [864] [865] [866] [867] [868] [869] [870] [871] [872] [873] [874] 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 rat cortex. Neuroreport 15, 1979-1982. Burgos-Ramos E, Martos-Moreno GA, Lopez MG, Herranz R, Aguado-Llera D, Egea J, Frechilla D, Cenarruzabeitia E, Leon R, Arilla-Ferreiro E, Argente J, Barrios V (2009) The N-terminal tripeptide of insulin-like growth factor-I protects against beta-amyloid-induced somatostatin depletion by calcium and glycogen synthase kinase 3 beta modulation. J Neurochem 109, 360-370. Bickerdike MJ, Thomas GB, Batchelor DC, Sirimanne ES, Leong W, Lin H, Sieg F, Wen J, Brimble MA, Harris PW, Gluckman PD (2009) NNZ-2566: A Gly-Pro-Glu analogue with neuroprotective efficacy in a rat model of acute focal stroke. J Neurol Sci 278, 85-90. Lu XC, Chen RW, Yao C, Wei H, Yang X, Liao Z, Dave JR, Tortella FC (2009) NNZ-2566, a glypromate analog, improves functional recovery and attenuates apoptosis and inflammation in a rat model of penetrating ballistic-type brain injury. J Neurotrauma 26, 141-154. Lu XC, Si Y, Williams AJ, Hartings JA, Gryder D, Tortella FC (2009) NNZ-2566, a glypromate analog, attenuates brain ischemia-induced non-convulsive seizures in rats. J Cereb Blood Flow Metab 29, 1924-1932. Wei HH, Lu XC, Shear DA, Waghray A, Yao C, Tortella FC, Dave JR (2009) NNZ-2566 treatment inhibits neuroinflammation and pro-inflammatory cytokine expression induced by experimental penetrating ballistic-like brain injury in rats. J Neuroinflammation 6, 19. Alonso De Diego SA, Munoz P, Gonzalez-Muniz R, Herranz R, Martin-Martinez M, Cenarruzabeitia E, Frechilla D, Del RJ, Jimeno ML, Garcia-Lopez MT (2005) Analogues of the neuroprotective tripeptide Gly-Pro-Glu (GPE): Synthesis and structure-activity relationships. Bioorg Med Chem Lett 15, 2279-2283. Alonso De Diego SA, Gutierrez-Rodriguez M, Perez d V, Casabona D, Cativiela C, Gonzalez-Muniz R, Herranz R, Cenarruzabeitia E, Frechilla D, Del RJ, Jimeno ML, Garcia-Lopez MT (2006) New Gly-Pro-Glu (GPE) analogues: Expedite solid-phase synthesis and biological activity. Bioorg Med Chem Lett 16, 1392-1396. Alonso De Diego SA, Gutierrez-Rodriguez M, Perez d V, Gonzalez-Muniz R, Herranz R, Martin-Martinez M, Cenarruzabeitia E, Frechilla D, Del RJ, Jimeno ML, Garcia-Lopez MT (2006) The neuroprotective activity of GPE tripeptide analogues does not correlate with glutamate receptor binding affinity. Bioorg Med Chem Lett 16, 3396-3400. Lai MY, Brimble MA, Callis DJ, Harris PW, Levi MS, Sieg F (2005) Synthesis and pharmacological evaluation of glycine-modified analogues of the neuroprotective agent glycyl-L-prolyl-L-glutamic acid (GPE). Bioorg Med Chem 13, 533-548. Holtje M, Djalali S, Hofmann F, Munster-Wandowski A, Hendrix S, Boato F, Dreger SC, Grosse G, Henneberger C, Grantyn R, Just I, Ahnert-Hilger G (2009) A 29amino acid fragment of Clostridium botulinum C3 protein enhances neuronal outgrowth, connectivity, and reinnervation. FASEB J 23, 1115-1126. Boato F, Hendrix S, Huelsenbeck SC, Hofmann F, Grosse G, Djalali S, Klimaschewski L, Auer M, Just I, hnertHilger G, Holtje M (2010) C3 peptide enhances recovery from spinal cord injury by improved regenerative growth of descending fiber tracts. J Cell Sci 123, 1652-1662. Just I, Rohrbeck A, Huelsenbeck SC, Hoeltje M (2011) Therapeutic effects of Clostridium botulinum C3 exoen- [875] [876] [877] [878] [879] [880] [881] [882] [883] [884] [885] [886] [887] [888] [889] zyme. Naunyn Schmiedebergs Arch Pharmacol 383, 247-252. Loske P, Boato F, Hendrix S, Piepgras J, Just I, hnertHilger G, Holtje M (2012) Minimal essential length of Clostridium botulinum C3 peptides to enhance neuronal regenerative growth and connectivity in a non-enzymatic mode. J Neurochem 120, 1084-1096. Jiang Y, Brody DL (2012) Administration of COG1410 reduces axonal amyloid precursor protein immunoreactivity and microglial activation after controlled cortical impact in mice. J Neurotrauma 29, 2332-2341. Ghosal K, Stathopoulos A, Thomas D, Phenis D, Vitek MP, Pimplikar SW (2012) The apolipoprotein-E-mimetic COG112 protects amyloid precursor protein intracellular domain-overexpressing animals from Alzheimer’s disease-like pathological features. Neurodegener Dis, DOI: 10.1159-000341299 [Epub ahead of print]. Corneveaux JJ, Liang WS, Reiman EM, Webster JA, Myers AJ, Zismann VL, Joshipura KD, Pearson JV, HuLince D, Craig DW, Coon KD, Dunckley T, Bandy D, Lee W, Chen K, Beach TG, Mastroeni D, Grover A, Ravid R, Sando SB, Aasly JO, Heun R, Jessen F, Kolsch H, Rogers J, Hutton ML, Melquist S, Petersen RC, Alexander GE, Caselli RJ, Papassotiropoulos A, Stephan DA, Huentelman MJ (2010) Evidence for an association between KIBRA and late-onset Alzheimer’s disease. Neurobiol Aging 31, 901-909. Hofmann SG, Smits JA, Asnaani A, Gutner CA, Otto MW (2011) Cognitive enhancers for anxiety disorders. Pharmacol Biochem Behav 99, 275-284. Marwarha G, Dasari B, Prasanthi JR, Schommer J, Ghribi O (2010) Leptin reduces the accumulation of Abeta and phosphorylated tau induced by 27-hydroxycholesterol in rabbit organotypic slices. J Alzheimers Dis 19, 1007-1019. Marwarha G, Dasari B, Prabhakara JP, Schommer J, Ghribi O (2010) beta-Amyloid regulates leptin expression and tau phosphorylation through the mTORC1 signaling pathway. J Neurochem 115, 373-384. Marwarha G, Prasanthi JR, Schommer J, Dasari B, Ghribi O (2011) Molecular interplay between leptin, insulin-like growth factor-1, and beta-amyloid in organotypic slices from rabbit hippocampus. Mol Neurodegener 6, 41. Harvey J, Shanley LJ, O’Malley D, Irving AJ (2005) Leptin: A potential cognitive enhancer? Biochem Soc Trans 33, 1029-1032. Farr SA, Banks WA, Morley JE (2006) Effects of leptin on memory processing. Peptides 27, 1420-1425. Beccano-Kelly D, Harvey J (2012) Leptin: A novel therapeutic target in Alzheimer’s disease? Int J Alzheimers Dis 2012, 594137. Perez-Gonzalez R, Antequera D, Vargas T, Spuch C, Bolos M, Carro E (2011) Leptin induces proliferation of neuronal progenitors and neuroprotection in a mouse model of Alzheimer’s disease. J Alzheimers Dis 24(Suppl 2), 17-25. Lourenco FC, Galvan V, Fombonne J, Corset V, Llambi F, Muller U, Bredesen DE, Mehlen P (2009) Netrin-1 interacts with amyloid precursor protein and regulates amyloid-beta production. Cell Death Differ 16, 655-663. Rama N, Goldschneider D, Corset V, Lambert J, Pays L, Mehlen P (2012) Amyloid precursor protein regulates netrin-1-mediated commissural axon outgrowth. J Biol Chem 287, 30014-30023. Gorba T, Bradoo P, Antonic A, Marvin K, Liu DX, Lobie PE, Reymann KG, Gluckman PD, Sieg F (2006) Neural regeneration protein is a novel chemoattractive and W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [890] [891] [892] [893] [894] [895] [896] [897] [898] [899] [900] [901] [902] [903] [904] [905] neuronal survival-promoting factor. Exp Cell Res 312, 3060-3074. Xue D, Zhao M, Wang YJ, Wang L, Yang Y, Wang SW, Zhang R, Zhao Y, Liu RT (2012) A multifunctional peptide rescues memory deficits in Alzheimer’s disease transgenic mice by inhibiting Abeta42-induced cytotoxicity and increasing microglial phagocytosis. Neurobiol Dis 46, 701-709. Schuster D, Rajendran A, Hui SW, Nicotera T, Srikrishnan T, Kruzel ML (2005) Protective effect of colostrinin on neuroblastoma cell survival is due to reduced aggregation of beta-amyloid. Neuropeptides 39, 419-426. Bourhim M, Kruzel M, Srikrishnan T, Nicotera T (2007) Linear quantitation of Abeta aggregation using Thioflavin T: Reduction in fibril formation by colostrinin. J Neurosci Methods 160, 264-268. Froud KE, Wardhaugh T, Banks D, Saffrey MJ, Stewart MG (2010) Colostrinin alleviates amyloid-beta induced toxicity in rat primary hippocampal cultures. J Alzheimers Dis 20, 423-426. Popik P, Bobula B, Janusz M, Lisowski J, Vetulani J (1999) Colostrinin, a polypeptide isolated from early milk, facilitates learning and memory in rats. Pharmacol Biochem Behav 64, 183-189. Stewart MG, Banks D (2006) Enhancement of long-term memory retention by Colostrinin in one-day-old chicks trained on a weak passive avoidance learning paradigm. Neurobiol Learn Mem 86, 66-71. Szaniszlo P, German P, Hajas G, Saenz DN, Woodberry MW, Kruzel ML, Boldogh I (2009) Effects of Colostrinin on gene expression-transcriptomal network analysis. Int Immunopharmacol 9, 181-193. Leszek J, Inglot AD, Janusz M, Byczkiewicz F, Kiejna A, Georgiades J, Lisowski J (2002) Colostrinin proline-rich polypeptide complex from ovine colostrum–a long-term study of its efficacy in Alzheimer’s disease. Med Sci Monit 8, I93-I96. Bilikiewicz A, Gaus W (2004) Colostrinin (a naturally occurring, proline-rich, polypeptide mixture) in the treatment of Alzheimer’s disease. J Alzheimers Dis 6, 17-26. Boldogh I, Kruzel ML (2008) Colostrinintrade mark: An oxidative stress modulator for prevention and treatment of age-related disorders. J Alzheimers Dis 13, 303-321. Szaniszlo P, German P, Hajas G, Saenz DN, Kruzel M, Boldogh I (2009) New insights into clinical trial for Colostrinin in Alzheimer’s disease. J Nutr Health Aging 13, 235-241. Janusz M, Zablocka A (2010) Colostral proline-rich polypeptides–immunoregulatory properties and prospects of therapeutic use in Alzheimer’s disease. Curr Alzheimer Res 7, 323-333. Stewart MG (2008) Colostrinin: A naturally occurring compound derived from mammalian colostrum with efficacy in treatment of neurodegenerative diseases, including Alzheimer’s. Expert Opin Pharmacother 9, 2553-2559. Zimecki M (2008) A proline-rich polypeptide from ovine colostrum: Colostrinin with immunomodulatory activity. Adv Exp Med Biol 606, 241-250. Taddei K, Laws SM, Verdile G, Munns S, D’Costa K, Harvey AR, Martins IJ, Hill F, Levy E, Shaw JE, Martins RN (2010) Novel phage peptides attenuate beta amyloid42 catalysed hydrogen peroxide production and associated neurotoxicity. Neurobiol Aging 31, 203-214. Hock FJ, Gerhards HJ, Wiemer G, Usinger P, Geiger R (1988) Learning and memory processes of an ACTH4-9 [906] [907] [908] [909] [910] [911] [912] [913] [914] [915] [916] [917] [918] [919] 85 analog (ebiratide; Hoe 427) in mice and rats. Peptides 9, 575-581. Wiemer G, Gerhards HJ, Hock FJ, Usinger P, Von RW, Geiger R (1988) Neurochemical effects of the synthetic ACTH4-9-analog Hoe 427 (Ebiratide) in rat brain. Peptides 9, 1081-1087. Siegfried KR (1991) First clinical impressions with an ACTH analog (HOE 427) in the treatment of Alzheimer’s disease. Ann N Y Acad Sci 640, 280-283. Manso Y, Adlard PA, Carrasco J, Vasak M, Hidalgo J (2011) Metallothionein and brain inflammation. J Biol Inorg Chem 16, 1103-1113. Manso Y, Carrasco J, Comes G, Meloni G, Adlard PA, Bush AI, Vasak M, Hidalgo J (2012) Characterization of the role of metallothionein-3 in an animal model of Alzheimer’s disease. Cell Mol Life Sci 69, 3683-3700. Pelsman A, Hoyo-Vadillo C, Gudasheva TA, Seredenin SB, Ostrovskaya RU, Busciglio J (2003) GVS-111 prevents oxidative damage and apoptosis in normal and Down’s syndrome human cortical neurons. Int J Dev Neurosci 21, 117-124. Ostrovskaya RU, Romanova GA, Barskov IV, Shanina EV, Gudasheva TA, Victorov IV, Voronina TA, Seredenin SB (1999) Memory restoring and neuroprotective effects of the proline-containing dipeptide, GVS-111, in a photochemical stroke model. Behav Pharmacol 10, 549553. Ostrovskaya RU, Mirsoev TK, Romanova GA, Gudasheva TA, Kravchenko EV, Trofimov CC, Voronina TA, Seredenin SB (2001) Proline-containing dipeptide GVS-111 retains nootropic activity after oral administration. Bull Exp Biol Med 132, 959-962. Ostrovskaya RU, Gruden MA, Bobkova NA, Sewell RD, Gudasheva TA, Samokhin AN, Seredinin SB, Noppe W, Sherstnev VV, Morozova-Roche LA (2007) The nootropic and neuroprotective proline-containing dipeptide noopept restores spatial memory and increases immunoreactivity to amyloid in an Alzheimer’s disease model. J Psychopharmacol 21, 611-619. Ostrovskaya RU, Belnik AP, Storozheva ZI (2008) Noopept efficiency in experimental Alzheimer disease (cognitive deficiency caused by beta-amyloid25-35 injection into Meynert basal nuclei of rats). Bull Exp Biol Med 146, 77-80. Ostrovskaya RU, Gudasheva TA, Zaplina AP, Vahitova JV, Salimgareeva MH, Jamidanov RS, Seredenin SB (2008) Noopept stimulates the expression of NGF and BDNF in rat hippocampus. Bull Exp Biol Med 146, 334-337. Kovalenko LP, Shipaeva EV, Alekseeva SV, Pronin AV, Durnev AD, Gudasheva TA, Ostrovskaja RU, Seredenin SB (2007) Immunopharmacological properties of noopept. Bull Exp Biol Med 144, 49-52. Neznamov GG, Teleshova ES (2009) Comparative studies of Noopept and piracetam in the treatment of patients with mild cognitive disorders in organic brain diseases of vascular and traumatic origin. Neurosci Behav Physiol 39, 311-321. Kondratenko RV, Derevyagin VI, Skrebitsky VG (2010) Novel nootropic dipeptide Noopept increases inhibitory synaptic transmission in CA1 pyramidal cells. Neurosci Lett 476, 70-73. Vorobyov V, Kaptsov V, Kovalev G, Sengpiel F (2011) Effects of nootropics on the EEG in conscious rats and their modification by glutamatergic inhibitors. Brain Res Bull 85, 123-132. 86 [920] [921] [922] [923] [924] [925] [926] [927] [928] [929] [930] [931] [932] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Jia X, Gharibyan AL, Ohman A, Liu Y, Olofsson A, Morozova-Roche LA (2011) Neuroprotective and nootropic drug noopept rescues alpha-synuclein amyloid cytotoxicity. J Mol Biol 414, 699-712. Hirano M, Hirate K, Miyake N (1998) NC-1900, a [pGlu4,Cyt6]AVP(4-9) analogue, facilitates the learning performance of rats on delayed nonmatching to sample task in Y-water maze. Methods Find Exp Clin Pharmacol 20, 567-573. Tanaka T, Yamada K, Senzaki K, Narimatsu H, Nishimura K, Kameyama T, Nabeshima T (1998) NC-1900, an active fragment analog of arginine vasopressin, improves learning and memory deficits induced by beta-amyloid protein in rats. Eur J Pharmacol 352, 135-142. 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 with transient forebrain ischemia. Brain Res Cogn Brain Res 13, 1-15. Ishida T, Sato T, Irifune M, Tanaka K, Hirate K, Nakamura N, Nishikawa T (2007) Inhibitory effect of cyclooxygenase inhibitors on the step-through passive avoidance performance in mice treated with NC-1900, an argininevasopressin fragment analog. Methods Find Exp Clin Pharmacol 29, 315-320. Sato T, Ishida T, Irifune M, Tanaka K, Hirate K, Nakamura N, Nishikawa T (2007) Effect of NC-1900, an active fragment analog of arginine vasopressin, and inhibitors of arachidonic acid metabolism on performance of a passive avoidance task in mice. Eur J Pharmacol 560, 36-41. Fotinopoulou A, Tsachaki M, Vlavaki M, Poulopoulos A, Rostagno A, Frangione B, Ghiso J, Efthimiopoulos S (2005) BRI2 interacts with amyloid precursor protein (APP) and regulates amyloid beta (Abeta) production. J Biol Chem 280, 30768-30772. Peng S, Fitzen M, Jornvall H, Johansson J (2010) The extracellular domain of Bri2 (ITM2B) binds the ABri peptide (1-23) and amyloid beta-peptide (Abeta1-40): Implications for Bri2 effects on processing of amyloid precursor protein and Abeta aggregation. Biochem Biophys Res Commun 393, 356-361. Matsuda S, Giliberto L, Matsuda Y, Davies P, McGowan E, Pickford F, Ghiso J, Frangione B, D’Adamio L (2005) The familial dementia BRI2 gene binds the Alzheimer gene amyloid-beta precursor protein and inhibits amyloid-beta production. J Biol Chem 280, 28912-28916. Matsuda S, Giliberto L, Matsuda Y, McGowan EM, D’Adamio L (2008) BRI2 inhibits amyloid beta-peptide precursor protein processing by interfering with the docking of secretases to the substrate. J Neurosci 28, 86688676. Matsuda S, Matsuda Y, Snapp EL, D’Adamio L (2011) Maturation of BRI2 generates a specific inhibitor that reduces APP processing at the plasma membrane and in endocytic vesicles. Neurobiol Aging 32, 14001408. 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 deposition in vivo. J Neurosci 28, 6030-6036. Tsachaki M, Ghiso J, Efthimiopoulos S (2008) BRI2 as a central protein involved in neurodegeneration. Biotechnol J 3, 1548-1554. [933] [934] [935] [936] [937] [938] [939] [940] [941] [942] [943] [944] [945] [946] [947] Tsachaki M, Serlidaki D, Fetani A, Zarkou V, Rozani I, Ghiso J, Efthimiopoulos S (2011) Glycosylation of BRI2 on asparagine 170 is involved in its trafficking to the cell surface but not in its processing by furin or ADAM10. Glycobiology 21, 1382-1388. Findeis MA (2007) The role of amyloid beta peptide 42 in Alzheimer’s disease. Pharmacol Ther 116, 266-286. Gunstad J, Spitznagel MB, Glickman E, Alexander T, Juvancic-Heltzel J, Walter K, Murray L (2008) betaAmyloid is associated with reduced cognitive function in healthy older adults. J Neuropsychiatry Clin Neurosci 20, 327-330. Cosentino SA, Stern Y, Sokolov E, Scarmeas N, Manly JJ, Tang MX, Schupf N, Mayeux RP (2010) Plasma beta-amyloid and cognitive decline. Arch Neurol 67, 14851490. Parihar MS, Brewer GJ (2010) Amyloid-beta as a modulator of synaptic plasticity. J Alzheimers Dis 22, 741-763. Koyama A, Okereke OI, Yang T, Blacker D, Selkoe DJ, Grodstein F (2012) Plasma amyloid-beta as a predictor of dementia and cognitive decline: A systematic review and meta-analysis. Arch Neurol 69, 824-831. Koivunen J, Karrasch M, Scheinin NM, Aalto S, Vahlberg T, Nagren K, Helin S, Viitanen M, Rinne JO (2012) Cognitive decline and amyloid accumulation in patients with mild cognitive impairment. Dement Geriatr Cogn Disord 34, 31-37. Chetelat G, Villemagne VL, Bourgeat P, Pike KE, Jones G, Ames D, Ellis KA, Szoeke C, Martins RN, O’Keefe GJ, Salvado O, Masters CL, Rowe CC (2010) Relationship between atrophy and beta-amyloid deposition in Alzheimer disease. Ann Neurol 67, 317-324. Chetelat G, Villemagne VL, Villain N, Jones G, Ellis KA, Ames D, Martins RN, Masters CL, Rowe CC (2012) Accelerated cortical atrophy in cognitively normal elderly with high beta-amyloid deposition. Neurology 78, 477484. Chetelat G, Villemagne VL, Pike KE, Ellis KA, Ames D, Masters CL, Rowe CC (2012) Relationship between memory performance and beta-amyloid deposition at different stages of Alzheimer’s disease. Neurodegener Dis 10, 141-144. Klyubin I, Cullen WK, Hu NW, Rowan MJ (2012) Alzheimer’s disease Abeta assemblies mediating rapid disruption of synaptic plasticity and memory. Mol Brain 5, 25. Cavallucci V, D’Amelio M, Cecconi F (2012) Abeta toxicity in Alzheimer’s disease. Mol Neurobiol 45, 366-378. Mucke L, Selkoe DJ (2012) Neurotoxicity of amyloid betaprotein: Synaptic and network dysfunction. Cold Spring Harb Perspect Med 2, a006338. Reitz C (2012) Alzheimer’s disease and the amyloid cascade hypothesis: A critical review. Int J Alzheimers Dis 2012, 369808. Nelson PT, Alafuzoff I, Bigio EH, Bouras C, Braak H, Cairns NJ, Castellani RJ, Crain BJ, Davies P, Del TK, Duyckaerts C, Frosch MP, Haroutunian V, Hof PR, Hulette CM, Hyman BT, Iwatsubo T, Jellinger KA, Jicha GA, Kovari E, Kukull WA, Leverenz JB, Love S, Mackenzie IR, Mann DM, Masliah E, McKee AC, Montine TJ, Morris JC, Schneider JA, Sonnen JA, Thal DR, Trojanowski JQ, Troncoso JC, Wisniewski T, Woltjer RL, Beach TG (2012) Correlation of Alzheimer disease neuropathologic changes with cognitive status: A review of the literature. J Neuropathol Exp Neurol 71, 362-381. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [948] [949] [950] [951] [952] [953] [954] [955] [956] [957] [958] [959] [960] [961] [962] [963] [964] [965] [966] [967] [968] [969] Kline A (2012) Apolipoprotein E, amyloid-beta clearance and therapeutic opportunities in Alzheimer’s disease. Alzheimers Res Ther 4, 32. Soto C, Kindy MS, Baumann M, Frangione B (1996) Inhibition of Alzheimer’s amyloidosis by peptides that prevent beta-sheet conformation. Biochem Biophys Res Commun 226, 672-680. Soto C, Sigurdsson EM, Morelli L, Kumar RA, Castano EM, Frangione B (1998) Beta-sheet breaker peptides inhibit fibrillogenesis in a rat brain model of amyloidosis: Implications for Alzheimer’s therapy. Nat Med 4, 822826. Zagorski MG (1998) Amyloid aggregation inhibitors. IDrugs 1, 17-18. Findeis MA (1999) -Amyloid aggregation inhibitors. Current Opin CNS Invest Druigs 1, 333-339. Findeis MA, Musso GM, rico-Muendel CC, Benjamin HW, Hundal AM, Lee JJ, Chin J, Kelley M, Wakefield J, Hayward NJ, Molineaux SM (1999) Modified-peptide inhibitors of amyloid beta-peptide polymerization. Biochemistry 38, 6791-6800. Soto C (1999) Plaque busters: Strategies to inhibit amyloid formation in Alzheimer’s disease. Mol Med Today 5, 343350. Soto C (1999) b-Amyloid disrupting drugs. CNS Drugs 12, 347-356. Moore CI, Wolfe MS (1999) Inhibition of -amyloid formation as a therapeutic strategy. Exp Opin Ther Patents 9, 135-146. Findeis MA, Lee JJ, Kelley M, Wakefield JD, Zhang MH, Chin J, Kubasek W, Molineaux SM (2001) Characterization of cholyl-leu-val-phe-phe-ala-OH as an inhibitor of amyloid beta-peptide polymerization. Amyloid 8, 231-241. Adessi C, Soto C (2002) Beta-sheet breaker strategy for the treatment of Alzheimer’s disease. Drug Dev Res 56, 184-193. Findeis MA (2002) Peptide inhibitors of beta amyloid aggregation. Curr Top Med Chem 2, 417-423. Levine H (2002) The challenge of inhibiting Abeta polymerization. Curr Med Chem 9, 1121-1133. Levine H III (2005) Multiple ligand binding sites on A beta(1-40) fibrils. Amyloid 12, 5-14. Soto C, Estrada L (2005) Amyloid inhibitors and betasheet breakers. Subcell Biochem 38, 351-364. Lansbury PT, Lashuel HA (2006) A century-old debate on protein aggregation and neurodegeneration enters the clinic. Nature 443, 774-779. Levine H III (2007) Small molecule inhibitors of Abeta assembly. Amyloid 14, 185-197. Stains CI, Mondal K, Ghosh I (2007) Molecules that target beta-amyloid. ChemMedChem 2, 1674-1692. Hawkes CA, Ng V, McLaurin J (2009) Small molecule inhibitors of A-aggregation and neurotoxicity. Drug Dev Res 70, 111-124. Haydar SN, Yun H, Staal RGW, Hirst WD (2009) Small-molecule protein-protein interaction inhibitors as therapeutic agents for neurodegenerative diseases: Recent progress and future directions. Ann Rep Med Chem 44, 51-69. Yadav A, Sonker M (2009) Perspectives in designing anti aggregation agents as Alzheimer disease drugs. Eur J Med Chem 44, 3866-3873. Kim HY, Kim Y, Han G, Kim DJ (2010) Regulation of in vitro Abeta1-40 aggregation mediated by small molecules. J Alzheimers Dis 22, 73-85. [970] [971] [972] [973] [974] [975] [976] [977] [978] [979] [980] [981] [982] [983] [984] [985] 87 Butterfield SM, Lashuel HA (2010) Amyloidogenic protein-membrane interactions: Mechanistic insight from model systems. Angew Chem Int Ed Engl 49, 56285654. Scherzer-Attali R, Pellarin R, Convertino M, FrydmanMarom A, Egoz-Matia N, Peled S, Levy-Sakin M, Shalev DE, Caflisch A, Gazit E, Segal D (2010) Complete phenotypic recovery of an Alzheimer’s disease model by a quinone-tryptophan hybrid aggregation inhibitor. PLoS One 5, e11101. Dorgeret B, Khemtemourian L, Correia I, Soulier JL, Lequin O, Ongeri S (2011) Sugar-based peptidomimetics inhibit amyloid beta-peptide aggregation. Eur J Med Chem 46, 5959-5969. Richman M, Wilk S, Skirtenko N, Perelman A, Rahimipour S (2011) Surface-modified protein microspheres capture amyloid-beta and inhibit its aggregation and toxicity. Chemistry 17, 11171-11177. Butterfield S, Hejjaoui M, Fauvet B, Awad L, Lashuel HA (2012) Chemical strategies for controlling protein folding and elucidating the molecular mechanisms of amyloid formation and toxicity. J Mol Biol 421, 204-236. Scherzer-Attali R, Shaltiel-Karyo R, Adalist YH, Segal D, Gazit E (2012) Generic inhibition of amyloidogenic proteins by two naphthoquinone-tryptophan hybrid molecules. Proteins 80, 1962-1973. Scherzer-Attali R, Farfara D, Cooper I, Levin A, BenRomano T, Trudler D, Vientrov M, Shaltiel-Karyo R, Shalev DE, Segev-Amzaleg N, Gazit E, Segal D, Frenkel D (2012) Naphthoquinone-tyrptophan reduces neurotoxic Abeta*56 levels and improves cognition in Alzheimer’s disease animal model. Neurobiol Dis 46, 663-672. Ratner M (2009) Spotlight focuses on protein-misfolding therapies. Nat Biotechnol 27, 874. Plante-Bordeneuve V, Said G (2011) Familial amyloid polyneuropathy. Lancet Neurol 10, 1086-1097. Yamamoto T, Muto K, Komiyama M, Canivet J, Yamaguchi J, Itami K (2011) Nickel-catalyzed C-H arylation of azoles with haloarenes: Scope, mechanism, and applications to the synthesis of bioactive molecules. Chemistry 17, 10113-10122. Bulawa CE, Connelly S, Devit M, Wang L, Weigel C, Fleming JA, Packman J, Powers ET, Wiseman RL, Foss TR, Wilson IA, Kelly JW, Labaudiniere R (2012) Tafamidis, a potent and selective transthyretin kinetic stabilizer that inhibits the amyloid cascade. Proc Natl Acad Sci U S A 109, 9629-9634. Du J, Cho PY, Yang DT, Murphy RM (2012) Identification of beta-amyloid-binding sites on transthyretin. Protein Eng Des Sel 25, 337-345. Johnson SM, Connelly S, Fearns C, Powers ET, Kelly JW (2012) The transthyretin amyloidoses: From delineating the molecular mechanism of aggregation linked to pathology to a regulatory-agency-approved drug. J Mol Biol 421, 185-203. Buxbaum JN, Linke RP (2012) A molecular history of the amyloidoses. J Mol Biol 421, 142-159. Dember LM, Hawkins PN, Hazenberg BP, Gorevic PD, Merlini G, Butrimiene I, Livneh A, Lesnyak O, Puechal X, Lachmann HJ, Obici L, Balshaw R, Garceau D, Hauck W, Skinner M (2007) Eprodisate for the treatment of renal disease in AA amyloidosis. N Engl J Med 356, 23492360. Manenti L, Tansinda P, Vaglio A (2007) Eprodisate in AA amyloidosis. N Engl J Med 357, 1153-1154. 