New Approaches for the Treatment of Alzheimer's Disease

Total Page:16

File Type:pdf, Size:1020Kb

New Approaches for the Treatment of Alzheimer's Disease Digest for Bioorg. Med. Chem. Lett. New Approaches for the Treatment of Alzheimer’s Disease Paul V. Fish,a,b,* David Steadman,a Elliott D. Bayle,a,b and Paul Whitinga,c a Alzheimer’s Research UK UCL Drug Discovery Institute, The Cruciform Building, University College London, Gower Street, London WC1N 1E 6BT, UK. b The Francis Crick Institute, 1 Midland Road, Kings Cross, London NW1 1AT, UK. c The Dementia Research Institute, The Cruciform Building, University College London, Gower Street, London WC1N 1E 6BT, UK. Key words: Alzheimer’s disease, neurodegeneration, neuroinflammation, drug discovery pipeline, symptomatic treatments, disease modification. 1 Digest for Bioorg. Med. Chem. Lett. Graphical Abstract New Approaches for the Treatment of Alzheimer’s Disease Paul V. Fish, David Steadman, Elliott D. Bayle, Paul Whiting 2 Digest for Bioorg. Med. Chem. Lett. Abstract Alzheimer’s disease (AD) is the most prevalent chronic neurodegenerative disease. Current approved therapies are symptomatic treatments having some effect on cognitive function. Therapies that target - amyloid (A) have been the focus of efforts to develop a disease modification treatment for AD but these approaches have failed to show any clinical benefit so far. Beyond the ‘A hypothesis’, there are a number of newer approaches to treat AD with neuroinflammation emerging as a very active area of research based on risk gene analysis. This short review will summarize approved drug therapies, recent clinical trials and new approaches for the treatment of AD. 3 Digest for Bioorg. Med. Chem. Lett. 1. Introduction Alzheimer’s disease (AD) is the most prevalent chronic neurodegenerative disease with 5.7 million people living with the disease in the USA alone and this is projected to increase to 13.8 million people by 2050.1 Globally, the number of people currently suffering with dementia is estimated to be 50 million of which 30-35 million have AD.2 The risk of developing the disease is influenced by both genetic and environmental factors, however the biggest risk factor by far is age; the older you are the more likely you are to develop the disease but it is not an inevitable part of ageing. For instance, about one in 50 people aged between 65 to 69 have dementia, and this figure rises to one in five for those aged between 85 to 89.2 Given the global increase in life expectancy, this represents a huge societal and economic challenge with the impact extending to those living with AD, along with their caregivers and family. The disease often manifests itself initially as short term memory loss and as the disease progresses symptoms include language problems, disorientation, mood swings and behavioural issues (agitation, sleep changes, psychosis). Eventually the disease progresses to loss of bodily function and ultimately to death. Aβ Ta u Figure 1: Postmortem tissue samples from Alzheimer’s disease (AD) patient brains reveal AD pathology including amyloid- (A) plaques and Tau tangles. Photographic image courtesy of Dr. Tammaryn Lashley at the Queen Square Brain Bank for Neurological Disorders (UCL Queen Square Institute of Neurology). Copyright Tammaryn Lashley 2018. There are two pathological hallmarks of the disease found in the brain of individuals with AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (Figure 1).3 These were first recognised in 1906 by Alois Alzheimer after whom the disease is named.3b We now recognise that the plaques are largely composed of -amyloid peptides (A) derived from amyloid precursor protein (APP), while the neurofibrillary tangles are composed of the tau protein (microtubule associated protein). As the disease advances, these pathologies progress along with brain shrinkage due to neuronal atrophy. (Figure 2). 4 Digest for Bioorg. Med. Chem. Lett. Figure 2: Comparison of MRI scans of healthy volunteer (left, control) and Alzheimer’s disease patient (right). The AD brain shows significant atrophy, dilation of the lateral ventricle and a smaller hippocampus. Images courtesy of Nick Fox (UCL Queen Square Institute of Neurology). Copyright Nick Fox. Human genetics has significantly enabled our understanding of the disease and the approaches that have been taken to discover new therapies. Rare autosomal dominant mutations in three genes, APP, presenilin 1 (PSEN1) and presenilin 2 (PSEN2) cause familial Alzheimer’s disease (FAD). FAD accounts for 2-3% of AD cases and usually has an earlier onset with symptoms developing in people in their 30s or 40s. In 1992, Hardy and Higgins postulated that the “deposition of amyloid protein (AβP) … is the causative agent of