88 [986] [987] [988] [989] [990] [991] [992] [993] [994] [995] [996] [997] [998] [999] [1000] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Manenti L, Tansinda P, Vaglio A (2008) Eprodisate in amyloid A amyloidosis: A novel therapeutic approach? Expert Opin Pharmacother 9, 2175-2180. Rule AD, Leung N (2007) Eprodisate slows the progression of renal disease in patients with AA amyloidosis. Nat Clin Pract Nephrol 3, 592-593. Loeb MB, Molloy DW, Smieja M, Standish T, Goldsmith CH, Mahony J, Smith S, Borrie M, Decoteau E, Davidson W, McDougall A, Gnarpe J, O’DONNell M, Chernesky M (2004) A randomized, controlled trial of doxycycline and rifampin for patients with Alzheimer’s disease. J Am Geriatr Soc 52, 381-387. Salloway S, Sperling R, Keren R, Porsteinsson AP, van Dyck CH, Tariot PN, Gilman S, Arnold D, Abushakra S, Hernandez C, Crans G, Liang E, Quinn G, Bairu M, Pastrak A, Cedarbaum JM (2011) A phase 2 randomized trial of ELND005, scyllo-inositol, in mild to moderate Alzheimer disease. Neurology 77, 1253-1262. McLaurin J, Golomb R, Jurewicz A, Antel JP, Fraser PE (2000) Inositol stereoisomers stabilize an oligomeric aggregate of Alzheimer amyloid beta peptide and inhibit abeta -induced toxicity. J Biol Chem 275, 18495-18502. 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, Fraser PE, Westaway D, St George-Hyslop P (2006) Cyclohexanehexol inhibitors of Abeta aggregation prevent and reverse Alzheimer phenotype in a mouse model. Nat Med 12, 801-808. Fenili D, Brown M, Rappaport R, McLaurin J (2007) Properties of scyllo-inositol as a therapeutic treatment of AD-like pathology. J Mol Med (Berl) 85, 603-611. Hawkes CA, Deng LH, Shaw JE, Nitz M, McLaurin J (2010) Small molecule beta-amyloid inhibitors that stabilize protofibrillar structures in vitro improve cognition and pathology in a mouse model of Alzheimer’s disease. Eur J Neurosci 31, 203-213. Townsend M, Cleary JP, Mehta T, Hofmeister J, Lesne S, O’Hare E, Walsh DM, Selkoe DJ (2006) Orally available compound prevents deficits in memory caused by the Alzheimer amyloid-beta oligomers. Ann Neurol 60, 668-676. Griffith HR, Den Hollander JA, Stewart CC, Evanochko WT, Buchthal SD, Harrell LE, Zamrini EY, Brockington JC, Marson DC (2007) Elevated brain scyllo-inositol concentrations in patients with Alzheimer’s disease. NMR Biomed 20, 709-716. Sun Y, Zhang G, Hawkes CA, Shaw JE, McLaurin J, Nitz M (2008) Synthesis of scyllo-inositol derivatives and their effects on amyloid beta peptide aggregation. Bioorg Med Chem 16, 7177-7184. Vasdev N, Chio J, van Oosten EM, Nitz M, McLaurin J, 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. Sinha S, Du Z, Maiti P, Klarner FG, Schrader T, Wang C, Bitan G (2012) Comparison of Three Amyloid Assembly Inhibitors: The Sugar scyllo-Inositol, the Polyphenol Epigallocatechin Gallate, and the Molecular Tweezer CLR01. ACS Chem Neurosci 3, 451-458. Gunther EC, Strittmatter SM (2010) Beta-amyloid oligomers and cellular prion protein in Alzheimer’s disease. J Mol Med (Berl) 88, 331-338. Lauren J, Gimbel DA, Nygaard HB, Gilbert JW, Strittmatter SM (2009) Cellular prion protein mediates impairment [1001] [1002] [1003] [1004] [1005] [1006] [1007] [1008] [1009] [1010] [1011] [1012] [1013] [1014] of synaptic plasticity by amyloid-beta oligomers. Nature 457, 1128-1132. Nygaard HB, Strittmatter SM (2009) Cellular prion protein mediates the toxicity of beta-amyloid oligomers: Implications for Alzheimer disease. Arch Neurol 66, 13251328. Chung E, Ji Y, Sun Y, Kascsak RJ, Kascsak RB, Mehta PD, Strittmatter SM, Wisniewski T (2010) Anti-PrPC monoclonal antibody infusion as a novel treatment for cognitive deficits in an Alzheimer’s disease model mouse. BMC Neurosci 11, 130. Gimbel DA, Nygaard HB, Coffey EE, Gunther EC, Lauren J, Gimbel ZA, Strittmatter SM (2010) Memory impairment in transgenic Alzheimer mice requires cellular prion protein. J Neurosci 30, 6367-6374. Kroth H, Ansaloni A, Varisco Y, Jan A, Sreenivasachary N, Rezaei-Ghaleh N, Giriens V, Lohmann S, Lopez-Deber MP, 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 and structure activity relationship of small molecule inhibitors of toxic beta-amyloid-42 fibril formation. J Biol Chem 287, 34786-34800. Rzepecki P, Wehner M, Molt O, Zadmard R, Harms K, Schrader T (2003) Aminopyrazole oligomers for betasheet stabilization of peptides. Synthesis, 1815-1826. Rzepecki P, Nagel-Steger L, Feuerstein S, Linne U, Molt O, Zadmard R, Aschermann K, Wehner M, Schrader T, Riesner D (2004) Prevention of Alzheimer’s diseaseassociated Abeta aggregation by rationally designed nonpeptidic beta-sheet ligands. J Biol Chem 279, 4749747505. Nagel-Steger L, Demeler B, Meyer-Zaika W, Hochdorffer K, Schrader T, Willbold D (2010) Modulation of aggregate size- and shape-distributions of the amyloid-beta peptide by a designed beta-sheet breaker. Eur Biophys J 39, 415422. Rzepecki P, Hochdorffer K, Schaller T, Zienau J, Harms K, Ochsenfeld C, Xie X, Schrader T (2008) Hierarchical self-assembly of aminopyrazole peptides into nanorosettes in water. J Am Chem Soc 130, 586-591. Hochdorffer K, Marz-Berberich J, Nagel-Steger L, Epple M, Meyer-Zaika W, Horn AH, Sticht H, Sinha S, Bitan G, Schrader T (2011) Rational design of beta-sheet ligands against Abeta42-induced toxicity. J Am Chem Soc 133, 4348-4358. Rzepecki P, Geib N, Peifer M, Biesemeier F, Schrader T (2007) Synthesis and binding studies of Alzheimer ligands on solid support. J Org Chem 72, 3614-3624. Cernovska K, Kemter M, Gallmeier HC, Rzepecki P, Schrader T, Konig B (2004) PEG-supported synthesis of pyrazole oligoamides with peptide beta-sheet affinity. Org Biomol Chem 2, 1603-1611. Rzepecki P, Schrader T (2005) beta-Sheet ligands in action: KLVFF recognition by aminopyrazole hybrid receptors in water. J Am Chem Soc 127, 3016-3025. Fricke H, Gerlach A, Unterberg C, Wehner M, Schrader T, Gerhards M (2009) Interactions of small protected peptides with aminopyrazole derivatives: The efficiency of blocking a beta-sheet model in the gas phase. Angew Chem Int Ed Engl 48, 900-904. Lecanu L, Tillement L, Rammouz G, Tillement JP, Greeson J, Papadopoulos V (2009) Caprospinol: Moving from a neuroactive steroid to a neurotropic drug. Expert Opin Investig Drugs 18, 265-276. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1015] [1016] [1017] [1018] [1019] [1020] [1021] [1022] [1023] [1024] [1025] [1026] [1027] [1028] Lecanu L, Rammouz G, McCourty A, Sidahmed EK, Greeson J, Papadopoulos V (2010) Caprospinol reduces amyloid deposits and improves cognitive function in a rat model of Alzheimer’s disease. Neuroscience 165, 427-435. Tillement L, Lecanu L, Papadopoulos V (2011) Further evidence on mitochondrial targeting of beta-amyloid and specificity of beta-amyloid-induced mitotoxicity in neurons. Neurodegener Dis 8, 331-344. Papadopoulos V, Lecanu L (2012) Caprospinol: Discovery of a steroid drug candidate to treat Alzheimer’s disease based on 22R-hydroxycholesterol structure and properties. J Neuroendocrinol 24, 93-101. Howlett DR, George AR, Owen DE, Ward RV, Markwell RE (1999) Common structural features determine the effectiveness of carvedilol, daunomycin and rolitetracycline as inhibitors of Alzheimer beta-amyloid fibril formation. Biochem J 343 Pt 2, 419-423. Fokkens M, Schrader T, Klarner FG (2005) A molecular tweezer for lysine and arginine. J Am Chem Soc 127, 14415-14421. 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) Lysinespecific molecular tweezers are broad-spectrum inhibitors of assembly and toxicity of amyloid proteins. J Am Chem Soc 133, 16958-16969. Prabhudesai S, Sinha S, Attar A, Kotagiri A, Fitzmaurice AG, Lakshmanan R, Ivanova MI, Loo JA, Klarner FG, Schrader T, Stahl M, Bitan G, Bronstein JM (2012) A novel “molecular tweezer” inhibitor of alpha-synuclein neurotoxicity in vitro and in vivo. Neurotherapeutics 9, 464-476. Klarner FG, Schrader T (2012) Aromatic interactions by molecular tweezers and clips in chemical and biological systems. Acc Chem Res, doi: 10.1021/ar300061c [Epub ahead of print]. Talbiersky P, Bastkowski F, Klarner FG, Schrader T (2008) Molecular clip and tweezer introduce new mechanisms of enzyme inhibition. J Am Chem Soc 130, 9824-9828. Klarner FG, Kahlert B, Nellesen A, Zienau J, Ochsenfeld C, Schrader T (2006) Molecular tweezer and clip in aqueous solution: Unexpected self-assembly, powerful host-guest complex formation, quantum chemical 1H NMR shift calculation. J Am Chem Soc 128, 4831-4841. Echeverria V, Zeitlin R (2012) Cotinine: A potential new therapeutic agent against Alzheimer’s disease. CNS Neurosci Ther 18, 517-523. Bergamaschini L, Rossi E, Storini C, Pizzimenti S, Distaso M, Perego C, De LA, Vergani C, De Simoni MG (2004) Peripheral treatment with enoxaparin, a low molecular weight heparin, reduces plaques and beta-amyloid accumulation in a mouse model of Alzheimer’s disease. J Neurosci 24, 4181-4186. Timmer NM, Herbert MK, Kleinovink JW, Kiliaan AJ, de Waal RM, Verbeek MM (2010) Limited expression of heparan sulphate proteoglycans associated with Abeta deposits in the APPswe/PS1dE9 mouse model for Alzheimer’s disease. Neuropathol Appl Neurobiol 36, 478-486. Timmer NM, van DL, van der Zee CE, Kiliaan A, de Waal RM, Verbeek MM (2010) Enoxaparin treatment administered at both early and late stages of amyloid beta deposition improves cognition of APPswe/PS1dE9 mice with differential effects on brain Abeta levels. Neurobiol Dis 40, 340-347. [1029] [1030] [1031] [1032] [1033] [1034] [1035] [1036] [1037] [1038] [1039] [1040] [1041] [1042] [1043] [1044] 89 Matharu B, Gibson G, Parsons R, Huckerby TN, Moore SA, Cooper LJ, Millichamp R, Allsop D, Austen B (2009) Galantamine inhibits beta-amyloid aggregation and cytotoxicity. J Neurol Sci 280, 49-58. Petrlova J, Kalai T, Maezawa I, Altman R, Harishchandra G, Hong HS, Bricarello DA, Parikh AN, Lorigan GA, Jin LW, Hideg K, Voss JC (2012) The influence of spin-labeled fluorene compounds on the assembly and toxicity of the abeta Peptide. PLoS One 7, e35443. Chavant F, Deguil J, Pain S, Ingrand I, Milin S, Fauconneau B, Perault-Pochat MC, Lafay-Chebassier C (2010) Imipramine, in part through tumor necrosis factor alpha inhibition, prevents cognitive decline and beta-amyloid accumulation in a mouse model of Alzheimer’s disease. J Pharmacol Exp Ther 332, 505-514. Byun JH, Kim H, Kim Y, Mook-Jung I, Kim DJ, Lee WK, Yoo KH (2008) Aminostyrylbenzofuran derivatives as potent inhibitors for Abeta fibril formation. Bioorg Med Chem Lett 18, 5591-5593. Lee YS, Kim HY, Kim Y, Seo JH, Roh EJ, Han H, Shin KJ (2012) Small molecules that protect against betaamyloid-induced cytotoxicity by inhibiting aggregation of beta-amyloid. Bioorg Med Chem 20, 4921-4935. 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 death and improves cognitive impairment in Alzheimer’s disease models. Neuropsychopharmacology 32, 2393-2404. Burgos-Ramos E, Puebla-Jimenez L, Rilla-Ferreiro E (2008) Minocycline provides protection against betaamyloid(25-35)-induced alterations of the somatostatin signaling pathway in the rat temporal cortex. Neuroscience 154, 1458-1466. Burgos-Ramos E, Puebla-Jimenez L, Rilla-Ferreiro E (2009) Minocycline prevents Abeta(25-35)-induced reduction of somatostatin and neprilysin content in rat temporal cortex. Life Sci 84, 205-210. Seabrook TJ, Jiang L, Maier M, Lemere CA (2006) Minocycline affects microglia activation, Abeta deposition, and behavior in APP-tg mice. Glia 53, 776-782. Fan R, Xu F, Previti ML, Davis J, Grande AM, Robinson JK, Van Nostrand WE (2007) Minocycline reduces microglial activation and improves behavioral deficits in a transgenic model of cerebral microvascular amyloid. J Neurosci 27, 3057-3063. Parachikova A, Vasilevko V, Cribbs DH, LaFerla FM, Green KN (2010) Reductions in amyloid-beta-derived neuroinflammation, with minocycline, restore cognition but do not significantly affect tau hyperphosphorylation. J Alzheimers Dis 21, 527-542. Kreutzmann P, Wolf G, Kupsch K (2010) Minocycline recovers MTT-formazan exocytosis impaired by amyloid beta peptide. Cell Mol Neurobiol 30, 979-984. Noble W, Garwood C, Stephenson J, Kinsey AM, Hanger DP, Anderton BH (2009) Minocycline reduces the development of abnormal tau species in models of Alzheimer’s disease. FASEB J 23, 739-750. Noble W, Garwood CJ, Hanger DP (2009) Minocycline as a potential therapeutic agent in neurodegenerative disorders characterised by protein misfolding. Prion 3, 78-83. Garwood CJ, Cooper JD, Hanger DP, Noble W (2010) Anti-inflammatory impact of minocycline in a mouse model of tauopathy. Front Psychiatr 1, 136. Song Y, Wei EQ, Zhang WP, Ge QF, Liu JR, Wang ML, Huang XJ, Hu X, Chen Z (2006) Minocycline protects 90 [1045] [1046] [1047] [1048] [1049] [1050] [1051] [1052] [1053] [1054] [1055] [1056] [1057] [1058] [1059] 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 inhibiting 5-lipoxygenase activation. Brain Res 1085, 57-67. Hashimoto K (2011) Can minocycline prevent the onset of Alzheimer’s disease? Ann Neurol 69, 739-740. Chaudhry IB, Hallak J, Husain N, Minhas F, Stirling J, Richardson P, Dursun S, Dunn G, Deakin B (2012) Minocycline benefits negative symptoms in early schizophrenia: A randomised double-blind placebocontrolled clinical trial in patients on standard treatment. J Psychopharmacol 26, 1185-1193. Tomiyama T, Asano S, Suwa Y, Morita T, Kataoka K, Mori H, Endo N (1994) Rifampicin prevents the aggregation and neurotoxicity of amyloid beta protein in vitro. Biochem Biophys Res Commun 204, 76-83. Tomiyama T, Shoji A, Kataoka K, Suwa Y, Asano S, Kaneko H, Endo N (1996) Inhibition of amyloid beta protein aggregation and neurotoxicity by rifampicin. Its possible function as a hydroxyl radical scavenger. J Biol Chem 271, 6839-6844. Tomiyama T, Kaneko H, Kataoka K, Asano S, Endo N (1997) Rifampicin inhibits the toxicity of pre-aggregated amyloid peptides by binding to peptide fibrils and preventing amyloid-cell interaction. Biochem J 322(Pt 3), 859-865. Qosa H, Abuznait AH, Hill RA, Kaddoumi A (2012) Enhanced brain amyloid-beta clearance by rifampicin and caffeine as a possible protective mechanism against Alzheimer’s disease. J Alzheimers Dis 31, 151-165. Forloni G, Colombo L, Girola L, Tagliavini F, Salmona M (2001) Anti-amyloidogenic activity of tetracyclines: Studies in vitro. FEBS Lett 487, 404-407. 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 elegans from beta amyloid-induced toxicity by targeting oligomers. Neurobiol Dis 40, 424-431. Averback P (1998) Spherons as a drug target in Alzheimer’s disease. Drug News Perspect 11, 469-479. Averback P, Morse D, Ghanbari H (1998) Bursting dense microspheres (spherons) in Alzheimer’s disease: A review of studies (1980–1997) on spherons and the pathogenesis of Alzheimer’s disease. J Alzheimers Dis 1, 1-34. Garofalo AW, Wone DW, Phuc A, Audia JE, Bales CA, Dovey HF, Dressen DB, Folmer B, Goldbach EG, Guinn AC, Latimer LH, Mabry TE, Nissen JS, Pleiss MA, Sohn S, Thorsett ED, Tung JS, Wu J (2002) A series of C-terminal amino alcohol dipeptide A beta inhibitors. Bioorg Med Chem Lett 12, 3051-3053. Friedman OM, Matsudaira P, Reis AH Jr, Simister N, Correia I, Kew D, Wei JY, Pochapsky T (2007) Substituted organosiloxanes as potential therapeutics for treatment and prevention of neurodegenerative diseases. J Alzheimers Dis 11, 291-300. Wood SJ, MacKenzie L, Maleeff B, Hurle MR, Wetzel R (1996) Selective inhibition of Abeta fibril formation. J Biol Chem 271, 4086-4092. Howlett DR, Perry AE, Godfrey F, Swatton JE, Jennings KH, Spitzfaden C, Wadsworth H, Wood SJ, Markwell RE (1999) Inhibition of fibril formation in beta-amyloid peptide by a novel series of benzofurans. Biochem J 340(Pt 1), 283-289. Parker MH, Chen R, Conway KA, Lee DH, Luo C, Boyd RE, Nortey SO, Ross TM, Scott MK, Reitz AB (2002) Synthesis of (-)-5,8-dihydroxy-3R-methyl2R-(dipropylamino)-1,2,3,4-tetrahydronaphthale ne: An [1060] [1061] [1062] [1063] [1064] [1065] [1066] [1067] [1068] [1069] [1070] [1071] [1072] [1073] [1074] inhibitor of beta-amyloid(1-42) aggregation. Bioorg Med Chem 10, 3565-3569. 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 structureactivity relationship of substituted N-phenyl anthranilic acid analogs as amyloid aggregation inhibitors. Bioorg Med Chem Lett 19, 654-657. Merlini G, Ascari E, Amboldi N, Bellotti V, Arbustini E, Perfetti V, Ferrari M, Zorzoli I, Marinone MG, Garini P, et al. (1995) Interaction of the anthracycline 4 -iodo4 -deoxydoxorubicin with amyloid fibrils: Inhibition of amyloidogenesis. Proc Natl Acad Sci U S A 92, 2959-2963. Higaki JN, Chakravarty S, Bryant CM, Cowart LR, Harden P, Scardina JM, Mavunkel B, Luedtke GR, Cordell B (1999) A combinatorial approach to the identification of dipeptide aldehyde inhibitors of beta-amyloid production. J Med Chem 42, 3889-3898. Kisilevsky R, Lemieux LJ, Fraser PE, Kong X, Hultin PG, Szarek WA (1995) Arresting amyloidosis in vivo using small-molecule anionic sulphonates or sulphates: Implications for Alzheimer’s disease. Nat Med 1, 143-148. Kisilevsky R (1996) Anti-amyloid drugs: Potential in the treatment of diseases associated with aging. Drugs Aging 8, 75-83. Kisilevsky R (1997) Can deposition of amyloid be prevented in Alzheimer’s disease? Ann N Y Acad Sci 826, 117-127. Tjernberg LO, Lilliehook C, Callaway DJ, Naslund J, Hahne S, Thyberg J, Terenius L, Nordstedt C (1997) Controlling amyloid beta-peptide fibril formation with protease-stable ligands. J Biol Chem 272, 12601-12605. Banks WA, Farr SA, Butt W, Kumar VB, Franko MW, Morley JE (2001) Delivery across the blood-brain barrier of antisense directed against amyloid beta: Reversal of learning and memory deficits in mice overexpressing amyloid precursor protein. J Pharmacol Exp Ther 297, 1113-1121. Heal JR, Roberts GW, Christie G, Miller AD (2002) Inhibition of beta-amyloid aggregation and neurotoxicity by complementary (antisense) peptides. Chembiochem 3, 8692. Gestwicki JE, Crabtree GR, Graef IA (2004) Harnessing chaperones to generate small-molecule inhibitors of amyloid beta aggregation. Science 306, 865-869. Lin SJ, Shiao YJ, Chi CW, Yang LM (2004) Abeta aggregation inhibitors. Part 1: Synthesis and biological activity of phenylazo benzenesulfonamides. Bioorg Med Chem Lett 14, 1173-1176. Bose M, Gestwicki JE, Devasthali V, Crabtree GR, Graef IA (2005) ‘Nature-inspired’ drug-protein complexes as inhibitors of Abeta aggregation. Biochem Soc Trans 33, 543-547. Lee KH, Shin BH, Shin KJ, Kim DJ, Yu J (2005) A hybrid molecule that prohibits amyloid fibrils and alleviates neuronal toxicity induced by beta-amyloid (1-42). Biochem Biophys Res Commun 328, 816-823. Schwarzman AL, Tsiper M, Gregori L, Goldgaber D, Frakowiak J, Mazur-Kolecka B, Taraskina A, Pchelina S, Vitek MP (2005) Selection of peptides binding to the amyloid -protein reveals potential inhibitors of amyloid formation. Amyloid 12, 199-209. Cohen T, Frydman-Marom A, Rechter M, Gazit E (2006) Inhibition of amyloid fibril formation and cytotoxicity by hydroxyindole derivatives. Biochemistry 45, 4727-4735. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1075] [1076] [1077] [1078] [1079] [1080] [1081] [1082] [1083] [1084] [1085] [1086] [1087] [1088] [1089] [1090] [1091] Dolphin GT, Dumy P, Garcia J (2006) Control of amyloid beta-peptide protofibril formation by a designed template assembly. Angew Chem Int Ed Engl 45, 2699-2702. Etienne MA, Aucoin JP, Fu Y, McCarley RL, Hammer RP (2006) Stoichiometric inhibition of amyloid betaprotein aggregation with peptides containing alternating alpha,alpha-disubstituted amino acids. J Am Chem Soc 128, 3522-3523. Inbar P, Yang J (2006) Inhibiting protein-amyloid interactions with small molecules: A surface chemistry approach. Bioorg Med Chem Lett 16, 1076-1079. Inbar P, Li CQ, Takayama SA, Bautista MR, Yang J (2006) Oligo(ethylene glycol) derivatives of thioflavin T as inhibitors of protein-amyloid interactions. Chembiochem 7, 1563-1566. Kwak JW, Kim HK, Chae CB (2006) Potential lead for an Alzheimer drug: A peptide that blocks intermolecular interaction and amyloid beta protein-induced cytotoxicity. J Med Chem 49, 4813-4817. Orner BP, Liu L, Murphy RM, Kiessling LL (2006) Phage display affords peptides that modulate beta-amyloid aggregation. J Am Chem Soc 128, 11882-11889. Pai AS, Rubinstein I, Onyuksel H (2006) PEGylated phospholipid nanomicelles interact with beta-amyloid(1-42) and mitigate its beta-sheet formation, aggregation and neurotoxicity in vitro. Peptides 27, 2858-2866. Porat Y, Abramowitz A, Gazit E (2006) Inhibition of amyloid fibril formation by polyphenols: Structural similarity and aromatic interactions as a common inhibition mechanism. Chem Biol Drug Des 67, 27-37. Byeon SR, Lee JH, Sohn JH, Kim DC, Shin KJ, Yoo KH, Mook-Jung I, Lee WK, Kim DJ (2007) Bis-styrylpyridine and bis-styrylbenzene derivatives as inhibitors for Abeta fibril formation. Bioorg Med Chem Lett 17, 1466-1470. Byeon SR, Jin YJ, Lim SJ, Lee JH, Yoo KH, Shin KJ, Oh SJ, Kim DJ (2007) Ferulic acid and benzothiazole dimer derivatives with high binding affinity to beta-amyloid fibrils. Bioorg Med Chem Lett 17, 4022-4025. Dolphin GT, Ouberai M, Dumy P, Garcia J (2007) Designed amyloid beta peptide fibril –a tool for highthroughput screening of fibril inhibitors. ChemMedChem 2, 1613-1623. Chafekar SM, Malda H, Merkx M, Meijer EW, Viertl D, Lashuel HA, Baas F, Scheper W (2007) Branched KLVFF tetramers strongly potentiate inhibition of beta-amyloid aggregation. Chembiochem 8, 1857-1864. Etienne MA, Edwin NJ, Aucoin JP, Russo PS, McCarley RL, Hammer RP (2007) Beta-amyloid protein aggregation. Methods Mol Biol 386, 203-225. Flaherty DP, Walsh SM, Kiyota T, Dong Y, Ikezu T, Vennerstrom JL (2007) Polyfluorinated bis-styrylbenzene beta-amyloid plaque binding ligands. J Med Chem 50, 4986-4992. Honson NS, Johnson RL, Huang W, Inglese J, Austin CP, Kuret J (2007) Differentiating Alzheimer diseaseassociated aggregates with small molecules. Neurobiol Dis 28, 251-260. Necula M, Kayed R, Milton S, Glabe CG (2007) Small molecule inhibitors of aggregation indicate that amyloid beta oligomerization and fibrillization pathways are independent and distinct. J Biol Chem 282, 10311-10324. Okuno H, Mori K, Okada T, Yokoyama Y, Suzuki H (2007) Development of aggregation inhibitors for amyloid-beta peptides and their evaluation by quartz-crystal microbalance. Chem Biol Drug Des 69, 356-361. [1092] [1093] [1094] [1095] [1096] [1097] [1098] [1099] [1100] [1101] [1102] [1103] [1104] [1105] [1106] 91 Riviere C, Richard T, Quentin L, Krisa S, Merillon JM, Monti JP (2007) Inhibitory activity of stilbenes on Alzheimer’s beta-amyloid fibrils in vitro. Bioorg Med Chem 15, 1160-1167. Sato J, Takahashi T, Oshima H, Matsumura S, Mihara H (2007) Design of peptides that form amyloid-like fibrils capturing amyloid beta1-42 peptides. Chemistry 13, 77457752. Takahashi T, Ohta K, Mihara H (2007) Embedding the amyloid beta-peptide sequence in green fluorescent protein inhibits Abeta oligomerization. Chembiochem 8, 985-988. Austen BM, Paleologou KE, Ali SA, Qureshi MM, Allsop D, El-Agnaf OM (2008) Designing peptide inhibitors for oligomerization and toxicity of Alzheimer’s beta-amyloid peptide. Biochemistry 47, 1984-1992. Bravo R, Arimon M, Valle-Delgado JJ, Garcia R, Durany N, Castel S, Cruz M, Ventura S, Fernandez-Busquets X (2008) Sulfated polysaccharides promote the assembly of amyloid beta(1-42) peptide into stable fibrils of reduced cytotoxicity. J Biol Chem 283, 32471-32483. 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 by polymeric nanoparticles. J Am Chem Soc 130, 1543715443. Cellamare S, Stefanachi A, Stolfa DA, Basile T, Catto M, Campagna F, Sotelo E, Acquafredda P, Carotti A (2008) Design, synthesis, and biological evaluation of glycine-based molecular tongs as inhibitors of Abeta1-40 aggregation in vitro. Bioorg Med Chem 16, 4810-4822. Dolphin GT, Chierici S, Ouberai M, Dumy P, Garcia J (2008) A multimeric quinacrine conjugate as a potential inhibitor of Alzheimer’s beta-amyloid fibril formation. Chembiochem 9, 952-963. Esteras-Chopo A, Pastor MT, Serrano L, Lopez de la PM (2008) New strategy for the generation of specific D-peptide amyloid inhibitors. J Mol Biol 377, 1372-1381. Feng BY, Toyama BH, Wille H, Colby DW, Collins SR, May BC, Prusiner SB, Weissman J, Shoichet BK (2008) Small-molecule aggregates inhibit amyloid polymerization. Nat Chem Biol 4, 197-199. Lee JH, Byeon SR, Lim SJ, Oh SJ, Moon DH, Yoo KH, Chung BY, Kim DJ (2008) Synthesis and evaluation of stilbenylbenzoxazole and stilbenylbenzothiazole derivatives for detecting beta-amyloid fibrils. Bioorg Med Chem Lett 18, 1534-1537. Velkova A, Tatarek-Nossol M, Andreetto E, Kapurniotu A (2008) Exploiting cross-amyloid interactions to inhibit protein aggregation but not function: Nanomolar affinity inhibition of insulin aggregation by an IAPP mimic. Angew Chem Int Ed Engl 47, 7114-7118. 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, Lindquist S, Shorter J (2008) Direct and selective elimination of specific prions and amyloids by 4,5-dianilinophthalimide and analogs. Proc Natl Acad Sci U S A 105, 7159-7164. Wisniewski T, Sadowski M (2008) Preventing betaamyloid fibrillization and deposition: Beta-sheet breakers and pathological chaperone inhibitors. BMC Neurosci 9(Suppl 2), S5. 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 92 [1107] [1108] [1109] [1110] [1111] [1112] [1113] [1114] [1115] [1116] [1117] [1118] [1119] 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. Campiglia P, Scrima M, Grimaldi M, Cioffi G, Bertamino A, Sala M, Aquino C, Gomez-Monterrey I, Grieco P, Novellino E, D’Ursi AM (2009) A new series of 1,3-dihidro-imidazo[1,5-c]thiazole-5,7-dione derivatives: Synthesis and interaction with Abeta(25-35) amyloid peptide. Chem Biol Drug Des 74, 224-233. Csuk R, Barthel A, Raschke C, Kluge R, Strohl D, Trieschmann L, Bohm G (2009) Synthesis of monomeric and dimeric acridine compounds as potential therapeutics in Alzheimer and prion diseases. Arch Pharm (Weinheim) 342, 699-709. Frydman-Marom A, Rechter M, Shefler I, Bram Y, Shalev DE, Gazit E (2009) Cognitive-performance recovery of Alzheimer’s disease model mice by modulation of early soluble amyloidal assemblies. Angew Chem Int Ed Engl 48, 1981-1986. Giordano C, Masi A, Pizzini A, Sansone A, Consalvi V, Chiaraluce R, Lucente G (2009) Synthesis and activity of fibrillogenesis peptide inhibitors related to the 17-21 beta-amyloid sequence. Eur J Med Chem 44, 179-189. Grillo-Bosch D, Carulla N, Cruz M, Sanchez L, PujolPina R, Madurga S, Rabanal F, Giralt E (2009) Retro-enantio N-methylated peptides as beta-amyloid aggregation inhibitors. ChemMedChem 4, 1488-1494. Madine J, Wang X, Brown DR, Middleton DA (2009) Evaluation of beta-alanine- and GABA-substituted peptides as inhibitors of disease-linked protein aggregation. Chembiochem 10, 1982-1987. Dolphin GT, Renaudet O, Ouberai M, Dumy P, Garcia J, Reymond JL (2009) Phenolic oxime oligomers inhibit Alzheimer’s amyloid fibril formation and disaggregate fibrils in vitro. Chembiochem 10, 1325-1329. Catto M, Aliano R, Carotti A, Cellamare S, Palluotto F, Purgatorio R, De SA, Campagna F (2010) Design, synthesis and biological evaluation of indane-2-arylhydrazinylmethylene-1,3-diones and indol-2-aryldiazenylmethylene-3-ones as beta-amyloid aggregation inhibitors. Eur J Med Chem 45, 1359-1366. Matharu B, El-Agnaf O, Razvi A, Austen BM (2010) Development of retro-inverso peptides as anti-aggregation drugs for beta-amyloid in Alzheimer’s disease. Peptides 31, 1866-1872. Suzuki M, Takahashi T, Sato J, Mie M, Kobatake E, Mihara H (2010) Designed short peptides that form amyloid-like fibrils in coassembly with amyloid beta-peptide (Abeta) decrease the toxicity of Abeta to neuronal PC12 cells. Chembiochem 11, 1525-1530. Zhou Y, Jiang C, Zhang Y, Liang Z, Liu W, Wang L, Luo C, Zhong T, Sun Y, Zhao L, Xie X, Jiang H, Zhou N, Liu D, Liu H (2010) Structural optimization and biological evaluation of substituted bisphenol A derivatives as betaamyloid peptide aggregation inhibitors. J Med Chem 53, 5449-5466. Ladiwala AR, Dordick JS, Tessier PM (2011) Aromatic small molecules remodel toxic soluble oligomers of amyloid beta through three independent pathways. J Biol Chem 286, 3209-3218. Ortega A, Rincon A, Jimenez-Aliaga KL, Bermejo-Bescos P, Martin-Aragon S, Molina MT, Csaky AG (2011) Synthesis and evaluation of arylquinones as BACE1 inhibitors, beta-amyloid peptide aggregation inhibitors, and destabilizers of preformed beta-amyloid fibrils. Bioorg Med Chem Lett 21, 2183-2187. [1120] [1121] [1122] [1123] [1124] [1125] [1126] [1127] [1128] [1129] [1130] [1131] [1132] [1133] [1134] [1135] Ryan TM, Griffin MD, Teoh CL, Ooi J, Howlett GJ (2011) High-affinity amphipathic modulators of amyloid fibril nucleation and elongation. J Mol Biol 406, 416-429. Ryan TM, Griffin MD, Bailey MF, Schuck P, Howlett GJ (2011) NBD-labeled phospholipid accelerates apolipoprotein C-II amyloid fibril formation but is not incorporated into mature fibrils. Biochemistry 50, 9579-9586. Frydman-Marom A, Shaltiel-Karyo R, Moshe S, Gazit E (2011) The generic amyloid formation inhibition effect of a designed small aromatic beta-breaking peptide. Amyloid 18, 119-127. Lukiw WJ (2012) Amyloid beta (Abeta) peptide modulators and other current treatment strategies for Alzheimer’s disease (AD). Expert Opin Emerg Drugs 17, 43-60. Di Santo R, Costi R, Cuzzucoli CG, Pescatori L, Rosi F, Scipione L, Celona D, Vertechy M, Ghirardi O, Piovesan P, Marzi M, Caccia S, Guiso G, Giorgi F, Minetti P (2012) Design, synthesis and structure-activity relationship of NAryl-naphthylamine derivatives as amyloid aggregation inhibitors. J Med Chem 55, 8538-8548. Cheng PN, Liu C, Zhao M, Eisenberg D, Nowick JS (2012) Amyloid beta-sheet mimics that antagonize protein aggregation and reduce amyloid toxicity. Nat Chem 4, 927-933. Huang L, Lu C, Sun Y, Mao F, Luo Z, Su T, Jiang H, Shan W, Li X (2012) Multi-target-directed benzylidene-indanone derivatives: Anti-beta amyloid (Abeta) aggregation, antioxidant, metal chelation and monoamine oxidase B (MAO-B) inhibition properties against Alzheimer’s disease. J Med Chem 55, 8483-8492. Liu R, Su R, Liang M, Huang R, Wang M, Qi W, He Z (2012) Physicochemical strategies for inhibition of amyloid fibril formation: An overview of recent advances. Curr Med Chem 19, 4157-4174. Ryan TM, Friedhuber A, Lind M, Howlett GJ, Masters C, Roberts BR (2012) Small amphipathic molecules modulate secondary structure and amyloid fibril-forming kinetics of Alzheimer disease peptide Abeta(1-42). J Biol Chem 287, 16947-16954. Lopez LC, Dos-Reis S, Espargaro A, Carrodeguas JA, Maddelein ML, Ventura S, Sancho J (2012) Discovery of novel inhibitors of amyloid beta-peptide 1-42 aggregation. J Med Chem 55, 9521-9530. Guo JT, Xu Y (2006) Amyloid fibril structure modeling using protein threading and molecular dynamics simulations. Methods Enzymol 412, 300-314. Guo JT, Xu Y (2008) Towards modeling of amyloid fibril structures. Front Biosci 13, 4039-4050. Esteras-Chopo A, Morra G, Moroni E, Serrano L, Lopez de la PM, Colombo G (2008) A molecular dynamics study of the interaction of D-peptide amyloid inhibitors with their target sequence reveals a potential inhibitory pharmacophore conformation. J Mol Biol 383, 266-280. Wu C, Wang Z, Lei H, Duan Y, Bowers MT, Shea JE (2008) The binding of thioflavin T and its neutral analog BTA1 to protofibrils of the Alzheimer’s disease Abeta(16-22) peptide probed by molecular dynamics simulations. J Mol Biol 384, 718-729. Wu C, Murray MM, Bernstein SL, Condron MM, Bitan G, Shea JE, Bowers MT (2009) The structure of Abeta42 C-terminal fragments probed by a combined experimental and theoretical study. J Mol Biol 387, 492-501. Chebaro Y, Mousseau N, Derreumaux P (2009) Structures and thermodynamics of Alzheimer’s amyloid-beta Abeta(16-35) monomer and dimer by replica exchange molecular dynamics simulations: Implication for full- W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1136] [1137] [1138] [1139] [1140] [1141] [1142] [1143] [1144] [1145] [1146] [1147] [1148] [1149] [1150] length Abeta fibrillation. J Phys Chem B 113, 76687675. Chebaro Y, Derreumaux P (2009) Targeting the early steps of Abeta16-22 protofibril disassembly by N-methylated inhibitors: A numerical study. Proteins 75, 442452. Convertino M, Pellarin R, Catto M, Carotti A, Caflisch A (2009) 9,10-Anthraquinone hinders beta-aggregation: How does a small molecule interfere with Abeta-peptide amyloid fibrillation? Protein Sci 18, 792-800. Frydman-Marom A, Convertino M, Pellarin R, Lampel A, Shaltiel-Karyo R, Segal D, Caflisch A, Shalev DE, Gazit E (2011) Structural basis for inhibiting beta-amyloid oligomerization by a non-coded beta-breaker-substituted endomorphin analogue. ACS Chem Biol 6, 1265-1276. Stempler S, Levy-Sakin M, Frydman-Marom A, Amir Y, Scherzer-Attali R, Buzhansky L, Gazit E, Senderowitz H (2011) Quantitative structure-activity relationship analysis of beta-amyloid aggregation inhibitors. J Comput Aided Mol Des 25, 135-144. Wise-Scira O, Xu L, Kitahara T, Perry G, Coskuner O (2011) Amyloid-beta peptide structure in aqueous solution varies with fragment size. J Chem Phys 135, 205101. Convertino M, Vitalis A, Caflisch A (2011) Disordered binding of small molecules to Abeta(12-28). J Biol Chem 286, 41578-41588. Chebaro Y, Jiang P, Zang T, Mu Y, Nguyen PH, Mousseau N, Derreumaux P (2012) Structures of abeta17-42 trimers in isolation and with five small-molecule drugs using a hierarchical computational procedure. J Phys Chem B 116, 8412-8422. Novick PA, Lopes DH, Branson KM, Esteras-Chopo A, Graef IA, Bitan G, Pande VS (2012) Design of betaamyloid aggregation inhibitors from a predicted structural motif. J Med Chem 55, 3002-3010. Srivastava A, Balaji PV (2012) Size, orientation and organization of oligomers that nucleate amyloid fibrils: Clues from MD simulations of pre-formed aggregates. Biochim Biophys Acta 1824, 963-973. Greenberg SM, Rosand J, Schneider AT, Creed PL, Gandy SE, Rovner B, Fitzsimmons BF, Smith EE, Edip GM, Schwab K, Laurin J, Garceau D (2006) A phase 2 study of tramiprosate for cerebral amyloid angiopathy. Alzheimer Dis Assoc Disord 20, 269-274. Wright TM (2006) Tramiprosate. Drugs Today (Barc) 42, 291-298. Gervais F, Paquette J, Morissette C, Krzywkowski P, Yu M, Azzi M, Lacombe D, Kong X, Aman A, Laurin J, Szarek WA, Tremblay P (2007) Targeting soluble Abeta peptide with Tramiprosate for the treatment of brain amyloidosis. Neurobiol Aging 28, 537-547. Gauthier S, Aisen PS, Ferris SH, Saumier D, Duong A, Haine D, Garceau D, Suhy J, Oh J, Lau W, Sampalis J (2009) Effect of tramiprosate in patients with mild-tomoderate Alzheimer’s disease: Exploratory analyses of the MRI sub-group of the Alphase study. J Nutr Health Aging 13, 550-557. 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 volumetric MRI in therapeutic trials in Alzheimer’s disease: The ALZHEMED (Tramiprosate) experience. J Nutr Health Aging 13, 370-372. Saumier D, Duong A, Haine D, Garceau D, Sampalis J (2009) Domain-specific cognitive effects of tramiprosate [1151] [1152] [1153] [1154] [1155] [1156] [1157] [1158] [1159] [1160] [1161] [1162] [1163] [1164] 93 in patients with mild to moderate Alzheimer’s disease: ADAS-cog subscale results from the Alphase Study. J Nutr Health Aging 13, 808-812. Aisen PS, Gauthier S, Ferris SH, Saumier D, Haine D, Garceau D, Duong A, Suhy J, Oh J, Lau WC, Sampalis J (2011) Tramiprosate in mild-to-moderate Alzheimer’s disease - a randomized, double-blind, placebo-controlled, multi-centre study (the Alphase Study). Arch Med Sci 7, 102-111. Swanoski MT (2009) Homotaurine: A failed drug for Alzheimer’s disease and now a nutraceutical for memory protection. Am J Health Syst Pharm 66, 19501953. 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 drug candidates for the treatment of Alzheimer’s disease. J Biol Chem 278, 13905-13911. Uryu S, Tokuhiro S, Murasugi T, Oda T (2002) A novel compound, RS-1178, specifically inhibits neuronal cell death mediated by beta-amyloid-induced macrophage activation in vitro. Brain Res 946, 298-306. Nakagami Y, Nishimura S, Murasugi T, Kaneko I, Meguro M, Marumoto S, Kogen H, Koyama K, Oda T (2002) A novel beta-sheet breaker, RS-0406, reverses amyloid beta-induced cytotoxicity and impairment of long-term potentiation in vitro. Br J Pharmacol 137, 676-682. Nakagami Y, Nishimura S, Murasugi T, Kubo T, Kaneko I, Meguro M, Marumoto S, Kogen H, Koyama K, Oda T (2002) A novel compound RS-0466 reverses betaamyloid-induced cytotoxicity through the Akt signaling pathway in vitro. Eur J Pharmacol 457, 11-17. Walsh DM, Townsend M, Podlisny MB, Shankar GM, Fadeeva JV, El AO, Hartley DM, Selkoe DJ (2005) Certain inhibitors of synthetic amyloid beta-peptide (Abeta) fibrillogenesis block oligomerization of natural Abeta and thereby rescue long-term potentiation. J Neurosci 25, 2455-2462. O’Hare E, Scopes DI, Treherne JM, Norwood K, Spanswick D, Kim EM (2010) RS-0406 arrests amyloid-beta oligomer-induced behavioural deterioration in vivo. Behav Brain Res 210, 32-37. Rodrigue KM, Kennedy KM, Park DC (2009) Betaamyloid deposition and the aging brain. Neuropsychol Rev 19, 436-450. Rodrigue KM, Kennedy KM, Devous MD, Rieck JR, Hebrank AC, Diaz-Arrastia R, Mathews D, Park DC (2012) beta-Amyloid burden in healthy aging: Regional distribution and cognitive consequences. Neurology 78, 387-395. Cohen RM (2007) The application of positron-emitting molecular imaging tracers in Alzheimer’s disease. Mol Imaging Biol 9, 204-216. Mathis CA, Wang Y, Holt DP, Huang GF, Debnath ML, Klunk WE (2003) Synthesis and evaluation of 11C-labeled 6-substituted 2-arylbenzothiazoles as amyloid imaging agents. J Med Chem 46, 2740-2754. Blennow K, de Leon MJ, Zetterberg H (2006) Alzheimer’s disease. Lancet 368, 387-403. Fagan AM, Mintun MA, Mach RH, Lee SY, Dence CS, 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 and cerebrospinal fluid Abeta42 in humans. Ann Neurol 59, 512-519. 94 [1165] [1166] [1167] [1168] [1169] [1170] [1171] [1172] [1173] [1174] [1175] [1176] [1177] [1178] [1179] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Quigley H, Colloby SJ, O’Brien JT (2011) PET imaging of brain amyloid in dementia: A review. Int J Geriatr Psychiatry 26, 991-999. Tarawneh R, Holtzman DM (2010) Biomarkers in translational research of Alzheimer’s disease. Neuropharmacology 59, 310-322. Forsberg A, Almkvist O, Engler H, Wall A, Langstrom B, Nordberg A (2010) High PIB retention in Alzheimer’s disease is an early event with complex relationship with CSF biomarkers and functional parameters. Curr Alzheimer Res 7, 56-66. Grimmer T, Henriksen G, Wester HJ, Forstl H, Klunk WE, Mathis CA, Kurz A, Drzezga A (2009) Clinical severity of Alzheimer’s disease is associated with PIB uptake in PET. Neurobiol Aging 30, 1902-1909. 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 human immunodeficiency virus-associated neurocognitive disorder. Arch Neurol 69, 72-77. Kung HF (2012) The beta-amyloid hypotheis in Alzheimer’s disease: Seeing is believing. ACS Med Chem Lett 3, 265-267. Ossenkoppele R, van Berckel BN, Prins ND (2011) Amyloid imaging in prodromal Alzheimer’s disease. Alzheimers Res Ther 3, 26. Ossenkoppele R, Tolboom N, Foster-Dingley JC, Adriaanse SF, Boellaard R, Yaqub M, Windhorst AD, Barkhof F, Lammertsma AA, Scheltens P, van der Flier WM, van Berckel BN (2012) Longitudinal imaging of Alzheimer pathology using [(11)C]PIB, [(18)F]FDDNP and [(18)F]FDG PET. Eur J Nucl Med Mol Imaging 39, 990-1000. Ossenkoppele R, Zwan MD, Tolboom N, van Assema DM, Adriaanse SF, Kloet RW, Boellaard R, Windhorst AD, Barkhof F, Lammertsma AA, Scheltens P, van der Flier WM, van Berckel BN (2012) Amyloid burden and metabolic function in early-onset Alzheimer’s disease: Parietal lobe involvement. Brain 135, 2115-2125. Small GW, Siddarth P, Kepe V, Ercoli LM, Burggren AC, Bookheimer SY, Miller KJ, Kim J, Lavretsky H, Huang SC, Barrio JR (2012) Prediction of cognitive decline by positron emission tomography of brain amyloid and tau. Arch Neurol 69, 215-222. Herholz K (2012) Imaging cerebral amyloid plaques: Clinical perspective. Lancet Neurol 11, 652-653. Driscoll I, Troncoso JC, Rudow G, Sojkova J, Pletnikova O, 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 amyloid imaging and postmortem, region-matched assessment of plaques. Acta Neuropathol, doi: 10.1007/s00401-0121025-1 Serdons K, Verduyckt T, Vanderghinste D, Borghgraef P, Cleynhens J, Van Leuven F, Kung H, Bormans G, Verbruggen A (2009) 11C-labelled PIB analogues as potential tracer agents for in vivo imaging of amyloid beta in Alzheimer’s disease. Eur J Med Chem 44, 1415-1426. Serdons K, Verbruggen A, Bormans GM (2009) Developing new molecular imaging probes for PET. Methods 48, 104-111. Choi SR, Golding G, Zhuang Z, Zhang W, Lim N, Hefti F, Benedum TE, Kilbourn MR, Skovronsky D, Kung HF (2009) Preclinical properties of 18F-AV-45: A PET agent for Abeta plaques in the brain. J Nucl Med 50, 1887-1894. [1180] [1181] [1182] [1183] [1184] [1185] [1186] [1187] [1188] [1189] [1190] [1191] [1192] Choi SR, Schneider JA, Bennett DA, Beach TG, Bedell BJ, Zehntner SP, Krautkramer MJ, Kung HF, Skovronsky DM, Hefti F, Clark CM (2012) Correlation of amyloid PET ligand florbetapir F 18 binding with abeta aggregation and neuritic plaque deposition in postmortem brain tissue. Alzheimer Dis Assoc Disord 26, 8-16. Kung HF, Choi SR, Qu W, Zhang W, Skovronsky D (2010) 18F stilbenes and styrylpyridines for PET imaging of A beta plaques in Alzheimer’s disease: A miniperspective. J Med Chem 53, 933-941. Lin KJ, Hsu WC, Hsiao IT, Wey SP, Jin LW, Skovronsky D, Wai YY, Chang HP, Lo CW, Yao CH, Yen TC, Kung MP (2010) Whole-body biodistribution and brain PET imaging with [18F]AV-45, a novel amyloid imaging agent–a pilot study. Nucl Med Biol 37, 497-508. Liu Y, Zhu L, Plossl K, Choi SR, Qiao H, Sun X, Li S, Zha Z, Kung HF (2010) Optimization of automated radiosynthesis of [18F]AV-45: A new PET imaging agent for Alzheimer’s disease. Nucl Med Biol 37, 917-925. Okamura N, Yanai K (2010) Florbetapir (18F), a PET imaging agent that binds to amyloid plaques for the potential detection of Alzheimer’s disease. IDrugs 13, 890-899. Wong DF, Rosenberg PB, Zhou Y, Kumar A, Raymont V, Ravert HT, Dannals RF, Nandi A, Brasic JR, Ye W, Hilton J, Lyketsos C, Kung HF, Joshi AD, Skovronsky DM, Pontecorvo MJ (2010) In vivo imaging of amyloid deposition in Alzheimer disease using the radioligand 18FAV-45 (florbetapir [corrected] F 18). J Nucl Med 51, 913920. Yao CH, Lin KJ, Weng CC, Hsiao IT, Ting YS, Yen TC, 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 in human brain. Appl Radiat Isot 68, 2293-2297. Lister-James J, Pontecorvo MJ, Clark C, Joshi AD, Mintun MA, Zhang W, Lim N, Zhuang Z, Golding G, Choi SR, Benedum TE, Kennedy P, Hefti F, Carpenter AP, Kung HF, Skovronsky DM (2011) Florbetapir f-18: A histopathologically validated Beta-amyloid positron emission tomography imaging agent. Semin Nucl Med 41, 300-304. Carome M, Wolfe S (2011) Florbetapir-PET imaging and postmortem beta-amyloid pathology. JAMA 305, 18571858. Clark CM, Schneider JA, Bedell BJ, Beach TG, Bilker WB, Mintun MA, Pontecorvo MJ, Hefti F, Carpenter AP, Flitter ML, Krautkramer MJ, Kung HF, Coleman RE, Doraiswamy PM, Fleisher AS, Sabbagh MN, Sadowsky CH, Reiman EP, Zehntner SP, Skovronsky DM (2011) Use of florbetapir-PET for imaging beta-amyloid pathology. JAMA 305, 275-283. Fleisher AS, Chen K, Liu X, Roontiva A, Thiyyagura P, Ayutyanont N, Joshi AD, Clark CM, Mintun MA, Pontecorvo MJ, Doraiswamy PM, Johnson KA, Skovronsky DM, Reiman EM (2011) Using positron emission tomography and florbetapir F18 to image cortical amyloid in patients with mild cognitive impairment or dementia due to Alzheimer disease. Arch Neurol 68, 1404-1411. Frisoni GB (2011) PET and 18F ligands in the diagnosis of Alzheimer’s disease. Lancet Neurol 10, 397-399. Joshi AD, Pontecorvo MJ, Clark CM, Carpenter AP, Jennings DL, Sadowsky CH, Adler LP, Kovnat KD, Seibyl JP, Arora A, Saha K, Burns JD, Lowrey MJ, Mintun MA, Skovronsky DM (2012) Performance characteristics of amyloid PET with florbetapir F 18 in patients with W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1193] [1194] [1195] [1196] [1197] [1198] [1199] [1200] [1201] [1202] [1203] Alzheimer’s disease and cognitively normal subjects. J Nucl Med 53, 378-384. Saint-Aubert L, Planton M, Hannequin D, Albucher JF, Delisle MB, Payoux P, Hitzel A, Viallard G, Peran P, Campion 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 duplication carrier. J Alzheimers Dis 28, 877-883. 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 tomography and neuropathologic estimates of fibrillar amyloid-beta in a patient with Down syndrome and Alzheimer disease. Arch Neurol 68, 1461-1466. Huang KL, Lin KJ, Ho MY, Chang YJ, Chang CH, Wey SP, Hsieh CJ, Yen TC, Hsiao IT, Lee TH (2012) Amyloid deposition after cerebral hypoperfusion: Evidenced on [(18)F]AV-45 positron emission tomography. J Neurol Sci 319, 124-129. Hsiao IT, Huang CC, Hsieh CJ, Hsu WC, Wey SP, Yen TC, Kung MP, Lin KJ (2012) Correlation of early-phase 18Fflorbetapir (AV-45/Amyvid) PET images to FDG images: Preliminary studies. Eur J Nucl Med Mol Imaging 39, 613620. Camus V, Payoux P, Barre L, Desgranges B, Voisin T, Tauber C, La JR, Tafani M, Hommet C, Chetelat G, Mondon K, de LS, V, Cottier JP, Beaufils E, Ribeiro MJ, Gissot V, Vierron E, Vercouillie J, Vellas B, Eustache F, Guilloteau 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. Newberg AB, Arnold SE, Wintering N, Rovner BW, Alavi A (2012) Initial clinical comparison of 18F-Florbetapir and 18F-FDG PET in patients with Alzheimer disease and controls. J Nucl Med 53, 902-907. Clark CM, Pontecorvo MJ, Beach TG, Bedell BJ, Coleman RE, Doraiswamy PM, Fleisher AS, Reiman EM, Sabbagh MN, Sadowsky CH, Schneider JA, Arora A, Carpenter AP, Flitter ML, Joshi AD, Krautkramer MJ, Lu M, Mintun MA, Skovronsky DM (2012) Cerebral PET with florbetapir compared with neuropathology at autopsy for detection of neuritic amyloid-beta plaques: A prospective cohort study. Lancet Neurol 11, 669-678. Kingwell K (2012) Alzheimer disease: Florbetapir-a useful tool to image amyloid load and predict cognitive decline in Alzheimer disease. Nat Rev Neurol 8, 471. 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, Kung MP, Skovronsky D, Dyrks T, Holl G, Krause S, Friebe M, Lehman L, Lindemann S, Dinkelborg LM, Masters CL, Villemagne VL (2008) Imaging of amyloid beta in Alzheimer’s disease with 18F-BAY94-9172, a novel PET tracer: Proof of mechanism. Lancet Neurol 7, 129-135. 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 PET with florbetaben (18F) in patients with Alzheimer’s disease and healthy controls: A multicentre phase 2 diagnostic study. Lancet Neurol 10, 424-435. Barthel H, Luthardt J, Becker G, Patt M, Hammerstein E, Hartwig K, Eggers B, Sattler B, Schildan A, Hesse S, Meyer PM, Wolf H, Zimmermann T, Reischl J, Rohde B, Gertz HJ, Reininger C, Sabri O (2011) Individualized [1204] [1205] [1206] [1207] [1208] [1209] [1210] [1211] [1212] [1213] [1214] [1215] [1216] 95 quantification of brain beta-amyloid burden: Results of a proof of mechanism phase 0 florbetaben PET trial in patients with Alzheimer’s disease and healthy controls. Eur J Nucl Med Mol Imaging 38, 1702-1714. Barthel H, Sabri O (2011) Florbetaben to trace amyloidbeta in the Alzheimer brain by means of PET. J Alzheimers Dis 26(Suppl 3), 117-121. Herholz K, Ebmeier K (2011) Clinical amyloid imaging in Alzheimer’s disease. Lancet Neurol 10, 667-670. Vallabhajosula S (2011) Positron emission tomography radiopharmaceuticals for imaging brain Beta-amyloid. Semin Nucl Med 41, 283-299. Villemagne VL, Ong K, Mulligan RS, Holl G, Pejoska S, Jones G, O’Keefe G, Ackerman U, Tochon-Danguy H, 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 dementias. J Nucl Med 52, 1210-1217. Villemagne VL, Okamura N, Pejoska S, Drago J, Mulligan RS, Chetelat G, O’Keefe G, Jones G, Kung HF, Pontecorvo M, Masters CL, Skovronsky DM, Rowe CC (2012) Differential diagnosis in Alzheimer’s disease and dementia with Lewy bodies via VMAT2 and amyloid imaging. Neurodegener Dis 10, 161-165. Wang H, Shi H, Yu H, Jiang S, Tang G (2011) Facile and rapid one-step radiosynthesis of [(18)F]BAY94-9172 with a new precursor. Nucl Med Biol 38, 121-127. O’Keefe GJ, Saunder TH, Ng S, Ackerman U, TochonDanguy 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 and 18F-BAY94-9172. J Nucl Med 50, 309-315. Villemagne VL, Mulligan RS, Pejoska S, Ong K, Jones G, O’Keefe G, Chan JG, Young K, Tochon-Danguy H, Masters CL, Rowe CC (2012) Comparison of (11)C-PiB and (18)F-florbetaben for Abeta imaging in ageing and Alzheimer’s disease. Eur J Nucl Med Mol Imaging 39, 983-989. Schipke CG, Peters O, Heuser I, Grimmer T, Sabbagh MN, Sabri O, Hock C, Kunz M, Kuhlmann J, Reininger C, Blankenburg M (2012) Impact of beta-amyloid-specific florbetaben PET imaging on confidence in early diagnosis of Alzheimer’s disease. Dement Geriatr Cogn Disord 33, 416-422. Nelissen N, Van LK, Thurfjell L, Owenius R, Vandenbulcke M, Koole M, Bormans G, Brooks DJ, Vandenberghe R (2009) Phase 1 study of the Pittsburgh compound B derivative 18F-flutemetamol in healthy volunteers and patients with probable Alzheimer disease. J Nucl Med 50, 12511259. Vandenberghe R, Van LK, Ivanoiu A, Salmon E, Bastin C, Triau E, Hasselbalch S, Law I, Andersen A, Korner A, Minthon L, Garraux G, Nelissen N, Bormans G, Buckley C, Owenius R, Thurfjell L, Farrar G, Brooks DJ (2010) 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: A phase 2 trial. Ann Neurol 68, 319-329. Thomas BA, Erlandsson K, Modat M, Thurfjell L, Vandenberghe R, Ourselin S, Hutton BF (2011) The importance of appropriate partial volume correction for PET quantification in Alzheimer’s disease. Eur J Nucl Med Mol Imaging 38, 1104-1119. Wolk DA, Grachev ID, Buckley C, Kazi H, Grady MS, Trojanowski JQ, Hamilton RH, Sherwin P, McLain R, Arnold SE (2011) Association between in vivo 96 [1217] [1218] [1219] [1220] [1221] [1222] [1223] [1224] [1225] [1226] [1227] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes fluorine 18-labeled flutemetamol amyloid positron emission tomography imaging and in vivo cerebral cortical histopathology. Arch Neurol 68, 1398-1403. Thurfjell L, Lotjonen J, Lundqvist R, Koikkalainen J, Soininen H, Waldemar G, Brooks DJ, Vandenberghe R (2012) Combination of biomarkers: PET [18F]flutemetamol imaging and structural MRI in dementia and mild cognitive impairment. Neurodegener Dis 10, 246-249. Vandenberghe R, Nelissen N, Salmon E, Ivanoiu A, Hasselbalch S, Andersen A, Korner A, Minthon L, Brooks DJ, Van LK, Dupont P (2012) Binary classification of (18)Fflutemetamol PET using machine learning: Comparison with visual reads and structural MRI. Neuroimage 64C, 517-525. Snellman A, Rokka J, Lopez-Picon FR, Eskola O, Wilson I, Farrar G, Scheinin M, Solin O, Rinne JO, HaaparantaSolin M (2012) Pharmacokinetics of [(18)F]flutemetamol in wild-type rodents and its binding to beta amyloid deposits in a mouse model of Alzheimer’s disease. Eur J Nucl Med Mol Imaging 39, 1784-1795. Jureus A, Swahn BM, Sandell J, Jeppsson F, Johnson AE, Johnstrom P, Neelissen JA, Sunnemark D, Farde L, Svensson SP (2010) Characterization of AZD4694, a novel fluorinated Abeta plaque neuroimaging PET radioligand. J Neurochem 114, 784-794. Brockschnieder D, Schmitt-Willich H, Heinrich T, Varrone A, Gulyas B, Toth M, Andersson J, Boemer U, Krause S, Friebe M, Dinkelborg L, Halldin C, Dyrks T (2012) Preclinical characterization of a novel class of 18F-Labeled PET tracers for amyloid-beta. J Nucl Med 53, 1794-1801. Furukawa K, Okamura N, Tashiro M, Waragai M, Furumoto S, Iwata R, Yanai K, Kudo Y, Arai H (2010) Amyloid PET in mild cognitive impairment and Alzheimer’s disease with BF-227: Comparison to FDG-PET. J Neurol 257, 721-727. 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 positron emission tomography probe [C]BF-227 uptake in mild cognitive impairment and alzheimer’s disease. Dement Geriatr Cogn Disord 30, 101-111. Kaneta T, Okamura N, Minoshima S, Furukawa K, Tashiro M, Furumoto S, Iwata R, Fukuda H, Takahashi S, Yanai K, Kudo Y, Arai H (2011) A modified method of 3D-SSP analysis for amyloid PET imaging using [(11)C]BF-227. Ann Nucl Med 25, 732-739. 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 by positron emission tomography and BF-227 in Alzheimer’s disease patients with or without diabetes mellitus. Geriatr Gerontol Int, doi: 10.1111/j.1447-0594.2012.00880.x [Epub ahead of print]. Fodero-Tavoletti MT, Mulligan RS, Okamura N, Furumoto S, Rowe CC, Kudo Y, Masters CL, Cappai R, Yanai K, Villemagne VL (2009) In vitro characterisation of BF227 binding to alpha-synuclein/Lewy bodies. Eur J Pharmacol 617, 54-58. Kikuchi A, Takeda A, Okamura N, Tashiro M, Hasegawa T, Furumoto S, Kobayashi M, Sugeno N, Baba T, Miki Y, Mori F, Wakabayashi K, Funaki Y, Iwata R, Takahashi S, Fukuda H, Arai H, Kudo Y, Yanai K, Itoyama Y (2010) In vivo visualization of alpha-synuclein deposition by carbon-11-labelled 2-[2-(2-dimethylaminothiazol-5- [1228] [1229] [1230] [1231] [1232] [1233] [1234] [1235] [1236] [1237] [1238] [1239] yl)ethenyl]-6-[2-(fluoro)ethoxy]benzoxazole positron emission tomography in multiple system atrophy. Brain 133, 1772-1778. Okamura N, Shiga Y, Furumoto S, Tashiro M, Tsuboi Y, Furukawa K, Yanai K, Iwata R, Arai H, Kudo Y, Itoyama Y, Doh-ura K (2010) In vivo detection of prion amyloid plaques using [(11)C]BF-227 PET. Eur J Nucl Med Mol Imaging 37, 934-941. Kung MP, Hou C, Zhuang ZP, Skovronsky D, Kung HF (2004) Binding of two potential imaging agents targeting amyloid plaques in postmortem brain tissues of patients with Alzheimer’s disease. Brain Res 1025, 98-105. Verhoeff NP, Wilson AA, Takeshita S, Trop L, Hussey D, Singh K, Kung HF, Kung MP, Houle S (2004) Invivo imaging of Alzheimer disease beta-amyloid with [11C]SB-13 PET. Am J Geriatr Psychiatry 12, 584-595. 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 for Abeta plaques in cerebral amyloid angiopathy (CAA). J Med Chem 54, 8085-8098. 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) Dicyanovinylnaphthalenes for neuroimaging of amyloids and relationships of electronic structures and geometries to binding affinities. Proc Natl Acad Sci U S A 109, 16492-16497. Cui M, Ono M, Kimura H, Ueda M, Nakamoto Y, Togashi K, Okamoto Y, Ihara M, Takahashi R, Liu B, Saji H (2012) Novel (18)F-labeled benzoxazole derivatives as potential positron emission tomography probes for imaging of cerebral beta-amyloid plaques in Alzheimer’s disease. J Med Chem 55, 9136-9145. Cui M, Wang X, Yu P, Zhang J, Li Z, Zhang X, Yang Y, Ono M, Jia H, Saji H, Liu B (2012) Synthesis and evaluation of novel (18)F labeled 2-pyridinylbenzoxazole and 2-pyridinylbenzothiazole derivatives as ligands for positron emission tomography (PET) imaging of betaamyloid plaques. J Med Chem 55, 9283-9296. Johnson AE, Jeppsson F, Sandell J, Wensbo D, Neelissen JA, Jureus A, Strom P, Norman H, Farde L, Svensson SP (2009) AZD2184: A radioligand for sensitive detection of beta-amyloid deposits. J Neurochem 108, 1177-1186. Nyberg S, Jonhagen ME, Cselenyi Z, Halldin C, Julin P, Olsson H, Freund-Levi Y, Andersson J, Varnas K, Svensson S, Farde L (2009) Detection of amyloid in Alzheimer’s disease with positron emission tomography using [11C]AZD2184. Eur J Nucl Med Mol Imaging 36, 1859-1863. Andersson JD, Varnas K, Cselenyi Z, Gulyas B, Wensbo D, Finnema SJ, Swahn BM, Svensson S, Nyberg S, Farde L, Halldin C (2010) Radiosynthesis of the candidate beta-amyloid radioligand [(11)C]AZD2184: Positron emission tomography examination and metabolite analysis in cynomolgus monkeys. Synapse 64, 733-741. Shoghi-Jadid K, Small GW, Agdeppa ED, Kepe V, Ercoli LM, Siddarth P, Read S, Satyamurthy N, Petric A, Huang SC, Barrio JR (2002) Localization of neurofibrillary tangles and beta-amyloid plaques in the brains of living patients with Alzheimer disease. Am J Geriatr Psychiatry 10, 24-35. Ossenkoppele R, Tolboom N, Foster-Dingley JC, Adriaanse SF, Boellaard R, Yaqub M, Windhorst AD, Barkhof F, Lammertsma AA, Scheltens P, van der Flier WM, van Berckel BN (2012) Longitudinal imaging of W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1240] [1241] [1242] [1243] [1244] [1245] [1246] [1247] [1248] [1249] [1250] [1251] Alzheimer pathology using [(11)C]PIB, [(18)F]FDDNP and [(18)F]FDG PET. Eur J Nucl Med Mol Imaging 39, 990-1000. Protas HD, Kepe V, Hayashi KM, Klunder AD, Braskie MN, Ercoli L, Siddarth P, Bookheimer SY, Thompson PM, Small GW, Barrio JR, Huang SC (2012) Prediction of cognitive decline based on hemispheric cortical surface maps of FDDNP PET. Neuroimage 61, 749-760. Cai L, Cuevas J, Temme S, Herman MM, Dagostin C, Widdowson DA, Innis RB, Pike VW (2007) Synthesis and structure-affinity relationships of new 4-(6-iodoH-imidazo[1,2-a]pyridin-2-yl)-N-dimethylbenzeneamine derivatives as ligands for human beta-amyloid plaques. J Med Chem 50, 4746-4758. Ryu EK, Chen X (2008) Development of Alzheimer’s disease imaging agents for clinical studies. Front Biosci 13, 777-789. Zhuang ZP, Kung MP, Wilson A, Lee CW, Plossl K, Hou C, Holtzman DM, Kung HF (2003) Structure-activity relationship of imidazo[1,2-a]pyridines as ligands for detecting beta-amyloid plaques in the brain. J Med Chem 46, 237-243. Newberg AB, Wintering NA, Plossl K, Hochold J, Stabin MG, Watson M, Skovronsky D, Clark CM, Kung MP, Kung HF (2006) Safety, biodistribution, and dosimetry of 123I-IMPY: A novel amyloid plaque-imaging agent for the diagnosis of Alzheimer’s disease. J Nucl Med 47, 748-754. Pepys MB, Herbert J, Hutchinson WL, Tennent GA, Lachmann 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, Murray S, Thompson D, Purvis A, Kolstoe S, Wood SP, Hawkins PN (2002) Targeted pharmacological depletion of serum amyloid P component for treatment of human amyloidosis. Nature 417, 254-259. Kolstoe SE, Wood SP (2010) Drug targets for amyloidosis. Biochem Soc Trans 38, 466-470. Millar DJ, Hutchinson WL, Pepys MB (2011) Immunoradiometric assay for human serum amyloid P component. J Immunol Methods 371, 18-24. Kolstoe SE, Ridha BH, Bellotti V, Wang N, Robinson CV, Crutch SJ, Keir G, Kukkastenvehmas R, Gallimore JR, Hutchinson WL, Hawkins PN, Wood SP, Rossor MN, Pepys MB (2009) Molecular dissection of Alzheimer’s disease neuropathology by depletion of serum amyloid P component. Proc Natl Acad Sci U S A 106, 7619-7623. Bodin K, Ellmerich S, Kahan MC, Tennent GA, Loesch A, Gilbertson JA, Hutchinson WL, Mangione PP, Gallimore JR, Millar DJ, Minogue S, Dhillon AP, Taylor GW, Bradwell AR, Petrie A, Gillmore JD, Bellotti V, Botto M, Hawkins PN, Pepys MB (2010) Antibodies to human serum amyloid P component eliminate visceral amyloid deposits. Nature 468, 93-97. Gillmore JD, Tennent GA, Hutchinson WL, Gallimore JR, Lachmann HJ, Goodman HJ, Offer M, Millar DJ, Petrie A, Hawkins PN, Pepys MB (2010) Sustained pharmacological depletion of serum amyloid P component in patients with systemic amyloidosis. Br J Haematol 148, 760-767. Schenk D, Barbour R, Dunn W, Gordon G, Grajeda H, Guido T, Hu K, Huang J, Johnson-Wood K, Khan K, Kholodenko D, Lee M, Liao Z, Lieberburg I, Motter R, Mutter L, Soriano F, Shopp G, Vasquez N, Vandevert C, Walker S, Wogulis M, Yednock T, Games D, Seubert P (1999) Immunization with amyloid-beta attenuates [1252] [1253] [1254] [1255] [1256] [1257] [1258] [1259] [1260] [1261] [1262] [1263] [1264] [1265] [1266] [1267] 97 Alzheimer-disease-like pathology in the PDAPP mouse. Nature 400, 173-177. Schenk D (2002) Amyloid-beta immunotherapy for Alzheimer’s disease: The end of the beginning. Nat Rev Neurosci 3, 824-828. Orgogozo JM, Gilman S, Dartigues JF, Laurent B, Puel M, Kirby LC, Jouanny P, Dubois B, Eisner L, Flitman S, Michel BF, Boada M, Frank A, Hock C (2003) Subacute meningoencephalitis in a subset of patients with AD after Abeta42 immunization. Neurology 61, 46-54. 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-amyloid by vaccination of patients with Alzheimer disease. Nat Med 8, 1270-1275. Hock C, Konietzko U, Streffer JR, Tracy J, Signorell A, Muller-Tillmanns B, Lemke U, Henke K, Moritz E, Garcia E, Wollmer MA, Umbricht D, de Quervain DJ, Hofmann M, Maddalena A, Papassotiropoulos A, Nitsch RM (2003) Antibodies against beta-amyloid slow cognitive decline in Alzheimer’s disease. Neuron 38, 547-554. Imbimbo BP (2002) Toxicity of beta-amyloid vaccination in patients with Alzheimer’s disease. Ann Neurol 51, 794. Imbimbo BP (2002) -Amyloid immunization approaches for Alzheimer’s disease. Drug Dev Res 56, 150-162. Nicoll JA, Wilkinson D, Holmes C, Steart P, Markham H, Weller RO (2003) Neuropathology of human Alzheimer disease after immunization with amyloid-beta peptide: A case report. Nat Med 9, 448-452. Nicoll JA, Barton E, Boche D, Neal JW, Ferrer I, Thompson P, Vlachouli C, Wilkinson D, Bayer A, Games D, Seubert P, Schenk D, Holmes C (2006) Abeta species removal after abeta42 immunization. J Neuropathol Exp Neurol 65, 1040-1048. Robinson SR, Bishop GM, Lee HG, Munch G (2004) Lessons from the AN 1792 Alzheimer vaccine: Lest we forget. Neurobiol Aging 25, 609-615. Bayer AJ, Bullock R, Jones RW, Wilkinson D, Paterson KR, Jenkins L, Millais SB, Donoghue S (2005) Evaluation of the safety and immunogenicity of synthetic Abeta42 (AN1792) in patients with AD. Neurology 64, 94-101. Boche D, Nicoll JA, Weller RO (2005) Immunotherapy for Alzheimer’s disease and other dementias. Curr Opin Neurol 18, 720-725. Boche D, Zotova E, Weller RO, Love S, Neal JW, Pickering RM, Wilkinson D, Holmes C, Nicoll JA (2008) Consequence of Abeta immunization on the vasculature of human Alzheimer’s disease brain. Brain 131, 3299-3310. Boche D, Donald J, Love S, Harris S, Neal JW, Holmes C, Nicoll JA (2010) Reduction of aggregated Tau in neuronal processes but not in the cell bodies after Abeta42 immunisation in Alzheimer’s disease. Acta Neuropathol 120, 13-20. Boche D, Denham N, Holmes C, Nicoll JA (2010) Neuropathology after active Abeta42 immunotherapy: Implications for Alzheimer’s disease pathogenesis. Acta Neuropathol 120, 369-384. Gilman S, Koller M, Black RS, Jenkins L, Griffith SG, Fox NC, 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. Hock C, Nitsch RM (2005) Clinical observations with AN1792 using TAPIR analyses. Neurodegener Dis 2, 273276. 98 [1268] [1269] [1270] [1271] [1272] [1273] [1274] [1275] [1276] [1277] [1278] [1279] [1280] [1281] [1282] [1283] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Lee M, Bard F, Johnson-Wood K, Lee C, Hu K, Griffith SG, Black RS, Schenk D, Seubert P (2005) Abeta42 immunization in Alzheimer’s disease generates Abeta N-terminal antibodies. Ann Neurol 58, 430-435. Masliah E, Hansen L, Adame A, Crews L, Bard F, Lee C, Seubert P, Games D, Kirby L, Schenk D (2005) Abeta vaccination effects on plaque pathology in the absence of encephalitis in Alzheimer disease. Neurology 64, 129-131. O’Toole M, Janszen DB, Slonim DK, Reddy PS, Ellis DK, Legault HM, Hill AA, Whitley MZ, Mounts WM, Zuberek K, Immermann FW, Black RS, Dorner AJ (2005) Risk factors associated with beta-amyloid(1-42) immunotherapy in preimmunization gene expression patterns of blood cells. Arch Neurol 62, 1531-1536. Schenk DB, Seubert P, Grundman M, Black R (2005) A beta immunotherapy: Lessons learned for potential treatment of Alzheimer’s disease. Neurodegener Dis 2, 255-260. Patton RL, Kalback WM, Esh CL, Kokjohn TA, Van Vickle GD, Luehrs DC, Kuo YM, Lopez J, Brune D, Ferrer I, Masliah E, Newel AJ, Beach TG, Castano EM, Roher AE (2006) Amyloid-beta peptide remnants in AN-1792immunized Alzheimer’s disease patients: A biochemical analysis. Am J Pathol 169, 1048-1063. Vasilevko V, Cribbs DH (2006) Novel approaches for immunotherapeutic intervention in Alzheimer’s disease. Neurochem Int 49, 113-126. Bombois S, Maurage CA, Gompel M, Deramecourt V, kowiak-Cordoliani MA, Black RS, Lavielle R, Delacourte A, Pasquier F (2007) Absence of beta-amyloid deposits after immunization in Alzheimer disease with Lewy body dementia. Arch Neurol 64, 583-587. Koepsell TD, Chi YY, Zhou XH, Lee WW, Ramos EM, Kukull WA (2007) An alternative method for estimating efficacy of the AN1792 vaccine for Alzheimer disease. Neurology 69, 1868-1872. Hawkes CA, McLaurin J (2008) Clinical immunotherapy trials in Alzheimer’s disease. Drug Discovery Today: Therapeutic Strategies 5, 177-183. Holmes C, Boche D, Wilkinson D, Yadegarfar G, Hopkins V, Bayer A, Jones RW, Bullock R, Love S, Neal JW, Zotova E, Nicoll JA (2008) Long-term effects of Abeta42 immunisation in Alzheimer’s disease: Follow-up of a randomised, placebo-controlled phase I trial. Lancet 372, 216-223. Holtzman DM (2008) Alzheimer’s disease: Moving towards a vaccine. Nature 454, 418-420. Pride M, Seubert P, Grundman M, Hagen M, Eldridge J, Black RS (2008) Progress in the active immunotherapeutic approach to Alzheimer’s disease: Clinical investigations into AN1792-associated meningoencephalitis. Neurodegener Dis 5, 194-196. St George-Hyslop PH, Morris JC (2008) Will anti-amyloid therapies work for Alzheimer’s disease? Lancet 372, 180182. Kokjohn TA, Roher AE (2009) Antibody responses, amyloid-beta peptide remnants and clinical effects of AN1792 immunization in patients with AD in an interrupted trial. CNS Neurol Disord Drug Targets 8, 88-97. Town T (2009) Alternative Abeta immunotherapy approaches for Alzheimer’s disease. CNS Neurol Disord Drug Targets 8, 114-127. Vellas B, Black R, Thal LJ, Fox NC, Daniels M, McLennan G, Tompkins C, Leibman C, Pomfret M, Grundman M (2009) Long-term follow-up of patients immunized with [1284] [1285] [1286] [1287] [1288] [1289] [1290] [1291] [1292] [1293] [1294] [1295] [1296] [1297] AN1792: Reduced functional decline in antibody responders. Curr Alzheimer Res 6, 144-151. Serrano-Pozo A, William CM, Ferrer I, Uro-Coste E, Delisle MB, Maurage CA, Hock C, Nitsch RM, Masliah E, Growdon JH, Frosch MP, Hyman BT (2010) Beneficial effect of human anti-amyloid-beta active immunization on neurite morphology and tau pathology. Brain 133, 13121327. Uro-Coste E, Russano de PG, Guilbeau-Frugier C, Sastre N, Ousset PJ, da Silva NA, Lavialle-Guillotreau V, Vellas B, Delisle MB (2010) Cerebral amyloid angiopathy and microhemorrhages after amyloid beta vaccination: Case report and brief review. Clin Neuropathol 29, 209-216. Zotova E, Holmes C, Johnston D, Neal JW, Nicoll JA, Boche D (2011) Microglial alterations in human Alzheimer’s disease following Abeta42 immunization. Neuropathol Appl Neurobiol 37, 513-524. Schneeberger A, Mandler M, Otawa O, Zauner W, Mattner F, Schmidt W (2009) Development of AFFITOPE vaccines for Alzheimer’s disease (AD)–from concept to clinical testing. J Nutr Health Aging 13, 264-267. Schneeberger A, Mandler M, Mattner F, Schmidt W (2010) AFFITOME(R) technology in neurodegenerative diseases: The doubling advantage. Hum Vaccin 6, 948-952. 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 of active Abeta immunotherapy with CAD106 in patients with Alzheimer’s disease: Randomised, double-blind, placebo-controlled, first-in-human study. Lancet Neurol 11, 597-604. Wisniewski T (2012) Active immunotherapy for Alzheimer’s disease. Lancet Neurol 11, 571-572. Kingwell K (2012) Alzheimer disease: Amyloid-beta immunotherapy CAD106 passes first safety test in patients with Alzheimer disease. Nat Rev Neurol 8, 414. Dyer MR, Renner WA, Bachmann MF (2006) A second vaccine revolution for the new epidemics of the 21st century. Drug Discov Today 11, 1028-1033. Jennings GT, Bachmann MF (2008) The coming of age of virus-like particle vaccines. Biol Chem 389, 521-536. Wiessner C, Wiederhold KH, Tissot AC, Frey P, Danner S, Jacobson LH, Jennings GT, Luond R, Ortmann R, Reichwald J, Zurini M, Mir A, Bachmann MF, Staufenbiel M (2011) The second-generation active Abeta immunotherapy CAD106 reduces amyloid accumulation in APP transgenic mice while minimizing potential side effects. J Neurosci 31, 9323-9331. Ryan JM, Grundman M (2009) Anti-amyloid-beta immunotherapy in Alzheimer’s disease: ACC-001 clinical trials are ongoing. J Alzheimers Dis 17, 243. Muhs A, Hickman DT, Pihlgren M, Chuard N, Giriens V, Meerschman C, Van der Auwera I, Van Leuven F, Sugawara M, Weingertner MC, Bechinger B, Greferath R, Kolonko N, Nagel-Steger L, Riesner D, Brady RO, Pfeifer A, Nicolau C (2007) Liposomal vaccines with conformation-specific amyloid peptide antigens define immune response and efficacy in APP transgenic mice. Proc Natl Acad Sci U S A 104, 9810-9815. 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) Sequenceindependent control of peptide conformation in liposomal W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1298] [1299] [1300] [1301] [1302] [1303] [1304] [1305] [1306] [1307] [1308] [1309] vaccines for targeting protein misfolding diseases. J Biol Chem 286, 13966-13976. Wang CY, Finstad CL, Walfield AM, Sia C, Sokoll KK, Chang TY, Fang XD, Hung CH, Hutter-Paier B, Windisch M (2007) Site-specific UBITh amyloid-beta vaccine for immunotherapy of Alzheimer’s disease. Vaccine 25, 30413052. Savage MJ, Wu G, McCampbell A, Wessner KR, Citron M, Liang X, Hsieh S, Wolfe AL, Kinney GG, Rosen LB, Renger JJ (2010) A novel multivariant Abetapeptide vaccine with preclinical evidence of a central immune response that generates antisera recognizing a wide range of Abeta peptide species. Alzheimers Dement 6, S142. Davtyan H, Ghochikyan A, Movsesyan N, Ellefsen B, Petrushina I, Cribbs DH, Hannaman D, Evans CF, Agadjanyan MG (2012) Delivery of a DNA vaccine for Alzheimer’s disease by electroporation versus gene gun generates potent and similar immune responses. Neurodegener Dis 10, 261-264. Xing XN, Zhang WG, Sha S, Li Y, Guo R, Wang C, Cao YP (2011) Amyloid beta 3-10 DNA vaccination suggests a potential new treatment for Alzheimer’s disease in BALB/c mice. Chin Med J (Engl) 124, 2636-2641. Xing X, Sha S, Li Y, Zong L, Jiang T, Cao Y (2012) Immunization with a new DNA vaccine for Alzheimer’s disease elicited Th2 immune response in BALB/c mice by in vivo electroporation. J Neurol Sci 313, 17-21. Ma Y, Li Y, Zong LX, Xing XN, Zhang WG, Cao YP (2011) Improving memory and decreasing cognitive impairment in Tg-APPswe/PSEN1dE9 mice with Abeta310 repeat fragment plasmid by reducing Abeta deposition and inflammatory response. Brain Res 1400, 112-124. Li Y, Ma Y, Zong LX, Xing XN, Sha S, Cao YP (2011) Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Abeta3-10 induces Th2 immune response against amyloid-beta in wild-type mouse. Neurosci Lett 505, 128-133. Li Y, Ma Y, Zong LX, Xing XN, Guo R, Jiang TZ, Sha S, Liu L, Cao YP (2012) Intranasal inoculation with an adenovirus vaccine encoding ten repeats of Abeta3-10 reduces AD-like pathology and cognitive impairment in TgAPPswe/PSEN1dE9 mice. J Neuroimmunol 249, 16-26. Santuccione AC, Merlini M, Shetty A, Tackenberg C, Bali J, Ferretti MT, McAfoose J, Kulic L, Bernreuther C, Welt T, Grimm J, Glatzel M, Rajendran L, Hock C, Nitsch RM (2012) Active vaccination with ankyrin G reduces beta-amyloid pathology in APP transgenic mice. Mol Psychiatry, doi: 10.1038/mp.2012.70 [Epub ahead of print]. Butovsky O, Koronyo-Hamaoui M, Kunis G, Ophir E, Landa G, Cohen H, Schwartz M (2006) Glatiramer acetate fights against Alzheimer’s disease by inducing dendriticlike microglia expressing insulin-like growth factor 1. Proc Natl Acad Sci U S A 103, 11784-11789. Hara H, Inoue M, Adachi K, Yonemitsu Y, Hasegawa M, Nabeshima T, Tabira T (2007) [Mucosal immunotherapy for alzheimer’s disease with viral vectors]. Nihon Shinkei Seishin Yakurigaku Zasshi 27, 53-56. 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, Fraser PE, St George-Hyslop P, Westaway D (2000) A beta peptide immunization reduces behavioural impairment and plaques in a model of Alzheimer’s disease. Nature 408, 979-982. [1310] [1311] [1312] [1313] [1314] [1315] [1316] [1317] [1318] [1319] [1320] [1321] [1322] 99 Morgan D, Diamond DM, Gottschall PE, Ugen KE, Dickey C, 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 loss in an animal model of Alzheimer’s disease. Nature 408, 982-985. Sigurdsson EM, Scholtzova H, Mehta PD, Frangione B, Wisniewski T (2001) Immunization with a nontoxic/nonfibrillar amyloid-beta homologous peptide reduces Alzheimer’s disease-associated pathology in transgenic mice. Am J Pathol 159, 439-447. Qu B, Rosenberg RN, Li L, Boyer PJ, Johnston SA (2004) Gene vaccination to bias the immune response to amyloidbeta peptide as therapy for Alzheimer disease. Arch Neurol 61, 1859-1864. Agadjanyan MG, Ghochikyan A, Petrushina I, Vasilevko V, Movsesyan N, Mkrtichyan M, Saing T, Cribbs DH (2005) Prototype Alzheimer’s disease vaccine using the immunodominant B cell epitope from beta-amyloid and promiscuous T cell epitope pan HLA DR-binding peptide. J Immunol 174, 1580-1586. Asuni AA, Boutajangout A, Scholtzova H, Knudsen E, Li YS, Quartermain D, Frangione B, Wisniewski T, Sigurdsson EM (2006) Vaccination of Alzheimer’s model mice with Abeta derivative in alum adjuvant reduces Abeta burden without microhemorrhages. Eur J Neurosci 24, 2530-2542. Lemere CA, Maier M, Jiang L, Peng Y, Seabrook TJ (2006) Amyloid-beta immunotherapy for the prevention and treatment of Alzheimer disease: Lessons from mice, monkeys, and humans. Rejuvenation Res 9, 77-84. Maier M, Seabrook TJ, Lazo ND, Jiang L, Das P, Janus C, Lemere CA (2006) Short amyloid-beta (Abeta) immunogens reduce cerebral Abeta load and learning deficits in an Alzheimer’s disease mouse model in the absence of an Abeta-specific cellular immune response. J Neurosci 26, 4717-4728. Seabrook TJ, Jiang L, Thomas K, Lemere CA (2006) Boosting with intranasal dendrimeric Abeta1-15 but not Abeta1-15 peptide leads to an effective immune response following a single injection of Abeta1-40/42 in APP-tg mice. J Neuroinflammation 3, 14. Seabrook TJ, Thomas K, Jiang L, Bloom J, Spooner E, Maier M, Bitan G, Lemere CA (2007) Dendrimeric Abeta1-15 is an effective immunogen in wildtype and APP-tg mice. Neurobiol Aging 28, 813-823. Lemere CA, Maier M, Peng Y, Jiang L, Seabrook TJ (2007) Novel Abeta immunogens: Is shorter better? Curr Alzheimer Res 4, 427-436. Mouri A, Noda Y, Hara H, Mizoguchi H, Tabira T, Nabeshima T (2007) Oral vaccination with a viral vector containing Abeta cDNA attenuates age-related Abeta accumulation and memory deficits without causing inflammation in a mouse Alzheimer model. FASEB J 21, 2135-2148. Nikolic WV, Bai Y, Obregon D, Hou H, Mori T, Zeng J, Ehrhart J, Shytle RD, Giunta B, Morgan D, Town T, Tan J (2007) Transcutaneous beta-amyloid immunization reduces cerebral beta-amyloid deposits without T cell infiltration and microhemorrhage. Proc Natl Acad Sci U S A 104, 2507-2512. Petrushina I, Ghochikyan A, Mktrichyan M, Mamikonyan G, Movsesyan N, Davtyan H, Patel A, Head E, Cribbs DH, Agadjanyan MG (2007) Alzheimer’s disease peptide epitope vaccine reduces insoluble but not soluble/oligomeric 100 [1323] [1324] [1325] [1326] [1327] [1328] [1329] [1330] [1331] [1332] [1333] [1334] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Abeta species in amyloid precursor protein transgenic mice. J Neurosci 27, 12721-12731. Petrushina I, Ghochikyan A, Mkrtichyan M, Mamikonyan G, Movsesyan N, Ajdari R, Vasilevko V, Karapetyan A, Lees A, Agadjanyan MG, Cribbs DH. (2008) MannanAbeta28 conjugate prevents Abeta-plaque deposition, but increases microhemorrhages in the brains of vaccinated Tg2576 (APPsw) mice. J Neuroinflammation 5, 42 Movsesyan N, Mkrtichyan M, Petrushina I, Ross TM, Cribbs DH, Agadjanyan MG, Ghochikyan A (2008) DNA epitope vaccine containing complement component C3d enhances anti-amyloid-beta antibody production and polarizes the immune response towards a Th2 phenotype. J Neuroimmunol 205, 57-63. Boutajangout A, Goni F, Knudsen E, Schreiber F, Asuni A, Quartermain D, Frangione B, Chabalgoity A, Wisniewski T, Sigurdsson EM (2009) Diminished amyloid-beta burden in Tg2576 mice following a prophylactic oral immunization with a salmonella-based amyloid-beta derivative vaccine. J Alzheimers Dis 18, 961-972. Kitazawa M, Vasilevko V, Cribbs DH, LaFerla FM (2009) Immunization with amyloid-beta attenuates inclusion body myositis-like myopathology and motor impairment in a transgenic mouse model. J Neurosci 29, 61326141. Trouche SG, Asuni A, Rouland S, Wisniewski T, Frangione B, Verdier JM, Sigurdsson EM, Mestre-Frances N (2009) Antibody response and plasma Abeta1-40 levels in young Microcebus murinus primates immunized with Abeta1-42 and its derivatives. Vaccine 27, 957-964. Wilcock DM, Gharkholonarehe N, Van Nostrand WE, Davis J, Vitek MP, Colton CA (2009) Amyloid reduction by amyloid-beta vaccination also reduces mouse tau pathology and protects from neuron loss in two mouse models of Alzheimer’s disease. J Neurosci 29, 79577965. 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 accumulation in Tg2576 transgenic mice by oral vaccination. Biochem Biophys Res Commun 399, 593-599. Vasilevko V, Pop V, Kim HJ, Saing T, Glabe CC, Milton S, Barrett EG, Cotman CW, Cribbs DH, Head E (2010) Linear and conformation specific antibodies in aged beagles after prolonged vaccination with aggregated Abeta. Neurobiol Dis 39, 301-310. Vollmar P, Kullmann JS, Thilo B, Claussen MC, Rothhammer V, Jacobi H, Sellner J, Nessler S, Korn T, Hemmer B (2010) Active immunization with amyloid-beta 1-42 impairs memory performance through TLR2/4-dependent activation of the innate immune system. J Immunol 185, 6338-6347. Wang CM, deVries S, Camboni M, Glass M, Martin PT (2010) Immunization with the SDPM1 peptide lowers amyloid plaque burden and improves cognitive function in the APPswePSEN1(A246E) transgenic mouse model of Alzheimer’s disease. Neurobiol Dis 39, 409-422. Mantile F, Basile C, Cicatiello V, De FD, Caivano A, De BP, Prisco A (2011) A multimeric immunogen for the induction of immune memory to beta-amyloid. Immunol Cell Biol 89, 604-609. Axelsen TV, Holm A, Christiansen G, Birkelund S (2011) Identification of the shortest Abeta-peptide generating highly specific antibodies against the C-terminal end of amyloid-beta42. Vaccine 29, 3260-3269. [1335] [1336] [1337] [1338] [1339] [1340] [1341] [1342] [1343] [1344] [1345] [1346] [1347] [1348] [1349] [1350] [1351] [1352] [1353] [1354] [1355] [1356] 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 nonhuman primates to beta-amyloid. J Neuroinflammation 9, 84. Esiri MM (2001) Is an effective immune intervention for Alzheimer’s disease in prospect? Trends Pharmacol Sci 22, 2-3. Ingram DK (2001) Vaccine development for Alzheimer’s disease: A shot of good news. Trends Neurosci 24, 305307. Sigurdsson EM, Wisniewski T, Frangione B (2002) A safer vaccine for Alzheimer’s disease? Neurobiol Aging 23, 1001-1008. Sigurdsson EM, Frangione B, Wisniewski T (2002) Immunization for Alzheimer’s disease. Drug Develop Res 56, 135-142. Solomon B (2002) Towards Alzheimer’s disease vaccination. Mini Rev Med Chem 2, 85-92. Wisniewski T, Sigurdsson EM (2002) Immunization treatment approaches in Alzheimer’s and prion diseases. Curr Neurol Neurosci Rep 2, 400-404. Dodart JC, Bales KR, Paul SM (2003) Immunotherapy for Alzheimer’s disease: Will vaccination work? Trends Mol Med 9, 85-87. Broytman O, Malter JS (2004) Anti-Abeta: The good, the bad, and the unforeseen. J Neurosci Res 75, 301-306. Brown ME, Dasilva KA, McLaurin J (2005) Refining an Alzheimer’s vaccine to avoid an inflammatory response. Expert Opin Biol Ther 5, 809-816. Goni F, Sigurdsson EM (2005) New directions towards safer and effective vaccines for Alzheimer’s disease. Curr Opin Mol Ther 7, 17-23. Rosenberg RN (2005) Immunotherapy for Alzheimer disease: The promise and the problem. Arch Neurol 62, 1506-1507. Dasilva KA, Aubert I, McLaurin J (2006) Vaccine development for Alzheimer’s disease. Curr Pharm Des 12, 4283-4293. Sigurdsson EM (2006) Immunotherapy for conformational diseases. Curr Pharm Des 12, 2569-2585. Solomon B (2006) Alzheimer’s disease immunotherapy: From in vitro amyloid immunomodulation to in vivo vaccination. J Alzheimers Dis 9, 433-438. Weiner HL, Frenkel D (2006) Immunology and immunotherapy of Alzheimer’s disease. Nat Rev Immunol 6, 404-416. Mohajeri MH (2007) The underestimated potential of the immune system in prevention of Alzheimer’s disease pathology. Bioessays 29, 927-932. Purcell AW, McCluskey J, Rossjohn J (2007) More than one reason to rethink the use of peptides in vaccine design. Nat Rev Drug Discov 6, 404-414. Solomon B (2007) Beta-amyloidbased immunotherapy as a treatment of Alzheimers disease. Drugs Today (Barc) 43, 333-342. Solomon B (2007) Clinical immunologic approaches for the treatment of Alzheimer’s disease. Expert Opin Investig Drugs 16, 819-828. Solomon B (2007) Active immunization against Alzheimer’s beta-amyloid peptide using phage display technology. Vaccine 25, 3053-3056. Brody DL, Holtzman DM (2008) Active and passive immunotherapy for neurodegenerative disorders. Annu Rev Neurosci 31, 175-193. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1357] [1358] [1359] [1360] [1361] [1362] [1363] [1364] [1365] [1366] [1367] [1368] [1369] [1370] [1371] [1372] [1373] [1374] [1375] [1376] [1377] Nitsch RM, Hock C (2008) Targeting beta-amyloid pathology in Alzheimer’s disease with Abeta immunotherapy. Neurotherapeutics 5, 415-420. Popovich PG, Longbrake EE (2008) Can the immune system be harnessed to repair the CNS? Nat Rev Neurosci 9, 481-493. Solomon B (2008) Immunological approaches for amyloid-beta clearance toward treatment for Alzheimer’s disease. Rejuvenation Res 11, 349-357. Agadjanyan MG, Cribbs DH (2009) Active and passive Abeta-immunotherapy: Preclinical and clinical studies and future directions: Part I. CNS Neurol Disord Drug Targets 8, 1-6. Cribbs DH, Agadjanyan MG (2009) Active and passive Abeta-immunotherapy: Preclinical and clinical studies and future directions: Part II. CNS Neurol Disord Drug Targets 8, 82-87. Foster JK, Verdile G, Bates KA, Martins RN (2009) Immunization in Alzheimer’s disease: Naive hope or realistic clinical potential? Mol Psychiatry 14, 239-251. Solomon B (2009) Immunotherapeutic strategies for Alzheimer’s disease treatment. ScientificWorldJournal 9, 909-919. Cribbs DH (2010) Abeta DNA vaccination for Alzheimer’s disease: Focus on disease prevention. CNS Neurol Disord Drug Targets 9, 207-216. Lemere CA, Masliah E (2010) Can Alzheimer disease be prevented by amyloid-beta immunotherapy? Nat Rev Neurol 6, 108-119. Maarouf CL, Daugs ID, Kokjohn TA, Kalback WM, Patton RL, Luehrs DC, Masliah E, Nicoll JA, Sabbagh MN, Beach TG, Castano EM, Roher AE (2010) The biochemical aftermath of anti-amyloid immunotherapy. Mol Neurodegener 5, 39. Solomon B, Frenkel D (2010) Immunotherapy for Alzheimer’s disease. Neuropharmacology 59, 303-309. von Bernhardi R (2010) Immunotherapy in Alzheimer’s disease: Where do we stand? Where should we go? J Alzheimers Dis 19, 405-421. Wang YJ, Zhou HD, Zhou XF (2010) Modified immunotherapies against Alzheimer’s disease: Toward safer and effective amyloid clearance. J Alzheimers Dis 21, 1065-1075. Wisniewski T, Boutajangout A (2010) Vaccination as a therapeutic approach to Alzheimer’s disease. Mt Sinai J Med 77, 17-31. Menendez-Gonzalez M, Perez-Pinera P, Martinez-Rivera M, Muniz AL, Vega JA (2011) Immunotherapy for Alzheimer’s disease: Rational basis in ongoing clinical trials. Curr Pharm Des 17, 508-520. Morgan D (2011) Immunotherapy for Alzheimer’s disease. J Internal Med 269, 54-63. Schnabel J (2011) Vaccines: Chasing the dream. Nature 475, S18-S19. Shah S, Federoff HJ (2011) Therapeutic potential of vaccines for Alzheimer’s disease. Immunotherapy 3, 287-298. Delrieu J, Ousset PJ, Caillaud C, Vellas B (2012) ‘Clinical trials in Alzheimer’s disease’: Immunotherapy approaches. J Neurochem 120(Suppl 1), 186-193. Lobello K, Ryan JM, Liu E, Rippon G, Black R (2012) Targeting Beta amyloid: A clinical review of immunotherapeutic approaches in Alzheimer’s disease. Int J Alzheimers Dis 2012, 628070. Bach P, Tschape JA, Kopietz F, Braun G, Baade JK, Wiederhold KH, Staufenbiel M, Prinz M, Deller T, Kalinke [1378] [1379] [1380] [1381] [1382] [1383] [1384] [1385] [1386] [1387] [1388] [1389] [1390] 101 U, Buchholz CJ, Muller UC (2009) Vaccination with Abeta-displaying virus-like particles reduces soluble and insoluble cerebral Abeta and lowers plaque burden in APP transgenic mice. J Immunol 182, 7613-7624. Zurbriggen R, Amacker M, Kammer AR, Westerfeld N, Borghgraef P, Van LF, Van dA I, Wera S (2005) Virosomebased active immunization targets soluble amyloid species rather than plaques in a transgenic mouse model of Alzheimer’s disease. J Mol Neurosci 27, 157-166. Salloway S, Sperling R, Gilman S, Fox NC, Blennow K, Raskind M, Sabbagh M, Honig LS, Doody R, van Dyck CH, Mulnard R, Barakos J, Gregg KM, Liu E, Lieberburg I, Schenk D, Black R, Grundman M (2009) A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology 73, 20612070. Black RS, Sperling RA, Safirstein B, Motter RN, Pallay A, Nichols A, Grundman M (2010) A single ascending dose study of bapineuzumab in patients with Alzheimer disease. Alzheimer Dis Assoc Disord 24, 198-203. Laskowitz DT, Kolls BJ (2010) A phase 2 multiple ascending dose trial of bapineuzumab in mild to moderate Alzheimer disease. Neurology 74, 2026-2027. Sperling R, Salloway S, Brooks DJ, Tampieri D, Barakos J, Fox NC, Raskind M, Sabbagh M, Honig LS, Porsteinsson AP, Lieberburg I, Arrighi HM, Morris KA, Lu Y, Liu E, Gregg KM, Brashear HR, Kinney GG, Black R, Grundman M (2012) Amyloid-related imaging abnormalities in patients with Alzheimer’s disease treated with bapineuzumab: A retrospective analysis. Lancet Neurol 11, 241-249. Rinne JO, Brooks DJ, Rossor MN, Fox NC, Bullock R, Klunk WE, Mathis CA, Blennow K, Barakos J, Okello AA, Rodriguez Martinez de LS, Liu E, Koller M, Gregg KM, Schenk D, Black R, Grundman M (2010) 11C-PiB PET assessment of change in fibrillar amyloid-beta load in patients with Alzheimer’s disease treated with bapineuzumab: A phase 2, double-blind, placebo-controlled, ascending-dose study. Lancet Neurol 9, 363-372. Blennow K, Zetterberg H, Rinne JO, Salloway S, Wei J, Black R, Grundman M, Liu E (2012) Effect of immunotherapy with bapineuzumab on cerebrospinal fluid biomarker levels in patients with mild to moderate Alzheimer disease. Arch Neurol 69, 1002-1010. Kerchner GA, Boxer AL (2010) Bapineuzumab. Expert Opin Biol Ther 10, 1121-1130. Wilcock GK (2010) Bapineuzumab in Alzheimer’s disease: Where now? Lancet Neurol 9, 134-136. Panza F, Frisardi V, Imbimbo BP, D’Onofrio G, Pietrarossa G, Seripa D, Pilotto A, Solfrizzi V (2010) Bapineuzumab: Anti-beta-amyloid monoclonal antibodies for the treatment of Alzheimer’s disease. Immunotherapy 2, 767-782. Panza F, Frisardi V, Imbimbo BP, Seripa D, Solfrizzi V, Pilotto A (2011) Monoclonal antibodies against betaamyloid (Abeta) for the treatment of Alzheimer’s disease: The Abeta target at a crossroads. Expert Opin Biol Ther 11, 679-686. 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 immunotherapy for Alzheimer’s disease: Focus on bapineuzumab. Curr Alzheimer Res 8, 808-817. Prins ND, Visser PJ, Scheltens P (2010) Can novel therapeutics halt the amyloid cascade? Alzheimers Res Ther 2, 5. 102 [1391] [1392] [1393] [1394] [1395] [1396] [1397] [1398] [1399] [1400] [1401] [1402] [1403] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Roher AE, Maarouf CL, Daugs ID, Kokjohn TA, Hunter JM, Sabbagh MN, Beach TG (2011) Neuropathology and amyloid-beta spectrum in a bapineuzumab immunotherapy recipient. J Alzheimers Dis 24, 315-325. Zago W, Buttini M, Comery TA, Nishioka C, Gardai SJ, Seubert P, Games D, Bard F, Schenk D, Kinney GG (2012) Neutralization of Soluble, synaptotoxic amyloid beta species by antibodies is epitope specific. J Neurosci 32, 2696-2702. Bard F, Cannon C, Barbour R, Burke RL, Games D, Grajeda H, Guido T, Hu K, Huang J, Johnson-Wood K, Khan K, Kholodenko D, Lee M, Lieberburg I, Motter R, Nguyen M, Soriano F, Vasquez N, Weiss K, Welch B, Seubert P, Schenk D, Yednock T (2000) Peripherally administered antibodies against amyloid beta-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat Med 6, 916-919. Siemers ER, Friedrich S, Dean RA, Gonzales CR, Farlow MR, Paul SM, DeMattos RB (2010) Safety and changes in plasma and cerebrospinal fluid amyloid beta after a single administration of an amyloid beta monoclonal antibody in subjects with Alzheimer disease. Clin Neuropharmacol 33, 67-73. Carlson C, Estergard W, Oh J, Suhy J, Jack CR Jr, Siemers E, Barakos J (2011) Prevalence of asymptomatic vasogenic edema in pretreatment Alzheimer’s disease study cohorts from phase 3 trials of semagacestat and solanezumab. Alzheimers Dement 7, 396-401. Reichert JM (2011) Antibody-based therapeutics to watch in 2011. MAbs 3, 76-99. Samadi H, Sultzer D (2011) Solanezumab for Alzheimer’s disease. Expert Opin Biol Ther 11, 787-798. Imbimbo BP, Ottonello S, Frisardi V, Solfrizzi V, Greco A, Seripa D, Pilotto A, Panza F (2012) Solanezumab for the treatment of mild-to-moderate Alzheimer’s disease. Expert Rev Clin Immunol 8, 135-149. Farlow M, Arnold SE, van Dyck CH, Aisen PS, Snider BJ, Porsteinsson AP, Friedrich S, Dean RA, Gonzales C, Sethuraman G, DeMattos RB, Mohs R, Paul SM, Siemers ER (2012) Safety and biomarker effects of solanezumab in patients with Alzheimer’s disease. Alzheimers Dement 8, 261-271. Ostrowitzki S, Deptula D, Thurfjell L, Barkhof F, Bohrmann B, Brooks DJ, Klunk WE, Ashford E, Yoo K, Xu ZX, Loetscher H, Santarelli L (2012) Mechanism of amyloid removal in patients with Alzheimer disease treated with gantenerumab. Arch Neurol 69, 198-207. 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, Winter M, Nordstedt C, Loetscher H (2012) Gantenerumab: A novel human anti-abeta antibody demonstrates sustained cerebral amyloid-beta binding and elicits cell-mediated removal of human amyloid-beta. J Alzheimers Dis 28, 49-69. Delrieu J, Ousset PJ, Vellas B (2012) Gantenerumab for the treatment of Alzheimer’s disease. Expert Opin Biol Ther 12, 1077-1086. Adolfsson O, Pihlgren M, Toni N, Varisco Y, Buccarello AL, Antoniello K, Lohmann S, Piorkowska K, Gafner V, Atwal JK, Maloney J, Chen M, Gogineni A, Weimer RM, Mortensen DL, Friesenhahn M, Ho C, Paul R, Pfeifer A, Muhs A, Watts RJ (2012) An effector-reduced anti-beta-amyloid (Abeta) antibody with unique Abeta binding properties promotes neuroprotec- [1404] [1405] [1406] [1407] [1408] [1409] [1410] [1411] [1412] [1413] [1414] [1415] [1416] [1417] tion and glial engulfment of Abeta. J Neurosci 32, 96779689. Balakrishnan K, ndrei-Selmer LC, Selmer T, Bacher M, Dodel R (2010) Comparison of intravenous immunoglobulins for naturally occurring autoantibodies against amyloid-beta. J Alzheimers Dis 20, 135-143. Dodel R, Neff F, Noelker C, Pul R, Du Y, Bacher M, Oertel W (2010) Intravenous immunoglobulins as a treatment for Alzheimer’s disease: Rationale and current evidence. Drugs 70, 513-528. Relkin NR (2008) Current state of immunotherapy for Alzheimer’s disease. CNS Spectr 13, 39-41. 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 immunoglobulin for treatment of mild Alzheimer disease. Neurobiol Aging 30, 1728-1736. Hughes RA, Dalakas MC, Cornblath DR, Latov N, Weksler ME, Relkin N (2009) Clinical applications of intravenous immunoglobulins in neurology. Clin Exp Immunol 158(Suppl 1), 34-42. Poetsch V, Neuhaus W, Noe CR (2010) Serum-derived immunoglobulins neutralize adverse effects of amyloidbeta peptide on the integrity of a blood-brain barrier in vitro model. J Alzheimers Dis 21, 303-314. Poetsch V, Benani-Baiti B, Neuhaus W, Muchitsch EM, Noe CR (2010) Serum-derived immunoglobulins alter amyloid beta transport across a blood-brain barrier in vitro model. Pharmazie 65, 267-273. Lanz TA, Schachter JB (2008) Solid-phase extraction enhances detection of beta-amyloid peptides in plasma and enables Abeta quantification following passive immunization with Abeta antibodies. J Neurosci Methods 169, 16-22. 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, Li W, Pons J, Lin JC, Bowes RC (2011) Anti-amyloid therapy protects against retinal pigmented epithelium damage and vision loss in a model of age-related macular degeneration. Proc Natl Acad Sci U S A 108, E279-E287. Venkataramani V, Wirths O, Budka H, Hartig W, Kovacs GG, Bayer TA (2012) Antibody 9D5 recognizes oligomeric pyroglutamate amyloid-beta in a fraction of amyloid-beta deposits in Alzheimer’s disease without cross-reactivity with other protein aggregates. J Alzheimers Dis 29, 361-371. 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, Wang W, Wu Y, Tessier-Lavigne M, Watts RJ (2011) A therapeutic antibody targeting BACE1 inhibits amyloid-beta production in vivo. Sci Transl Med 3, 84ra43. Yu YJ, Zhang Y, Kenrick M, Hoyte K, Luk W, Lu Y, Atwal J, Elliott JM, Prabhu S, Watts RJ, Dennis MS (2011) Boosting brain uptake of a therapeutic antibody by reducing its affinity for a transcytosis target. Sci Transl Med 3, 84ra44. Boado RJ, Zhang Y, Wang Y, Pardridge WM (2009) Engineering and expression of a chimeric transferrin receptor monoclonal antibody for blood-brain barrier delivery in the mouse. Biotechnol Bioeng 102, 1251-1258. Boado RJ, Zhou QH, Lu JZ, Hui EK, Pardridge WM (2010) Pharmacokinetics and brain uptake of a genetically engineered bifunctional fusion antibody targeting the mouse transferrin receptor. Mol Pharm 7, 237-244. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1418] [1419] [1420] [1421] [1422] [1423] [1424] [1425] [1426] [1427] [1428] [1429] [1430] [1431] [1432] [1433] Zhou QH, Fu A, Boado RJ, Hui EK, Lu JZ, Pardridge WM (2011) Receptor-mediated abeta amyloid antibody targeting to Alzheimer’s disease mouse brain. Mol Pharm 8, 280-285. Zhou QH, Boado RJ, Hui EK, Lu JZ, Pardridge WM (2011) Chronic dosing of mice with a transferrin receptor monoclonal antibody-glial-derived neurotrophic factor fusion protein. Drug Metab Dispos 39, 1149-1154. Zhou QH, Boado RJ, Pardridge WM (2012) Selective plasma pharmacokinetics and brain uptake in the mouse of enzyme fusion proteins derived from species-specific receptor-targeted antibodies. J Drug Target 20, 715-719. Pardridge WM, Boado RJ (2012) Reengineering biopharmaceuticals for targeted delivery across the blood-brain barrier. Methods Enzymol 503, 269-292. Boado RJ, Pardridge WM (2011) The Trojan Horse liposome technology for nonviral gene transfer across the blood-brain barrier. J Drug Deliv 2011, 296151. Goldshmit Y, Matteo R, Sztal T, Ellett F, Frisca F, Moreno K, Crombie D, Lieschke GJ, Currie PD, Sabbadini RA, Pebay A (2012) Blockage of lysophosphatidic Acid signaling improves spinal cord injury outcomes. Am J Pathol 181, 978-992. Solomon B, Koppel R, Hanan E, Katzav T (1996) Monoclonal antibodies inhibit in vitro fibrillar aggregation of the Alzheimer beta-amyloid peptide. Proc Natl Acad Sci U S A 93, 452-455. Solomon B, Koppel R, Frankel D, Hanan-Aharon E (1997) Disaggregation of Alzheimer beta-amyloid by sitedirected mAb. Proc Natl Acad Sci U S A 94, 41094112. Buttini M, Masliah E, Barbour R, Grajeda H, Motter R, Johnson-Wood K, Khan K, Seubert P, Freedman S, Schenk D, Games D (2005) Beta-amyloid immunotherapy prevents synaptic degeneration in a mouse model of Alzheimer’s disease. J Neurosci 25, 9096-9101. Morgan D (2005) Mechanisms of A beta plaque clearance following passive A beta immunization. Neurodegener Dis 2, 261-266. Levites Y, Das P, Price RW, Rochette MJ, Kostura LA, McGowan EM, Murphy MP, Golde TE (2006) Anti-Abeta42- and anti-Abeta40-specific mAbs attenuate amyloid deposition in an Alzheimer disease mouse model. J Clin Invest 116, 193-201. Levites Y, Smithson LA, Price RW, Dakin RS, Yuan B, Sierks MR, Kim J, McGowan E, Reed DK, Rosenberry TL, Das P, Golde TE (2006) Insights into the mechanisms of action of anti-Abeta antibodies in Alzheimer’s disease mouse models. FASEB J 20, 2576-2578. Chang WP, Downs D, Huang XP, Da H, Fung KM, Tang J (2007) Amyloid-beta reduction by memapsin 2 (betasecretase) immunization. FASEB J 21, 3184-3196. Chauhan NB (2007) Intracerebroventricular passive immunization with anti-oligoAbeta antibody in TgCRND8. J Neurosci Res 85, 451-463. 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 of Alzheimer’s disease. Proc Natl Acad Sci U S A 104, 1565915664. 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 conformational antibody domain that prevents mature amyloid fibril for- [1434] [1435] [1436] [1437] [1438] [1439] [1440] [1441] [1442] [1443] [1444] [1445] 103 mation by stabilizing Abeta protofibrils. Proc Natl Acad Sci U S A 104, 19232-19237. Kayed R, Head E, Sarsoza F, Saing T, Cotman CW, Necula M, Margol L, Wu J, Breydo L, Thompson JL, Rasool S, Gurlo T, Butler P, Glabe CG (2007) Fibril specific, conformation dependent antibodies recognize a generic epitope common to amyloid fibrils and fibrillar oligomers that is absent in prefibrillar oligomers. Mol Neurodegener 2, 18. Kayed R, Canto I, Breydo L, Rasool S, Lukacsovich T, Wu J, Albay R III, Pensalfini A, Yeung S, Head E, Marsh JL, Glabe C (2010) Conformation dependent monoclonal antibodies distinguish different replicating strains or conformers of prefibrillar Abeta oligomers. Mol Neurodegener 5, 57. Mamikonyan G, Necula M, Mkrtichyan M, Ghochikyan A, Petrushina I, Movsesyan N, Mina E, Kiyatkin A, Glabe CG, Cribbs DH, Agadjanyan MG (2007) Anti-A beta 1-11 antibody binds to different beta-amyloid species, inhibits fibril formation, and disaggregates preformed fibrils but not the most toxic oligomers. J Biol Chem 282, 2237622386. Moretto N, Bolchi A, Rivetti C, Imbimbo BP, Villetti G, Pietrini V, Polonelli L, Del SS, Smith KM, Ferrante RJ, Ottonello S (2007) Conformation-sensitive antibodies against alzheimer amyloid-beta by immunization with a thioredoxin-constrained B-cell epitope peptide. J Biol Chem 282, 11436-11445. Poduslo JF, Ramakrishnan M, Holasek SS, RamirezAlvarado M, Kandimalla KK, Gilles EJ, Curran GL, Wengenack TM (2007) In vivo targeting of antibody fragments to the nervous system for Alzheimer’s disease immunotherapy and molecular imaging of amyloid plaques. J Neurochem 102, 420-433. Rakover I, Arbel M, Solomon B (2007) Immunotherapy against APP beta-secretase cleavage site improves cognitive function and reduces neuroinflammation in Tg2576 mice without a significant effect on brain abeta levels. Neurodegener Dis 4, 392-402. Song MS, Mook-Jung I, Lee HJ, Min JY, Park MH (2007) Serum anti-amyloid-beta antibodies and Alzheimer’s disease in elderly Korean patients. J Int Med Res 35, 301-306. Tampellini D, Magrane J, Takahashi RH, Li F, Lin MT, Almeida CG, Gouras GK (2007) Internalized antibodies to the Abeta domain of APP reduce neuronal Abeta and protect against synaptic alterations. J Biol Chem 282, 18895-18906. Bolukbasi Hatip FF, Matsunaga Y, Yamada T (2008) Specific reactivity of mild/severe Alzheimer’s disease patient’s sera to antibody against Abeta1-40 epitope 17-21. Acta Neurol Scand 117, 404-408. Koenigsknecht-Talboo J, Meyer-Luehmann M, Parsadanian M, Garcia-Alloza M, Finn MB, Hyman BT, Bacskai BJ, Holtzman DM (2008) Rapid microglial response around amyloid pathology after systemic anti-Abeta antibody administration in PDAPP mice. J Neurosci 28, 14156-14164. Manea M, Kalaszi A, Mezo G, Horvati K, Bodor A, Horvath A, Farkas O, Perczel A, Przybylski M, Hudecz F (2008) Antibody recognition and conformational flexibility of a plaque-specific beta-amyloid epitope modulated by non-native peptide flanking regions. J Med Chem 51, 1150-1161. Schroeter S, Khan K, Barbour R, Doan M, Chen M, Guido T, Gill D, Basi G, Schenk D, Seubert P, Games D (2008) 104 [1446] [1447] [1448] [1449] [1450] [1451] [1452] [1453] [1454] [1455] [1456] [1457] [1458] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Immunotherapy reduces vascular amyloid-beta in PDAPP mice. J Neurosci 28, 6787-6793. Tucker SM, Borchelt DR, Troncoso JC (2008) Limited clearance of pre-existing amyloid plaques after intracerebral injection of Abeta antibodies in two mouse models of Alzheimer disease. J Neuropathol Exp Neurol 67, 30-40. 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 and Abeta42 in patients with Alzheimer’s disease and normal controls. Brain Res 1219, 169-179. Itoh T, Satou T, Nishida S, Tsubaki M, Hashimoto S, Ito H (2009) Improvement of cerebral function by anti-amyloid precursor protein antibody infusion after traumatic brain injury in rats. Mol Cell Biochem 324, 191-199. Lord A, Gumucio A, Englund H, Sehlin D, Sundquist VS, Soderberg L, Moller C, Gellerfors P, Lannfelt L, Pettersson FE, Nilsson LN (2009) An amyloid-beta protofibrilselective antibody prevents amyloid formation in a mouse model of Alzheimer’s disease. Neurobiol Dis 36, 425-434. Rozkalne A, Spires-Jones TL, Stern EA, Hyman BT (2009) A single dose of passive immunotherapy has extended benefits on synapses and neurites in an Alzheimer’s disease mouse model. Brain Res 1280, 178-185. Spires-Jones TL, Mielke ML, Rozkalne A, MeyerLuehmann M, de CA, Bacskai BJ, Schenk D, Hyman BT (2009) Passive immunotherapy rapidly increases structural plasticity in a mouse model of Alzheimer disease. Neurobiol Dis 33, 213-220. Xu YX, Wang HQ, Yan J, Sun XB, Guo JC, Zhu CQ (2009) Antibody binding to cell surface amyloid precursor protein induces neuronal injury by deregulating the phosphorylation of focal adhesion signaling related proteins. Neurosci Lett 465, 276-281. 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, HongGang H, Xiang-Lei P, Yan-Peng Z, Ling-Ling H, Tao H (2009) Preparation and characterization of a monoclonal antibody with high affinity for soluble Abeta oligomers. Hybridoma (Larchmt) 28, 349-354. Axelsen TV, Holm A, Birkelund S, Christiansen G, Ploug M, Holm IE (2009) Specific recognition of the C-terminal end of A beta 42 by a high affinity monoclonal antibody. Mol Immunol 46, 2267-2273. Adekar SP, Klyubin I, Macy S, Rowan MJ, Solomon A, Dessain SK, O’Nuallain B (2010) Inherent antiamyloidogenic activity of human immunoglobulin gamma heavy chains. J Biol Chem 285, 1066-1074. Basi GS, Feinberg H, Oshidari F, Anderson J, Barbour R, Baker J, Comery TA, Diep L, Gill D, Johnson-Wood K, Goel A, Grantcharova K, Lee M, Li J, Partridge A, Griswold-Prenner I, Piot N, Walker D, Widom A, Pangalos MN, Seubert P, Jacobsen JS, Schenk D, Weis WI (2010) Structural correlates of antibodies associated with acute reversal of amyloid beta-related behavioral deficits in a mouse model of Alzheimer disease. J Biol Chem 285, 3417-3427. Luo F, Rustay NR, Seifert T, Roesner B, Hradil V, Hillen H, Ebert U, Severin JM, Cox BF, Llano DA, Day M, Fox GB (2010) Magnetic resonance imaging detection and time course of cerebral microhemorrhages during passive immunotherapy in living amyloid precursor protein transgenic mice. J Pharmacol Exp Ther 335, 580-588. Szabo P, Mujalli DM, Rotondi ML, Sharma R, Weber A, Schwarz HP, Weksler ME, Relkin N (2010) Measure- [1459] [1460] [1461] [1462] [1463] [1464] [1465] [1466] [1467] [1468] [1469] [1470] ment of anti-beta amyloid antibodies in human blood. J Neuroimmunol 227, 167-174. Winkler DT, Abramowski D, Danner S, Zurini M, Paganetti P, Tolnay M, Staufenbiel M (2010) Rapid cerebral amyloid binding by Abeta antibodies infused into beta-amyloid precursor protein transgenic mice. Biol Psychiatry 68, 971-974. Lindhagen-Persson M, Brannstrom K, Vestling M, Steinitz M, Olofsson A (2010) Amyloid-beta oligomer specificity mediated by the IgM isotype–implications for a specific protective mechanism exerted by endogenous auto-antibodies. PLoS One 5, e13928. Miller DL, Potempska A, Wegiel J, Mehta PD (2011) High-affinity rabbit monoclonal antibodies specific for amyloid peptides amyloid-beta40 and amyloid-beta42. J Alzheimers Dis 23, 293-305. 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-beta42 oligomer-specific monoclonal antibody improved memory performance in SAMP8 mice. J Alzheimers Dis 23, 551-561. O’Nuallain B, Klyubin I, Mc Donald JM, Foster JS, Welzel A, Barry A, Dykoski RK, Cleary JP, Gebbink MF, Rowan MJ, Walsh DM (2011) A monoclonal antibody against synthetic Abeta dimer assemblies neutralizes brain-derived synaptic plasticity-disrupting Abeta. J Neurochem 119, 189-201. Dodel R, Balakrishnan K, Keyvani K, Deuster O, Neff F, ndrei-Selmer LC, Roskam S, Stuer C, Al-Abed Y, Noelker C, Balzer-Geldsetzer M, Oertel W, Du Y, Bacher M (2011) Naturally occurring autoantibodies against beta-amyloid: Investigating their role in transgenic animal and in vitro models of Alzheimer’s disease. J Neurosci 31, 58475854. Tanaka K, Nishimura M, Yamaguchi Y, Hashiguchi S, Takiguchi S, Yamaguchi M, Tahara H, Gotanda T, Abe R, Ito Y, Sugimura K (2011) A mimotope peptide of Abeta42 fibril-specific antibodies with Abeta42 fibrillation inhibitory activity induces anti-Abeta42 conformer antibody response by a displayed form on an M13 phage in mice. J Neuroimmunol 236, 27-38. 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 basis of beta-amyloid oligomer recognition with a conformational antibody fragment. Proc Natl Acad Sci U S A 109, 12503-12508. 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 typical and atypical Alzheimer disease. Arch Neurol 69, 11811185. Arbel M, Solomon B (2007) A novel immunotherapy for Alzheimer’s disease: Antibodies against the beta-secretase cleavage site of APP. Curr Alzheimer Res 4, 437-445. Arbel M, Solomon B (2007) Immunotherapy for Alzheimer’s disease: Attacking amyloid-beta from the inside. Trends Immunol 28, 511-513. Britschgi M, Wyss-Coray T (2007) Systemic and acquired immune responses in Alzheimer’s disease. Int Rev Neurobiol 82, 205-233. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1471] [1472] [1473] [1474] [1475] [1476] [1477] [1478] [1479] [1480] [1481] [1482] [1483] [1484] [1485] [1486] [1487] Cardinale A, Biocca S (2008) The potential of intracellular antibodies for therapeutic targeting of protein-misfolding diseases. Trends Mol Med 14, 373-380. Lichtlen P, Mohajeri MH (2008) Antibody-based approaches in Alzheimer’s research: Safety, pharmacokinetics, metabolism, and analytical tools. J Neurochem 104, 859-874. Bednar MM (2009) Anti-amyloid antibody drugs in clinical testing for Alzheimer’s disease. IDrugs 12, 566575. Jicha GA (2009) Is passive immunization for Alzheimer’s disease ‘alive and well’ or ‘dead and buried’? Expert Opin Biol Ther 9, 481-491. Lambert MP, Velasco PT, Viola KL, Klein WL (2009) Targeting generation of antibodies specific to conformational epitopes of amyloid beta-derived neurotoxins. CNS Neurol Disord Drug Targets 8, 65-81. Pul R, Dodel R, Stangel M (2011) Antibody-based therapy in Alzheimer’s disease. Expert Opin Biol Ther 11, 343357. Hermann DM, Keyvani K, van de NJ, Weimar C, Wiltfang J, Nitsch RM, Szodorai A (2011) Brain-reactive betaamyloid antibodies in primary CNS angiitis with cerebral amyloid angiopathy. Neurology 77, 503-505. Merlini G (2011) Reducing the amyloid burden through immunotherapy: A major therapeutic advance. Nephrol Dial Transplant 26, 1471-1473. Perchiacca JM, Ladiwala AR, Bhattacharya M, Tessier PM (2012) Structure-based design of conformation- and sequence-specific antibodies against amyloid beta. Proc Natl Acad Sci U S A 109, 84-89. Liu YH, Giunta B, Zhou HD, Tan J, Wang YJ (2012) Immunotherapy for Alzheimer disease-the challenge of adverse effects. Nat Rev Neurol 8, 465-469. Dodart JC, Bales KR, Gannon KS, Greene SJ, DeMattos RB, Mathis C, DeLong CA, Wu S, Wu X, Holtzman DM, Paul SM (2002) Immunization reverses memory deficits without reducing brain Abeta burden in Alzheimer’s disease model. Nat Neurosci 5, 452-457. Yamada K, Yabuki C, Seubert P, Schenk D, Hori Y, Ohtsuki S, Terasaki T, Hashimoto T, Iwatsubo T (2009) Abeta immunotherapy: Intracerebral sequestration of Abeta by an anti-Abeta monoclonal antibody 266 with high affinity to soluble Abeta. J Neurosci 29, 11393-11398. Freeman GB, Lin JC, Pons J, Raha NM (2012) 39week toxicity and toxicokinetic study of ponezumab (PF-04360365) in cynomolgus monkeys with 12-week recovery period. J Alzheimers Dis 28, 531-541. Freeman GB, Brown TP, Wallace K, Bales KR (2012) Chronic administration of an aglycosylated murine antibody of ponezumab does not worsen microhemorrhages in aged Tg2576 mice. Curr Alzheimer Res, [Epub ahead of print]. La Porte SL, Bollini SS, Lanz TA, Abdiche YN, Rusnak AS, Ho WH, Kobayashi D, Harrabi O, Pappas D, Mina EW, Milici AJ, Kawabe TT, Bales K, Lin JC, Pons J (2012) Structural basis of C-terminal beta-amyloid peptide binding by the antibody ponezumab for the treatment of Alzheimer’s disease. J Mol Biol 421, 525-536. Crespo-Biel N, Theunis C, Van Leuven F (2012) Protein tau: Prime cause of synaptic and neuronal degeneration in Alzheimer’s disease. Int J Alzheimers Dis 2012, 251426. Morris M, Maeda S, Vossel K, Mucke L (2011) The many faces of tau. Neuron 70, 410-426. [1488] [1489] [1490] [1491] [1492] [1493] [1494] [1495] [1496] [1497] [1498] [1499] [1500] [1501] [1502] 105 Kolarova M, Garcia-Sierra F, Bartos A, Ricny J, Ripova D (2012) Structure and pathology of tau protein in Alzheimer disease. Int J Alzheimers Dis 2012, 731526. Mandelkow EM, Mandelkow E (2012) Biochemistry and cell biology of tau protein in neurofibrillary degeneration. Cold Spring Harb Perspect Med 2, a006247. Lee VM, Brunden KR, Hutton M, Trojanowski JQ (2011) Developing therapeutic approaches to tau, selected kinases, and related neuronal protein targets. Cold Spring Harb Perspect Med 1, a006437. Santacruz K, Lewis J, Spires T, Paulson J, Kotilinek L, Ingelsson M, Guimaraes A, DeTure M, Ramsden M, McGowan E, Forster C, Yue M, Orne J, Janus C, Mariash A, Kuskowski M, Hyman B, Hutton M, Ashe KH (2005) Tau suppression in a neurodegenerative mouse model improves memory function. Science 309, 476481. Berger Z, Roder H, Hanna A, Carlson A, Rangachari V, Yue M, Wszolek Z, Ashe K, Knight J, Dickson D, Andorfer C, Rosenberry TL, Lewis J, Hutton M, Janus C (2007) Accumulation of pathological tau species and memory loss in a conditional model of tauopathy. J Neurosci 27, 36503662. 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 [Epub ahead of print]. Martin L, Latypova X, Wilson CM, Magnaudeix A, Perrin ML, Yardin C, Terro F (2012) Tau protein kinases: Involvement in Alzheimer’s disease. Ageing Res Rev 12, 289-309. Bekris LM, Millard S, Lutz F, Li G, Galasko DR, Farlow MR, Quinn JF, Kaye JA, Leverenz JB, Tsuang DW, Yu CE, Peskind ER (2012) Tau phosphorylation pathway genes and cerebrospinal fluid tau levels in Alzheimer’s disease. Am J Med Genet B Neuropsychiatr Genet 159B, 874-883. Kester MI, van d V, Blankenstein MA, Pijnenburg YA, van Elk EJ, Scheltens P, van der Flier WM (2009) CSF biomarkers predict rate of cognitive decline in Alzheimer disease. Neurology 73, 1353-1358. Ittner LM, Gotz J (2011) Amyloid-beta and tau–a toxic pas de deux in Alzheimer’s disease. Nat Rev Neurosci 12, 65-72. 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 function of tau mediates amyloid-beta toxicity in Alzheimer’s disease mouse models. Cell 142, 387-397. Ittner A, Ke YD, van EJ, Gladbach A, Gotz J, Ittner LM (2011) Brief update on different roles of tau in neurodegeneration. IUBMB Life 63, 495-502. Roberson ED, Halabisky B, Yoo JW, Yao J, Chin J, Yan F, Wu T, Hamto P, Devidze N, Yu GQ, Palop JJ, Noebels JL, Mucke L (2011) Amyloid-beta/Fyn-induced synaptic, network, and cognitive impairments depend on tau levels in multiple mouse models of Alzheimer’s disease. J Neurosci 31, 700-711. Gotz J, Ittner A, Ittner LM (2012) Tau-targeted treatment strategies in Alzheimer’s disease. Br J Pharmacol 165, 1246-1259. Mondragon-Rodriguez S, Perry G, Zhu X, Boehm J (2012) Amyloid Beta and tau proteins as therapeutic targets for Alzheimer’s disease treatment: Rethinking the current strategy. Int J Alzheimers Dis 2012, 630182. 106 [1503] [1504] [1505] [1506] [1507] [1508] [1509] [1510] [1511] [1512] [1513] [1514] [1515] [1516] [1517] [1518] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Tai HC, Serrano-Pozo A, Hashimoto T, Frosch MP, SpiresJones TL, Hyman BT (2012) The synaptic accumulation of hyperphosphorylated tau oligomers in Alzheimer disease is associated with dysfunction of the ubiquitin-proteasome system. Am J Pathol 181, 1426-1435. Jack CR Jr, Knopman DS, Jagust WJ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Trojanowski JQ (2010) Hypothetical model of dynamic biomarkers of the Alzheimer’s pathological cascade. Lancet Neurol 9, 119-128. 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 ordering of Alzheimer disease biomarkers. Arch Neurol 68, 15261535. Jack CR Jr, Vemuri P, Wiste HJ, Weigand SD, Lesnick TG, Lowe V, Kantarci K, Bernstein MA, Senjem ML, Gunter JL, Boeve BF, Trojanowski JQ, Shaw LM, Aisen PS, Weiner MW, Petersen RC, Knopman DS (2012) Shapes of the trajectories of 5 major biomarkers of Alzheimer disease. Arch Neurol 69, 856-867. Han SD, Gruhl J, Beckett L, Dodge HH, Stricker NH, Farias S, Mungas D (2012) Beta amyloid, tau, neuroimaging, and cognition: Sequence modeling of biomarkers for Alzheimer’s disease. Brain Imaging Behav, doi: 10.1007/s11682-012-9177-0 [Epub ahead of print]. Wischik CM, Bentham P, Wishik DJ, SK (2008) Tau aggregation inhibitor (TAI) therapy with Rember™ arrests disease progression in mild to moderate Alzheimer’s disease over 50 weeks. Alzheimers Dementia 4, T167. Wischik C, Staff R (2009) Challenges in the conduct of disease-modifying trials in AD: Practical experience from a phase 2 trial of Tau-aggregation inhibitor therapy. J Nutr Health Aging 13, 367-369. Wischik CM, Edwards PC, Lai RY, Roth M, Harrington CR (1996) Selective inhibition of Alzheimer disease-like tau aggregation by phenothiazines. Proc Natl Acad Sci U S A 93, 11213-11218. Atamna H, Nguyen A, Schultz C, Boyle K, Newberry J, Kato H, Ames BN (2008) Methylene blue delays cellular senescence and enhances key mitochondrial biochemical pathways. FASEB J 22, 703-712. Oz M, Lorke DE, Petroianu GA (2009) Methylene blue and Alzheimer’s disease. Biochem Pharmacol 78, 927-932. Oz M, Lorke DE, Hasan M, Petroianu GA (2011) Cellular and molecular actions of Methylene Blue in the nervous system. Med Res Rev 31, 93-117. Oz M, Isaev D, Lorke DE, Hasan M, Petroianu G, Shippenberg TS (2012) Methylene blue inhibits function of the 5-HT transporter. Br J Pharmacol 166, 168-176. Atamna H, Kumar R (2010) Protective role of methylene blue in Alzheimer’s disease via mitochondria and cytochrome c oxidase. J Alzheimers Dis 20(Suppl 2), S439-S452. Schirmer RH, Adler H, Pickhardt M, Mandelkow E (2011) “Lest we forget you–methylene blue...”. Neurobiol Aging 32, 2325-2316. 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, Gestwicki JE, Dickey CA (2010) Phenothiazine-mediated rescue of cognition in tau transgenic mice requires neuroprotection and reduced soluble tau burden. Mol Neurodegener 5, 45. Medina DX, Caccamo A, Oddo S (2011) Methylene blue reduces abeta levels and rescues early cognitive deficit by increasing proteasome activity. Brain Pathol 21, 140-149. [1519] [1520] [1521] [1522] [1523] [1524] [1525] [1526] [1527] [1528] [1529] [1530] [1531] [1532] [1533] Necula M, Breydo L, Milton S, Kayed R, van d V, Tone P, Glabe CG (2007) Methylene blue inhibits amyloid Abeta oligomerization by promoting fibrillization. Biochemistry 46, 8850-8860. del Ser T, Steinwachs KC, Gertz HJ, Andres MV, GomezCarrillo B, Medina M, Vericat JA, Redondo P, Fleet D, Leon T (2012) Treatment of Alzheimer’s disease with the GSK-3 inhibitor tideglusib: A pilot study. J Alzheimers Dis, doi: 10.3233/JAD-2012-120805 [Epub ahead of print]. Rojo LE, Alzate-Morales J, Saavedra IN, Davies P, Maccioni RB (2010) Selective interaction of lansoprazole and astemizole with tau polymers: Potential new clinical use in diagnosis of Alzheimer’s disease. J Alzheimers Dis 19, 573-589. Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M, Starkova N, Calingasan NY, Yang L, Tampellini D, Starkov AA, Chan RB, Di PG, Pujol A, Beal MF (2012) Bezafibrate administration improves behavioral deficits and tau pathology in P301S mice. Hum Mol Genet 21, 5091-5105. Brunden KR, Zhang B, Carroll J, Yao Y, Potuzak JS, Hogan AM, Iba M, James MJ, Xie SX, Ballatore C, Smith AB III, Lee VM, Trojanowski JQ (2010) Epothilone D improves microtubule density, axonal integrity, and cognition in a transgenic mouse model of tauopathy. J Neurosci 30, 13861-13866. Yang Y, Ma D, Wang Y, Jiang T, Hu S, Zhang M, Yu X, Gong CX (2012) Intranasal insulin ameliorates tau hyperphosphorylation in a rat model of type 2 diabetes. J Alzheimers Dis, doi: 10.3233/JAD-2012-121294-2012 [Epub ahead of print]. 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 selective tau-PHF ligands. J Alzheimers Dis 27, 835-843. Martin L, Latypova X, Wilson CM, Magnaudeix A, Perrin ML, Terro F (2012) Tau protein phosphatases in Alzheimer’s disease: The leading role of PP2A. Ageing Res Rev 12, 39-49. Braithwaite SP, Stock JB, Lombroso PJ, Nairn AC (2012) Protein phosphatases and Alzheimer’s disease. Prog Mol Biol Transl Sci 106, 343-379. 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’s disease models. Proc Natl Acad Sci U S A 107, 1388813893. Yuzwa SA, Macauley MS, Heinonen JE, Shan X, Dennis RJ, He Y, Whitworth GE, Stubbs KA, McEachern EJ, Davies GJ, Vocadlo DJ (2008) A potent mechanism-inspired O-GlcNAcase inhibitor that blocks phosphorylation of tau in vivo. Nat Chem Biol 4, 483-490. Yuzwa SA, Vocadlo DJ (2009) O-GlcNAc modification and the tauopathies: Insights from chemical biology. Curr Alzheimer Res 6, 451-454. Yuzwa SA, Yadav AK, Skorobogatko Y, Clark T, Vosseller K, Vocadlo DJ (2011) Mapping O-GlcNAc modification sites on tau and generation of a site-specific O-GlcNAc tau antibody. Amino Acids 40, 857-868. Yuzwa SA, Shan X, Macauley MS, Clark T, Skorobogatko Y, Vosseller K, Vocadlo DJ (2012) Increasing O-GlcNAc slows neurodegeneration and stabilizes tau against aggregation. Nat Chem Biol 8, 393-399. Shen DL, Gloster TM, Yuzwa SA, Vocadlo DJ (2012) Insights into O-GlcNAc processing and dynamics W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1534] [1535] [1536] [1537] [1538] [1539] [1540] [1541] [1542] [1543] [1544] [1545] [1546] [1547] [1548] [1549] through kinetic analysis of O-GlcNAc transferase and O-GlcNAcase activity on protein substrates. J Biol Chem 287, 15395-15408. Fischer PM (2008) Turning down tau phosphorylation. Nat Chem Biol 4, 448-449. Lefebvre T (2012) Recall sugars, forget Alzheimer’s. Nature Chem Biol 8, 325-326. Navarrete LP, Perez P, Morales I, Maccioni RB (2011) Novel drugs affecting tau behavior in the treatment of Alzheimer’s disease and tauopathies. Curr Alzheimer Res 8, 678-685. Navarrete LP, Guzman L, San MA, studillo-Saavedra L, Maccioni RB (2012) Molecules of the quinoline family block tau self-aggregation: Implications toward a therapeutic approach for Alzheimer’s disease. J Alzheimers Dis 29, 79-88. Calcul L, Zhang B, Jinwal UK, Dickey CA, Baker BJ (2012) Natural products as a rich source of tau-targeting drugs for Alzheimer’s disease. Future Med Chem 4, 17511761. Chirita C, Necula M, Kuret J (2004) Ligand-dependent inhibition and reversal of tau filament formation. Biochemistry 43, 2879-2887. Necula M, Chirita CN, Kuret J (2005) Cyanine dye N744 inhibits tau fibrillization by blocking filament extension: Implications for the treatment of tauopathic neurodegenerative diseases. Biochemistry 44, 10227-10237. Pickhardt M, von BM, Gazova Z, Hascher A, Biernat J, Mandelkow EM, Mandelkow E (2005) Screening for inhibitors of tau polymerization. Curr Alzheimer Res 2, 219-226. Pickhardt M, Gazova Z, von BM, Khlistunova I, Wang Y, Hascher A, Mandelkow EM, Biernat J, Mandelkow E (2005) Anthraquinones inhibit tau aggregation and dissolve Alzheimer’s paired helical filaments in vitro and in cells. J Biol Chem 280, 3628-3635. Khlistunova I, Biernat J, Wang Y, Pickhardt M, von BM, Gazova Z, Mandelkow E, Mandelkow EM (2006) Inducible expression of Tau repeat domain in cell models of tauopathy: Aggregation is toxic to cells but can be reversed by inhibitor drugs. J Biol Chem 281, 1205-1214. Bulic B, Pickhardt M, Khlistunova I, Biernat J, Mandelkow EM, Mandelkow E, Waldmann H (2007) Rhodanine-based tau aggregation inhibitors in cell models of tauopathy. Angew Chem Int Ed Engl 46, 9215-9219. Khlistunova I, Pickhardt M, Biernat J, Wang Y, Mandelkow EM, Mandelkow E (2007) Inhibition of tau aggregation in cell models of tauopathy. Curr Alzheimer Res 4, 544-546. Pickhardt M, Larbig G, Khlistunova I, Coksezen A, Meyer B, Mandelkow EM, Schmidt B, Mandelkow E (2007) Phenylthiazolyl-hydrazide and its derivatives are potent inhibitors of tau aggregation and toxicity in vitro and in cells. Biochemistry 46, 10016-10023. Pickhardt M, Biernat J, Khlistunova I, Wang YP, Gazova Z, Mandelkow EM, Mandelkow E (2007) N-phenylamine derivatives as aggregation inhibitors in cell models of tauopathy. Curr Alzheimer Res 4, 397-402. Crowe A, Ballatore C, Hyde E, Trojanowski JQ, Lee VM (2007) High throughput screening for small molecule inhibitors of heparin-induced tau fibril formation. Biochem Biophys Res Commun 358, 1-6. Honson NS, Jensen JR, Darby MV, Kuret J (2007) Potent inhibition of tau fibrillization with a multivalent ligand. Biochem Biophys Res Commun 363, 229-234. [1550] [1551] [1552] [1553] [1554] [1555] [1556] [1557] [1558] [1559] [1560] [1561] [1562] [1563] [1564] 107 Larbig G, Pickhardt M, Lloyd DG, Schmidt B, Mandelkow E (2007) Screening for inhibitors of tau protein aggregation into Alzheimer paired helical filaments: A ligand based approach results in successful scaffold hopping. Curr Alzheimer Res 4, 315-323. Bulic B, Pickhardt M, Schmidt B, Mandelkow EM, Waldmann H, Mandelkow E (2009) Development of tau aggregation inhibitors for Alzheimer’s disease. Angew Chem Int Ed Engl 48, 1740-1752. Chang E, Congdon EE, Honson NS, Duff KE, Kuret J (2009) Structure-activity relationship of cyanine tau aggregation inhibitors. J Med Chem 52, 3539-3547. Chang E, Honson NS, Bandyopadhyay B, Funk KE, Jensen JR, Kim S, Naphade S, Kuret J (2009) Modulation and detection of tau aggregation with small-molecule ligands. Curr Alzheimer Res 6, 409-414. Crowe A, Huang W, Ballatore C, Johnson RL, Hogan AM, 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 of aminothienopyridazine inhibitors of tau assembly by quantitative high-throughput screening. Biochemistry 48, 77327745. Honson NS, Jensen JR, Abraha A, Hall GF, Kuret J (2009) Small-molecule mediated neuroprotection in an in situ model of tauopathy. Neurotox Res 15, 274-283. Ballatore C, Brunden KR, Piscitelli F, James MJ, Crowe A, Yao Y, Hyde E, Trojanowski JQ, Lee VM, Smith AB III (2010) Discovery of brain-penetrant, orally bioavailable aminothienopyridazine inhibitors of tau aggregation. J Med Chem 53, 3739-3747. Bulic B, Pickhardt M, Mandelkow EM, Mandelkow E (2010) Tau protein and tau aggregation inhibitors. Neuropharmacology 59, 276-289. Schafer KN, Murale DP, Kim K, Cisek K, Kuret J, Churchill DG (2011) Structure-activity relationship of cyclic thiacarbocyanine tau aggregation inhibitors. Bioorg Med Chem Lett 21, 3273-3276. Martin L, Magnaudeix A, Wilson CM, Yardin C, Terro F (2011) The new indirubin derivative inhibitors of glycogen synthase kinase-3, 6-BIDECO and 6-BIMYEO, prevent tau phosphorylation and apoptosis induced by the inhibition of protein phosphatase-2A by okadaic acid in cultured neurons. J Neurosci Res 89, 1802-1811. Ballatore C, Crowe A, Piscitelli F, James M, Lou K, Rossidivito G, Yao Y, Trojanowski JQ, Lee VM, Brunden KR, Smith AB III (2012) Aminothienopyridazine inhibitors of tau aggregation: Evaluation of structure-activity relationship leads to selection of candidates with desirable in vivo properties. Bioorg Med Chem 20, 4451-4461. Ballatore C, Brunden KR, Huryn DM, Trojanowski JQ, Lee VM, Smith AB III (2012) Microtubule stabilizing agents as potential treatment for Alzheimer’s disease and related neurodegenerative tauopathies. J Med Chem 55, 8979-8996. Castro A, Martinez A (2000) Inhibition of tau phosphorylation: A new therapeutic strategy for the treatment of Alzheimer’s disease and other neurodegenerative disorders. Expert Opin Ther Patents 10, 1519-1527. Lau LF, Schachter JB, Seymour PA, Sanner MA (2002) Tau protein phosphorylation as a therapeutic target in Alzheimer’s disease. Curr Top Med Chem 2, 395-415. Dickey CA, Petrucelli L (2006) Current strategies for the treatment of Alzheimer’s disease and other tauopathies. Expert Opin Ther Targets 10, 665-676. 108 [1565] [1566] [1567] [1568] [1569] [1570] [1571] [1572] [1573] [1574] [1575] [1576] [1577] [1578] [1579] [1580] [1581] [1582] [1583] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Ballatore C, Lee VM, Trojanowski JQ (2007) Taumediated neurodegeneration in Alzheimer’s disease and related disorders. Nat Rev Neurosci 8, 663-672. Gong CX, Iqbal K (2008) Hyperphosphorylation of microtubule-associated protein tau: A promising therapeutic target for Alzheimer disease. Curr Med Chem 15, 2321-2328. Honson NS, Kuret J (2008) Tau aggregation and toxicity in tauopathic neurodegenerative diseases. J Alzheimers Dis 14, 417-422. Iqbal K, Grundke-Iqbal I (2008) Alzheimer neurofibrillary degeneration: Significance, etiopathogenesis, therapeutics and prevention. J Cell Mol Med 12, 38-55. Schneider A, Mandelkow E (2008) Tau-based treatment strategies in neurodegenerative diseases. Neurotherapeutics 5, 443-457. Brunden KR, Trojanowski JQ, Lee VM (2009) Advances in tau-focused drug discovery for Alzheimer’s disease and related tauopathies. Nat Rev Drug Discov 8, 783-793. Hanger DP, Anderton BH, Noble W (2009) Tau phosphorylation: The therapeutic challenge for neurodegenerative disease. Trends Mol Med 15, 112-119. Brunden KR, Ballatore C, Crowe A, Smith AB III, Lee VM, Trojanowski JQ (2010) Tau-directed drug discovery for Alzheimer’s disease and related tauopathies: A focus on tau assembly inhibitors. Exp Neurol 223, 304-310. Gong CX, Grundke-Iqbal I, Iqbal K (2010) Targeting tau protein in Alzheimer’s disease. Drugs Aging 27, 351-365. Meraz-Rios MA, Lira-De Leon KI, Campos-Pena V, De Anda-Hernandez MA, Mena-Lopez R (2010) Tau oligomers and aggregation in Alzheimer’s disease. J Neurochem 112, 1353-1367. Ballatore C, Brunden KR, Trojanowski JQ, Lee VM, Smith AB, Huryn DM (2011) Modulation of protein-protein interactions as a therapeutic strategy for the treatment of neurodegenerative tauopathies. Curr Top Med Chem 11, 317-330. Corbo CP, Alonso AC (2011) Therapeutic targets in Alzheimer’s disease and related tauopathies. Prog Mol Biol Transl Sci 98, 47-83. Noble W, Pooler AM, Hanger DP (2011) Advances in taubased drug discovery. Expert Opin Drug Discov 6, 797810. Fuentes P, Catalan J (2011) A clinical perspective: Anti tau’s treatment in Alzheimer’s disease. Curr Alzheimer Res 8, 686-688. Himmelstein DS, Ward SM, Lancia JK, Patterson KR, Binder LI (2012) Tau as a therapeutic target in neurodegenerative disease. Pharmacol Ther 136, 8-22. Ward SM, Himmelstein DS, Lancia JK, Binder LI (2012) Tau oligomers and tau toxicity in neurodegenerative disease. Biochem Soc Trans 40, 667-671. Karakaya T, Fusser F, Prvulovic D, Hampel H (2012) Treatment options for tauopathies. Curr Treat Options Neurol 14, 126-136. Miller Y, Ma B, Nussinov R (2011) Synergistic interactions between repeats in tau protein and Abeta amyloids may be responsible for accelerated aggregation via polymorphic states. Biochemistry 50, 5172-5181. Zhang W, Arteaga J, Cashion DK, Chen G, Gangadharmath U, Gomez LF, Kasi D, Lam C, Liang Q, Liu C, Mocharla VP, Mu F, Sinha A, Szardenings AK, Wang E, Walsh JC, Xia C, Yu C, Zhao T, Kolb HC (2012) A highly selective and specific PET tracer for imaging of tau pathologies. J Alzheimers Dis 31, 601-612. [1584] [1585] [1586] [1587] [1588] [1589] [1590] [1591] [1592] [1593] [1594] [1595] [1596] [1597] Fodero-Tavoletti MT, Okamura N, Furumoto S, Mulligan RS, Connor AR, McLean CA, Cao D, Rigopoulos A, Cartwright GA, O’Keefe G, Gong S, Adlard PA, Barnham KJ, Rowe CC, Masters CL, Kudo Y, Cappai R, Yanai K, Villemagne VL (2011) 18F-THK523: A novel in vivo tau imaging ligand for Alzheimer’s disease. Brain 134, 1089-1100. Okamura N, Furumoto S, Harada R, Furukawa K, Arai H, Yanai K, Kudo Y (2011) Phenylquinoline derivatives for in vivo imaging of tau pathology in Alzheimer’s disease. Alzheimers Dement 7 (Suppl), e38. Jones C, Nairne J (2012) Use of cyanine dyes for the detection of tau for diagnosis of early-stage tauopathies. WO 2012/068072, Prior. Nov. 19, 2010. Watanabe H, Ono M, Kimura H, Matsumura K, Yoshimura M, Okamoto Y, Ihara M, Takahashi R, Saji H (2012) Synthesis and biological evaluation of novel oxindole derivatives for imaging neurofibrillary tangles in Alzheimer’s disease. Bioorg Med Chem Lett 22, 57005703. Bolander A, Kieser D, Voss C, Bauer S, Schon C, Burgold S, Bittner T, Holzer J, Heyny-von HR, Mall G, Goetschy V, Czech C, Knust H, Berger R, Herms J, Hilger I, Schmidt B (2012) Bis(arylvinyl)pyrazines, -pyrimidines, and -pyridazines as imaging agents for tau fibrils and betaamyloid plaques in Alzheimer’s disease models. J Med Chem 55, 9170-9180. Jensen JR, Cisek K, Funk KE, Naphade S, Schafer KN, Kuret J (2011) Research towards tau imaging. J Alzheimers Dis 26(Suppl 3), 147-157. Jensen JR, Cisek K, Honson NS, Kuret J (2011) Ligand polarizability contributes to tau fibril binding affinity. Bioorg Med Chem 19, 5147-5154. Boimel M, Grigoriadis N, Lourbopoulos A, Haber E, Abramsky O, Rosenmann H (2010) Efficacy and safety of immunization with phosphorylated tau against neurofibrillary tangles in mice. Exp Neurol 224, 472-485. Vana L, Kanaan NM, Ugwu IC, Wuu J, Mufson EJ, Binder LI (2011) Progression of tau pathology in cholinergic Basal forebrain neurons in mild cognitive impairment and Alzheimer’s disease. Am J Pathol 179, 25332550. Patterson KR, Remmers C, Fu Y, Brooker S, Kanaan NM, Vana L, Ward S, Reyes JF, Philibert K, Glucksman MJ, Binder LI (2011) Characterization of prefibrillar Tau oligomers in vitro and in Alzheimer disease. J Biol Chem 286, 23063-23076. Rosenmann H, Meiner Z, Geylis V, Abramsky O, Steinitz M (2006) Detection of circulating antibodies against tau protein in its unphosphorylated and in its neurofibrillary tangles-related phosphorylated state in Alzheimer’s disease and healthy subjects. Neurosci Lett 410, 90-93. Yen SH, Crowe A, Dickson DW (1985) Monoclonal antibodies to Alzheimer neurofibrillary tangles. 1. Identification of polypeptides. Am J Pathol 120, 282-291. Taniguchi T, Sumida M, Hiraoka S, Tomoo K, Kakehi T, Minoura K, Sugiyama S, Inaka K, Ishida T, Saito N, Tanaka C (2005) Effects of different anti-tau antibodies on tau fibrillogenesis: RTA-1 and RTA-2 counteract tau aggregation. FEBS Lett 579, 1399-1404. Asuni AA, Boutajangout A, Quartermain D, Sigurdsson EM (2007) Immunotherapy targeting pathological tau conformers in a tangle mouse model reduces brain pathology with associated functional improvements. J Neurosci 27, 9115-9129. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1598] [1599] [1600] [1601] [1602] [1603] [1604] [1605] [1606] [1607] [1608] [1609] [1610] [1611] [1612] [1613] [1614] [1615] [1616] [1617] Sigurdsson EM (2008) Immunotherapy targeting pathological tau protein in Alzheimer’s disease and related tauopathies. J Alzheimers Dis 15, 157-168. Zilka N, Kontsekova E, Novak M (2008) Chaperone-like antibodies targeting misfolded tau protein: New vistas in the immunotherapy of neurodegenerative foldopathies. J Alzheimers Dis 15, 169-179. Kayed R, Jackson GR (2009) Prefilament tau species as potential targets for immunotherapy for Alzheimer disease and related disorders. Curr Opin Immunol 21, 359-363. Sigurdsson EM (2009) Tau-focused immunotherapy for Alzheimer’s disease and related tauopathies. Curr Alzheimer Res 6, 446-450. Boutajangout A, Quartermain D, Sigurdsson EM (2010) Immunotherapy targeting pathological tau prevents cognitive decline in a new tangle mouse model. J Neurosci 30, 16559-16566. Kayed R (2010) Anti-tau oligomers passive vaccination for the treatment of Alzheimer disease. Hum Vaccin 6, 931-935. Boutajangout A, Ingadottir J, Davies P, Sigurdsson EM (2011) Passive immunization targeting pathological phospho-tau protein in a mouse model reduces functional decline and clears tau aggregates from the brain. J Neurochem 118, 658-667. Boutajangout A, Sigurdsson EM, Krishnamurthy PK (2011) Tau as a therapeutic target for Alzheimer’s disease. Curr Alzheimer Res 8, 666-677. Gu J, Sigurdsson EM (2011) Immunotherapy for tauopathies. J Mol Neurosci 45, 690-695. Krishnamurthy PK, Deng Y, Sigurdsson EM (2011) Mechanistic studies of antibody-mediated clearance of tau aggregates using an ex vivo brain slice model. Front Psychiatry 2, 59. Mattson MP (2000) Emerging neuroprotective strategies for Alzheimer’s disease: Dietary restriction, telomerase activation, and stem cell therapy. Exp Gerontol 35, 489502. Sugaya K, Brannen CL (2001) Stem cell strategies for neuroreplacement therapy in Alzheimer’s disease. Med Hypotheses 57, 697-700. Tanne JH (2005) Activating stem cells may treat Alzheimer’s. BMJ 330, 622. Wang QH, Xu RX, Nagao S (2005) Transplantation of cholinergic neural stem cells in a mouse model of Alzheimer’s disease. Chin Med J (Engl) 118, 508-511. Heese K, Low JW, Inoue N (2006) Nerve growth factor, neural stem cells and Alzheimer’s disease. Neurosignals 15, 1-12. Wang Q, Matsumoto Y, Shindo T, Miyake K, Shindo A, Kawanishi M, Kawai N, Tamiya T, Nagao S (2006) Neural stem cells transplantation in cortex in a mouse model of Alzheimer’s disease. J Med Invest 53, 61-69. Sugaya K, Alvarez A, Marutle A, Kwak YD, Choumkina E (2006) Stem cell strategies for Alzheimer’s disease therapy. Panminerva Med 48, 87-96. Fullwood NJ (2007) Neural stem cells, acetylcholine and Alzheimer’s disease. Nat Chem Biol 3, 435. Sugaya K, Kwak YD, Ohmitsu O, Marutle A, Greig NH, Choumrina E (2007) Practical issues in stem cell therapy for Alzheimer’s disease. Curr Alzheimer Res 4, 370377. Sugaya K, Merchant S (2008) How to approach Alzheimer’s disease therapy using stem cell technologies. J Alzheimers Dis 15, 241-254. [1618] [1619] [1620] [1621] [1622] [1623] [1624] [1625] [1626] [1627] [1628] [1629] [1630] [1631] [1632] [1633] [1634] [1635] [1636] 109 Waldau B, Shetty AK (2008) Behavior of neural stem cells in the Alzheimer brain. Cell Mol Life Sci 65, 2372-2384. Lee JK, Jin HK, Bae JS (2009) Bone marrow-derived mesenchymal stem cells reduce brain amyloid-beta deposition and accelerate the activation of microglia in an acutely induced Alzheimer’s disease mouse model. Neurosci Lett 450, 136-141. Feng Z, Zhao G, Yu L (2009) Neural stem cells and Alzheimer’s disease: Challenges and hope. Am J Alzheimer’s Dis Other Demen 24, 52-57. Taupin P (2009) Adult neurogenesis and the pathogenesis of Alzheimer’s disease. Med Sci Monit 15, LE1. Taupin P (2009) Adult neurogenesis, neural stem cells and Alzheimer’s disease: Developments, limitations, problems and promises. Curr Alzheimer Res 6, 461-470. Dantuma E, Merchant S, Sugaya K (2010) Stem cells for the treatment of neurodegenerative diseases. Stem Cell Res Ther 1, 37. Habisch HJ, Schmid B, von Arnim CA, Ludolph AC, Brenner R, Storch A (2010) Efficient processing of Alzheimer’s disease amyloid-beta peptides by neuroectodermally converted mesenchymal stem cells. Stem Cells Dev 19, 629-633. Lee HJ, Lee JK, Lee H, Shin JW, Carter JE, Sakamoto T, Jin HK, Bae JS (2010) The therapeutic potential of human umbilical cord blood-derived mesenchymal stem cells in Alzheimer’s disease. Neurosci Lett 481, 30-35. Lee JK, Jin HK, Endo S, Schuchman EH, Carter JE, Bae JS (2010) Intracerebral transplantation of bone marrowderived mesenchymal stem cells reduces amyloid-beta deposition and rescues memory deficits in Alzheimer’s disease mice by modulation of immune responses. Stem Cells 28, 329-343. Stellos K, Panagiota V, Sachsenmaier S, Trunk T, Straten G, Leyhe T, Seizer P, Geisler T, Gawaz M, Laske C (2010) Increased circulating progenitor cells in Alzheimer’s disease patients with moderate to severe dementia: Evidence for vascular repair and tissue regeneration? J Alzheimers Dis 19, 591-600. Taupin P (2010) Adult neurogenesis and neural stem cells as a model for the discovery and development of novel drugs. Expert Opin Drug Discov 5, 921-925. Taupin P (2010) Aging and neurogenesis, a lesion from Alzheimer’s disease. Aging Dis 1, 158-168. Schwarz SC, Schwarz J (2010) Translation of stem cell therapy for neurological diseases. Transl Res 156, 155160. Couillard-Despres S, Iglseder B, Aigner L (2011) Neurogenesis, cellular plasticity and cognition: The impact of stem cells in the adult and aging brain–a mini-review. Gerontology 57, 559-564. Lunn JS, Sakowski SA, Hur J, Feldman EL (2011) Stem cell technology for neurodegenerative diseases. Ann Neurol 70, 353-361. Taupin P (2011) Neurogenesis, NSCs, pathogenesis and therapies for Alzheimer’s disease. Front Biosci (Schol Ed) 3, 178-190. Abdel-Salam OM (2011) Stem cell therapy for Alzheimer’s disease. CNS Neurol Disord Drug Targets 10, 459-485. Chen C, Xiao SF (2011) Induced pluripotent stem cells and neurodegenerative diseases. Neurosci Bull 27, 107-114. Borlongan CV (2012) Recent preclinical evidence advancing cell therapy for Alzheimer’s disease. Exp Neurol 237, 142-146. 110 [1637] [1638] [1639] [1640] [1641] [1642] [1643] [1644] [1645] [1646] [1647] [1648] [1649] [1650] [1651] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Rachubinski AL, Maclean KN, Evans JR, Bjugstad KB (2012) Modulating cognitive deficits and tau accumulation in a mouse model of aging down syndrome through neonatal implantation of neural progenitor cells. Exp Gerontol 47, 723-733. Rachubinski AL, Crowley SK, Sladek Jr, Maclean KN, Bjugstad KB (2012) Effects of neonatal neural progenitor cell implantation on adult neuroanatomy and cognition in the Ts65Dn model of Down syndrome. PLoS One 7, e36082. Njie e, Kantorovich S, Astary GW, Green C, Zheng T, Semple-Rowland SL, Steindler DA, Sarntinoranont M, Streit WJ, Borchelt DR (2012) A preclinical assessment of neural stem cells as delivery vehicles for anti-amyloid therapeutics. PLoS One 7, e34097. Khandekar N, Lie KH, Sachdev PS, Sidhu KS (2012) Amyloid precursor proteins, neural differentiation of pluripotent stem cells and its relevance to Alzheimer’s disease. Stem Cells Dev 21, 997-1006. Feng Z, Gao F (2012) Stem cell challenges in the treatment of neurodegenerative disease. CNS Neurosci Ther 18, 142148. Kitiyanant N, Kitiyanant Y, Svendsen CN, Thangnipon W (2012) BDNF-, IGF-1- and GDNF-secreting human neural progenitor cells rescue amyloid beta-induced toxicity in cultured rat septal neurons. Neurochem Res 37, 143-152. Park D, Joo SS, Kim TK, Lee SH, Kang H, Lee HJ, Lim I, Matsuo A, Tooyama I, Kim YB, Kim SU (2012) Human neural stem cells overexpressing choline acetyltransferase restore cognitive function of kainic acid-induced learning and memory deficit animals. Cell Transplant 21, 365-371. Park D, Lee HJ, Joo SS, Bae DK, Yang G, Yang YH, Lim I, Matsuo A, Tooyama I, Kim YB, Kim SU (2012) Human neural stem cells over-expressing choline acetyltransferase restore cognition in rat model of cognitive dysfunction. Exp Neurol 234, 521-526. Kern DS, Maclean KN, Jiang H, Synder EY, Sladek JR, Bjugstad KB (2011) Neural stem cells reduce hippocampal tau and reelin accumulation in aged Ts65Dn down syndrome mice. Cell Transplant 20, 371-379. Xuan AG, Long DH, Gu HG, Yang DD, Hong LP, Leng SL (2008) BDNF improves the effects of neural stem cells on the rat model of Alzheimer’s disease with unilateral lesion of fimbria-fornix. Neurosci Lett 440, 331-335. Xuan AG, Luo M, Ji WD, Long DH (2009) Effects of engrafted neural stem cells in Alzheimer’s disease rats. Neurosci Lett 450, 167-171. Blurton-Jones M, Kitazawa M, Martinez-Coria H, Castello NA, Muller FJ, Loring JF, Yamasaki TR, Poon WW, Green KN, LaFerla FM (2009) Neural stem cells improve cognition via BDNF in a transgenic model of Alzheimer disease. Proc Natl Acad Sci U S A 106, 13594-13599. Lee HJ, Lim IJ, Park SW, Ko YB, Kim SU (2012) Human neural stem cells genetically modified to express human nerve growth factor (NGF) gene restore cognition in mouse with ibotenic acid-induced cognitive dysfunction. Cell Transplant, doi: http://dx.doi.org/10.3727/ 096368912X638964 Lee HJ, Lim IJ, Lee MC, Kim SU (2010) Human neural stem cells genetically modified to overexpress brainderived neurotrophic factor promote functional recovery and neuroprotection in a mouse stroke model. J Neurosci Res 88, 3282-3294. Wu S, Sasaki A, Yoshimoto R, Kawahara Y, Manabe T, Kataoka K, Asashima M, Yuge L (2008) Neural stem cells [1652] [1653] [1654] [1655] [1656] [1657] [1658] [1659] [1660] [1661] [1662] [1663] [1664] improve learning and memory in rats with Alzheimer’s disease. Pathobiology 75, 186-194. Wu L, Sluiter AA, Guo HF, Balesar RA, Swaab DF, Zhou JN, Verwer RW (2008) Neural stem cells improve neuronal survival in cultured postmortem brain tissue from aged and Alzheimer patients. J Cell Mol Med 12, 16111621. Yagi T, Ito D, Okada Y, Akamatsu W, Nihei Y, Yoshizaki T, Yamanaka S, Okano H, Suzuki N (2011) Modeling familial Alzheimer’s disease with induced pluripotent stem cells. Hum Mol Genet 20, 4530-4539. Israel MA, Yuan SH, Bardy C, Reyna SM, Mu Y, Herrera C, Hefferan MP, Van GS, Nazor KL, Boscolo FS, Carson CT, Laurent LC, Marsala M, Gage FH, Remes AM, Koo EH, Goldstein LS (2012) Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 482, 216-220. Choi SH, Tanzi RE (2012) iPSCs to the rescue in Alzheimer’s research. Cell Stem Cell 10, 235-236. Shi Y, Kirwan P, Smith J, MacLean G, Orkin SH, Livesey FJ (2012) A human stem cell model of early Alzheimer’s disease pathology in Down syndrome. Sci Transl Med 4, 124ra29. Camnasio S, Carri AD, Lombardo A, Grad I, Mariotti C, 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 several induced pluripotent stem cell lines from homozygous and heterozygous Huntington’s disease patients demonstrates mutation related enhanced lysosomal activity. Neurobiol Dis 46, 41-51. Sadan O, Bahat-Stromza M, Barhum Y, Levy YS, Pisnevsky A, Peretz H, Ilan AB, Bulvik S, Shemesh N, Krepel D, Cohen Y, Melamed E, Offen D (2009) Protective effects of neurotrophic factor-secreting cells in a 6-OHDA rat model of Parkinson disease. Stem Cells Dev 18, 11791190. Sadan O, Shemesh N, Barzilay R, Bahat-Stromza M, Melamed E, Cohen Y, Offen D (2008) Migration of neurotrophic factors-secreting mesenchymal stem cells toward a quinolinic acid lesion as viewed by magnetic resonance imaging. Stem Cells 26, 2542-2551. McGill TJ, Cottam B, Lu B, Wang S, Girman S, Tian C, Huhn SL, Lund RD, Capela A (2012) Transplantation of human central nervous system stem cells - neuroprotection in retinal degeneration. Eur J Neurosci 35, 468-477. Lu P, Wang Y, Graham L, McHale K, Gao M, Wu D, Brock J, Blesch A, Rosenzweig ES, Havton LA, Zheng B, Conner JM, Marsala M, Tuszynski MH (2012) Long-distance growth and connectivity of neural stem cells after severe spinal cord injury. Cell 150, 1264-1273. Glass JD, Boulis NM, Johe K, Rutkove SB, Federici T, Polak M, Kelly C, Feldman EL (2012) Lumbar intraspinal injection of neural stem cells in patients with amyotrophic lateral sclerosis: Results of a phase I trial in 12 patients. Stem Cells 30, 1144-1151. Borlongan CV, Skinner SJ, Geaney M, Vasconcellos AV, Elliott RB, Emerich DF (2004) CNS grafts of rat choroid plexus protect against cerebral ischemia in adult rats. Neuroreport 15, 1543-1547. Borlongan CV, Skinner SJ, Geaney M, Vasconcellos AV, Elliott RB, Emerich DF (2004) Neuroprotection by encapsulated choroid plexus in a rodent model of Huntington’s disease. Neuroreport 15, 2521-2525. W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1665] [1666] [1667] [1668] [1669] [1670] [1671] [1672] [1673] [1674] [1675] [1676] [1677] [1678] [1679] [1680] [1681] Brinton RD, Wang JM (2006) Therapeutic potential of neurogenesis for prevention and recovery from Alzheimer’s disease: Allopregnanolone as a proof of concept neurogenic agent. Curr Alzheimer Res 3, 185-190. Wang JM, Singh C, Liu L, Irwin RW, Chen S, Chung EJ, Thompson RF, Brinton RD (2010) Allopregnanolone reverses neurogenic and cognitive deficits in mouse model of Alzheimer’s disease. Proc Natl Acad Sci U S A 107, 6498-6503. Singh C, Liu L, Wang JM, Irwin RW, Yao J, Chen S, Henry S, Thompson RF, Brinton RD (2012) Allopregnanolone restores hippocampal-dependent learning and memory and neural progenitor survival in aging 3xTgAD and nonTg mice. Neurobiol Aging 33, 1493-1506. Tsai KJ, Tsai YC, Shen CK (2007) G-CSF rescues the memory impairment of animal models of Alzheimer’s disease. J Exp Med 204, 1273-1280. Zhang XZ, Li XJ, Zhang HY (2010) Valproic acid as a promising agent to combat Alzheimer’s disease. Brain Res Bull 81, 3-6. Ling S, Zhou J, Rudd JA, Hu Z, Fang M (2011) The recent updates of therapeutic approaches against abeta for the treatment of Alzheimer’s disease. Anat Rec (Hoboken) 294, 1307-1318. Ulloa CM, Towfigh A, Safdieh J (2009) Review of levetiracetam, with a focus on the extended release formulation, as adjuvant therapy in controlling partial-onset seizures. Neuropsychiatr Dis Treat 5, 467-476. Lyseng-Williamson KA (2011) Levetiracetam: A review of its use in epilepsy. Drugs 71, 489-514. Lyseng-Williamson KA (2011) Spotlight on levetiracetam in epilepsy. CNS Drugs 25, 901-905. Lynch BA, Lambeng N, Nocka K, Kensel-Hammes P, Bajjalieh SM, Matagne A, Fuks B (2004) The synaptic vesicle protein SV2A is the binding site for the antiepileptic drug levetiracetam. Proc Natl Acad Sci U S A 101, 98619866. Lambeng N, Gillard M, Vertongen P, Fuks B, Chatelain P (2005) Characterization of [(3)H]ucb 30889 binding to synaptic vesicle protein 2A in the rat spinal cord. Eur J Pharmacol 520, 70-76. Gillard M, Chatelain P, Fuks B (2006) Binding characteristics of levetiracetam to synaptic vesicle protein 2A (SV2A) in human brain and in CHO cells expressing the human recombinant protein. Eur J Pharmacol 536, 102-108. Dong M, Yeh F, Tepp WH, Dean C, Johnson EA, Janz R, Chapman ER (2006) SV2 is the protein receptor for botulinum neurotoxin A. Science 312, 592-596. Yeh FL, Dong M, Yao J, Tepp WH, Lin G, Johnson EA, Chapman ER (2010) SV2 mediates entry of tetanus neurotoxin into central neurons. PLoS Pathog 6, e1001207. Vogl C, Mochida S, Wolff C, Whalley BJ, Stephens GJ (2012) The synaptic vesicle glycoprotein 2A ligand levetiracetam inhibits presynaptic Ca2+ channels through an intracellular pathway. Mol Pharmacol 82, 199-208. Angehagen M, Margineanu DG, Ben-Menachem E, Ronnback L, Hansson E, Klitgaard H (2003) Levetiracetam reduces caffeine-induced Ca2+ transients and epileptiform potentials in hippocampal neurons. Neuroreport 14, 471-475. Ueda Y, Doi T, Nagatomo K, Tokumaru J, Takaki M, Willmore LJ (2007) Effect of levetiracetam on molecular regulation of hippocampal glutamate and GABA transporters in rats with chronic seizures induced by amygdalar FeCl3 injection. Brain Res 1151, 55-61. [1682] [1683] [1684] [1685] [1686] [1687] [1688] [1689] [1690] [1691] [1692] [1693] [1694] [1695] [1696] [1697] 111 Noyer M, Gillard M, Matagne A, Henichart JP, Wulfert E (1995) The novel antiepileptic drug levetiracetam (ucb L059) appears to act via a specific binding site in CNS membranes. Eur J Pharmacol 286, 137-146. Loscher W, Honack D, Bloms-Funke P (1996) The novel antiepileptic drug levetiracetam (ucb L059) induces alterations in GABA metabolism and turnover in discrete areas of rat brain and reduces neuronal activity in substantia nigra pars reticulata. Brain Res 735, 208-216. Sills GJ, Leach JP, Fraser CM, Forrest G, Patsalos PN, Brodie MJ (1997) Neurochemical studies with the novel anticonvulsant levetiracetam in mouse brain. Eur J Pharmacol 325, 35-40. Celikyurt IK, Ulak G, Mutlu O, Akar FY, Mulayim S, Erden F, Komsuoglu SS (2012) Positive impact of levetiracetam on emotional learning and memory in naive mice. Life Sci 90, 185-189. Sanchez PE, Zhu L, Verret L, Vossel KA, Orr AG, Cirrito JR, Devidze N, Ho K, Yu GQ, Palop JJ, Mucke L (2012) Levetiracetam suppresses neuronal network dysfunction and reverses synaptic and cognitive deficits in an Alzheimer’s disease model. Proc Natl Acad Sci U S A 109, E2895-E2903. Cho JR, Koo DL, Joo EY, Yoon SM, Ju E, Lee J, Kim DY, Hong SB (2012) Effect of levetiracetam monotherapy on background EEG activity and cognition in drug-naive epilepsy patients. Clin Neurophysiol 123, 883-891. Schiemann-Delgado J, Yang H, Loge CL, Stalvey TJ, Jones J, Legoff D, Mintz M (2012) A long-term open-label extension study assessing cognition and behavior, tolerability, safety, and efficacy of adjunctive levetiracetam in children aged 4 to 16 years with partial-onset seizures. J Child Neurol 27, 80-89. Schulze-Bonhage A (2011) Brivaracetam for the treatment of epilepsy. Expert Opin Pharmacother 12, 1959-1966. Stephen LJ, Brodie MJ (2011) Pharmacotherapy of epilepsy: Newly approved and developmental agents. CNS Drugs 25, 89-107. Gillard M, Fuks B, Leclercq K, Matagne A (2011) Binding characteristics of brivaracetam, a selective, high affinity SV2A ligand in rat, mouse and human brain: Relationship to anti-convulsant properties. Eur J Pharmacol 664, 36-44. Zona C, Pieri M, Carunchio I, Curcio L, Klitgaard H, Margineanu DG (2010) Brivaracetam (ucb 34714) inhibits Na(+) current in rat cortical neurons in culture. Epilepsy Res 88, 46-54. Detrait ER, Leclercq K, Loscher W, Potschka H, Niespodziany I, Hanon E, Kaminski RM, Matagne A, Lamberty Y (2010) Brivaracetam does not alter spatial learning and memory in both normal and amygdalakindled rats. Epilepsy Res 91, 74-83. Meador KJ, Gevins A, Leese PT, Otoul C, Loring DW (2011) Neurocognitive effects of brivaracetam, levetiracetam, and lorazepam. Epilepsia 52, 264-272. Selkoe DJ (2012) Preventing Alzheimer’s disease. Science 337, 1488-1492. Froestl W, Muhs A, Pfeifer A (2012) Cognitive Enhancers (Nootropics). Part 2: Drugs Interacting with Enzymes. J Alzheimers Dis 33, 547-658. Morimoto K, Horio J, Satoh H, Sue L, Beach T, Arita S, Tooyama I, Konishi Y (2011) Expression profiles of cytokines in the brains of Alzheimer’s disease (AD) patients compared to the brains of non-demented patients with and without increasing AD pathology. J Alzheimers Dis 25, 59-76. 112 [1698] [1699] [1700] [1701] [1702] [1703] [1704] [1705] [1706] [1707] [1708] [1709] [1710] [1711] W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes Zhou W, Zhong GF, Roa Xr, Xie H, Zeng SG, Chi TY, Zou LB, Wu DH, Hu WH (2012) Identification of aminopyridazine-derived antineuroinflammatory agents effective in an Alzheimer’s mouse model. ACS Med Chem Lett, dx.doi.org/10.1021/ml3001769Vom 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’s disease-like pathology and cognitive decline. Nat Med, doi: 10.1038/nm.2965Chan D (2012) American Chemical Society - 33rd National Medicinal Chemistry Symposium (NMCS). Drugs of the Future 37, 599-603. Thompson AD, Scaglione KM, Prensner J, Gillies AT, Chinnaiyan A, Paulson HL, Jinwal UK, Dickey CA, Gestwicki JE (2012) Analysis of the tau-associated proteome reveals that exchange of hsp70 for hsp90 is involved in tau degradation. ACS Chem Biol 7, 1677-1686. Berridge MJ (2013) Dysregulation of neural calcium signaling in Alzheimer disease, bipolar disorder and schizophrenia. Prion 7 [Epub ahead of print]. Corbett A, Pickett J, Burns A, Corcoran J, Dunnett SB, Edison P, Hagan JJ, Holmes C, Jones E, Katona C, Kearns I, Kehoe P, Mudher A, Passmore A, Shepherd N, Walsh F, Ballard C (2012) Drug repositioning for Alzheimer’s disease. Nat Rev Drug Discov 11, 833-846. Gholamipour-Badie H, Naderi N, Khodagholi F, Shaerzadeh F, Motamedi F (2013) L-type calcium channel blockade alleviates molecular and reversal spatial learning and memory alterations induced by entorhinal amyloid pathology in rats. Behav Brain Res 237, 190-199. Mufson EJ, He B, Nadeem M, Perez SE, Counts SE, Leurgans S, Fritz J, Lah J, Ginsberg SD, Wuu J, Scheff SW (2012) Hippocampal ProNGF Signaling Pathways and beta-Amyloid Levels in Mild Cognitive Impairment and Alzheimer Disease. J Neuropathol Exp Neurol 71, 10181029. 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. Bush AI (2012) The Metal Theory of Alzheimer’s Disease. J Alzheimers Dis, doi: 10.3233/JAD-2012-129011Wei G, Yunbo C, Chen DF, Lai XP, Liu DH, Deng RD, Zhou JH, Zhang SX, Li YW, Li H, Liu LF (2012) beta-Asarone Inhibits Neuronal Apoptosis via the CaMKII/CREB/Bcl-2 Signaling Pathway in an in vitro Model and AbetaPP/PS1 Mice. J Alzheimers Dis, doi: 10.3233/JAD-2012-120865Brunhofer G, Fallarero A, Karlsson D, Batista-Gonzalez A, Shinde P, Gopi MC, Vuorela P (2012) Exploration of natural compounds as sources of new bifunctional scaffolds targeting cholinesterases and beta amyloid aggregation: The case of chelerythrine. Bioorg Med Chem 20, 6669-6679. Zhao X, Zou Y, Xu H, Fan L, Guo H, Li X, Li G, Zhang X, Dong M (2012) Gastrodin protect primary cultured rat hippocampal neurons against amyloid-beta peptide-induced neurotoxicity via ERK1/2-Nrf2 pathway. Brain Res 1482, 13-21. 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 [1712] [1713] [1714] [1715] [1716] [1717] [1718] [1719] [1720] [1721] [1722] [1723] [1724] [1725] beta-amyloid in APP/PS1 transgenic mice. Aging Cell, doi: 10.1111/acel.12022Zou Y, Hong B, Fan L, Zhou L, Liu Y, Wu Q, Zhang X, Dong M (2012) Protective effect of puerarin against beta-amyloid-induced oxidative stress in neuronal cultures from rat hippocampus: involvement of the GSK-3beta/Nrf2 signaling pathway. Free Radic Res, doi:10.3109/10715762.2012.742518Ho L, Ferruzzi MG, Janle EM, Wang J, Gong B, Chen TY, Lobo J, Cooper B, Wu QL, Talcott ST, Percival SS, Simon JE, Pasinetti GM (2012) Identification of braintargeted bioactive dietary quercetin-3-O-glucuronide as a novel intervention for Alzheimer’s disease. FASEB J, doi: 10.1096/fj.12-212118McCracken C, Hudson P, Ellis R, McCaddon A (2006) Methylmalonic acid and cognitive function in the Medical Research Council Cognitive Function and Ageing Study. Am J Clin Nutr 84, 1406-1411. Moore E, Mander A, Ames D, Carne R, Sanders K, Watters D (2012) Cognitive impairment and vitamin B12: a review. Int Psychogeriatr, 1-16. Llewellyn DJ, Langa KM, Lang IA (2009) Serum 25-hydroxyvitamin D concentration and cognitive impairment. J Geriatr Psychiatry Neurol 22, 188-195. 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. Llewellyn DJ, Lang IA, Langa KM, Melzer D (2011) Vitamin D and cognitive impairment in the elderly U.S. population. J Gerontol A Biol Sci Med Sci 66, 59-65. Wang L, Ankati H, Akubathini SK, Balderamos M, Storey CA, Patel AV, Price V, Kretzschmar D, Biehl ER, D’Mello SR (2010) Identification of novel 1,4-benzoxazine compounds that are protective in tissue culture and in vivo models of neurodegeneration. J Neurosci Res 88, 19701984. Hampel H (2012) Amyloid-beta and Cognition in Aging and Alzheimer’s Disease: Molecular and Neurophysiological Mechanisms. J Alzheimers Dis, doi: 10.3233/JAD-2012-129003Lim YY, Pietrzak RH, Ellis KA, Jaeger J, Harrington K, Ashwood T, Szoeke C, Martins RN, Bush AI, Masters CL, Rowe CC, Villemagne VL, Ames D, Darby D, Maruff P (2012) Rapid Decline in Episodic Memory in Healthy Older Adults with High Amyloid-beta. J Alzheimers Dis, doi: 10.3233/JAD-2012-121516Lim YY, Ellis KA, Pietrzak RH, Ames D, Darby D, Harrington K, Martins RN, Masters CL, Rowe C, Savage G, Szoeke C, Villemagne VL, Maruff P (2012) Stronger effect of amyloid load than APOE genotype on cognitive decline in healthy older adults. Neurology 79, 16451652. Lim YY, Ellis KA, Harrington K, Pietrzak RH, Gale J, Ames D, Bush AI, Darby D, Martins RN, Masters CL, Rowe CC, Savage G, Szoeke C, Villemagne VL, Maruff P (2012) Cognitive Decline in Adults with Amnestic Mild Cognitive Impairment and High Amyloid-beta: Prodromal Alzheimer’s Disease? J Alzheimers Dis, doi: 10.3233/JAD-121771Said G, Grippon S, Kirkpatrick P (2012) Tafamidis. Nat Rev Drug Discov 11, 185-186. Echeverria V, Zeitlin R, Burgess S, Patel S, Barman A, Thakur G, Mamcarz M, Wang L, Sattelle DB, Kirschner W. Froestl et al. / Cognitive Enhancers (Nootropics). Part 3: Drugs Interacting with Targets other than Receptors or Enzymes [1726] [1727] [1728] [1729] [1730] [1731] [1732] [1733] [1734] [1735] [1736] [1737] [1738] DA, Mori T, Leblanc RM, Prabhakar R, Arendash GW (2011) Cotinine reduces amyloid-beta aggregation and improves memory in Alzheimer’s disease mice. J Alzheimers Dis 24, 817-835. Ghosh S (2012) Alzheimer’s Association International Conference. Drugs of the Future 37, 663-670. O’Hare E, Scopes DI, Kim EM, Palmer P, Jones M, Whyment AD, Spanswick D, Amijee H, Nerou E, McMahon B, Treherne JM, Jeggo R (2012) Orally bioavailable small molecule drug protects memory in Alzheimer’s disease models. Neurobiol Aging, doi:10.1016/j.neurobiolaging.2012.10.016McKoy AF, Chen J, Schupbach T, Hecht MH (2012) A Novel Inhibitor of Amyloid beta (Abeta) Peptide Aggregation: From high throughput screening to efficacy in an animal model of Alzheimer disease 1. J Biol Chem 287, 38992-39000. Amijee H, Bate C, Williams A, Virdee J, Jeggo R, Spanswick D, Scopes DI, Treherne JM, Mazzitelli S, Chawner R, Eyers CE, Doig AJ (2012) The N-Methylated Peptide SEN304 Powerfully Inhibits Abeta(1-42) Toxicity by Perturbing Oligomer Formation. Biochemistry, doi: 10.1021/bi300415vMathis CA, Mason NS, Lopresti BJ, Klunk WE (2012) Development of Positron Emission Tomography betaAmyloid Plaque Imaging Agents. Semin Nucl Med 42, 423-432. Landau SM, Breault C, Joshi AD, Pontecorvo M, Mathis CA, Jagust WJ, Mintun MA (2012) Amyloid-beta Imaging with Pittsburgh Compound B and Florbetapir: Comparing Radiotracers and Quantification Methods. J Nucl Med, doi: 10.2967/jnumed.112.109009Handen BL, Cohen AD, Channamalappa U, Bulova P, Cannon SA, Cohen WI, Mathis CA, Price JC, Klunk WE (2012) Imaging brain amyloid in nondemented young adults with Down syndrome using Pittsburgh compound B. Alzheimers Dement 8, 496-501. Doraiswamy PM, Sperling RA, Coleman RE, Johnson KA, Reiman EM, Davis MD, Grundman M, Sabbagh MN, Sadowsky CH, Fleisher AS, Carpenter A, Clark CM, Joshi AD, Mintun MA, Skovronsky DM, Pontecorvo MJ (2012) Amyloid-beta assessed by florbetapir F 18 PET and 18month cognitive decline: A multicenter study. Neurology 79, 1636-1644. Furst AJ, Kerchner GA (2012) From Alois to Amyvid: Seeing Alzheimer disease. Neurology 79, 1628-1629. Poisnel G, Dhilly M, Moustie O, Delamare J, Abbas A, Guilloteau D, Barre L (2012) PET imaging with [18F]AV45 in an APP/PS1-21 murine model of amyloid plaque deposition. Neurobiol Aging 33, 2561-2571. 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, Dyrks T, Cappai R (2012) In vitro characterization of [(18)F]florbetaben, an Abeta imaging radiotracer. Nucl Med Biol 39, 1042-1048. Swahn BM, Sandell J, Pyring D, Bergh M, Jeppsson F, Jureus A, Neelissen J, Johnstrom P, Schou M, Svensson S (2012) Synthesis and evaluation of pyridylbenzofuran, pyridylbenzothiazole and pyridylbenzoxazole derivatives as (1)(8)F-PET imaging agents for beta-amyloid plaques. Bioorg Med Chem Lett 22, 4332-4337. Swahn BM, Wensbo D, Sandell J, Sohn D, Slivo C, Pyring D, Malmstrom J, Arzel E, Vallin M, Bergh M, Jeppsson F, Johnson AE, Jureus A, Neelissen J, Svensson S (2010) [1739] [1740] [1741] [1742] [1743] [1744] [1745] [1746] [1747] [1748] [1749] [1750] [1751] [1752] [1753] 113 Synthesis and evaluation of 2-pyridylbenzothiazole, 2pyridylbenzoxazole and 2-pyridylbenzofuran derivatives as 11C-PET imaging agents for beta-amyloid plaques. Bioorg Med Chem Lett 20, 1976-1980. Nelson LD, Siddarth P, Kepe V, Scheibel KE, Huang SC, Barrio JR, Small GW (2011) Positron emission tomography of brain beta-amyloid and tau levels in adults with Down syndrome. Arch Neurol 68, 768-774. Shin J, Kepe V, Barrio JR, Small GW (2011) The merits of FDDNP-PET imaging in Alzheimer’s disease. J Alzheimers Dis 26(Suppl 3), 135-145. Montanes M, Casabona D, Sarasa L, Pesini P, Sarasa M (2012) Prevention of Amyloid-beta Fibril Formation Using Antibodies Against the C-terminal Region of Amyloidbeta1-40 and Amyloid-beta1-42. J Alzheimers Dis, doi: 10.3233/JAD-120850Bard F, Fox M, Friedrich S, Seubert P, Schenk D, Kinney GG, Yednock T (2012) Sustained levels of antibodies against Abeta in amyloid-rich regions of the CNS following intravenous dosing in human APP transgenic mice. Exp Neurol 238, 38-43. Solomon B (2007) Intravenous immunoglobulin and Alzheimer’s disease immunotherapy. Curr Opin Mol Ther 9, 79-85. Robert R, Wark KL (2012) Engineered antibody approaches for Alzheimer’s disease immunotherapy. Arch Biochem Biophys 526, 132-138. Yu G, Li Y, Tian Q, Liu R, Wang Q, Wang JZ, Wang X (2011) Berberine attenuates calyculin A-induced cytotoxicity and Tau hyperphosphorylation in HEK293 cells. J Alzheimers Dis 24, 525-535. Peng Y, Hu Y, Xu S, Li P, Li J, Lu L, Yang H, Feng N, Wang L, Wang X (2012) L-3-n-butylphthalide reduces tau phosphorylation and improves cognitive deficits in AbetaPP/PS1-Alzheimer’s transgenic mice. J Alzheimers Dis 29, 379-391. Tolstykh T, Lee J, Vafai S, Stock JB (2000) Carboxyl methylation regulates phosphoprotein phosphatase 2A by controlling the association of regulatory B subunits. EMBO J 19, 5682-5691. Vafai SB, Stock JB (2002) Protein phosphatase 2A methylation: A link between elevated plasma homocysteine and Alzheimer’s Disease. FEBS Lett 518, 1-4. Xing Y, Li Z, Chen Y, Stock JB, Jeffrey PD, Shi Y (2008) Structural mechanism of demethylation and inactivation of protein phosphatase 2A. Cell 133, 154-163. Butler KV, Kalin J, Brochier C, Vistoli G, Langley B, Kozikowski AP (2010) Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin A. J Am Chem Soc 132, 10842-10846. Benner L, Kalin JH, Kozikowski A, Gordon MN, Morgan D, Selenica MB (2012) Selective HDAC6 inhibition decreases tau levels and improves memory deficits in the rtg4510 mouse model. Society for Neuroscience, New Orleans, Poster 150.04-Sunday, November 14, 2012. Okamura N, Suemoto T, Furumoto S, Suzuki M, Shimadzu H, Akatsu H, Yamamoto T, Fujiwara H, Nemoto M, Maruyama M, Arai H, Yanai K, Sawada T, Kudo Y (2005) Quinoline and benzimidazole derivatives: Candidate probes for in vivo imaging of tau pathology in Alzheimer’s disease. J Neurosci 25, 10857-10862. Gu J, Anumala UR, Lo MF, Kramer T, Heyny von HR, Holzer J, Goetschy-Meyer V, Mall G, Hilger I, Czech C, Schmidt B (2012) 2-Styrylindolium based fluorescent probes visualize neurofibrillary tan- 114 [1754] [1755] [1756] [1757] [1758] [1759] 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.2012.09.109Chai X, Wu S, Murray TK, Kinley R, Cella CV, Sims H, Buckner N, Hanmer J, Davies P, O’Neill MJ, Hutton ML, Citron M (2011) Passive immunization with antiTau antibodies in two transgenic models: reduction of Tau pathology and delay of disease progression. J Biol Chem 286, 34457-34467. Shih HH, Tu C, Cao W, Klein A, Ramsay R, Fennell BJ, Lambert M, Ni SD, Autin B, Kouranova E, Laxmanan S, Braithwaite S, Wu L, it-Zahra M, Milici AJ, Dumin JA, Lavallie ER, Arai M, Corcoran C, Paulsen JE, Gill D, Cunningham O, Bard J, Mosyak L, Finlay WJ (2012) An ultra-specific avian antibody to phosphorylated tau reveals a unique mechanism for phosphoepitope recognition. J Biol Chem, doi: 10.1074/jbc.M112.415935Wang W, Fan L, Xu D, Wen Z, Yu R, Ma Q (2012) Immunotherapy for Alzheimer’s disease. Acta Biochim Biophys Sin (Shanghai) 44, 807-814. Young JE, Goldstein LS (2012) Alzheimer’s disease in a dish: promises and challenges of human stem cell models. Hum Mol Genet 21, R82-R89. Grskovic M, Javaherian A, Strulovici B, Daley GQ (2011) Induced pluripotent stem cells–opportunities for disease modelling and drug discovery. Nat Rev Drug Discov 10, 915-929. Uchida N, Chen K, Dohse M, Hansen KD, Dean J, Buser JR, 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, [1760] [1761] [1762] [1763] [1764] [1765] [1766] Back SA (2012) Human neural stem cells induce functional myelination in mice with severe dysmyelination. Sci Transl Med 4, 155ra136Gupta N, Henry RG, Strober J, Kang SM, Lim DA, Bucci M, Caverzasi E, Gaetano L, Mandelli ML, Ryan T, Perry R, Farrell J, Jeremy RJ, Ulman M, Huhn SL, Barkovich AJ, Rowitch DH (2012) Neural stem cell engraftment and myelination in the human brain. Sci Transl Med 4, 155ra137Moreira PI, Santos RX, Zhu X, Lee HG, Smith MA, Casadesus G, Perry G (2010) Autophagy in Alzheimer’s disease. Expert Rev Neurother 10, 1209-1218. Barnett A, Brewer GJ (2011) Autophagy in aging and Alzheimer’s disease: Pathologic or protective? J Alzheimers Dis 25, 385-394. Schaeffer V, Goedert M (2012) Stimulation of autophagy is neuroprotective in a mouse model of human tauopathy. Autophagy 8, http://dx.doi.org/10.4161/auto.21488Schaeffer V, Lavenir I, Ozcelik S, Tolnay M, Winkler DT, Goedert M (2012) Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy. Brain 135, 2169-2177. Brown JR, Crawford BE, Esko JD (2007) Glycan antagonists and inhibitors: A fount for drug discovery. Crit Rev Biochem Mol Biol 42, 481-515. 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 improves cognition in amnestic mild cognitive impairment. Neuron 74, 467-474.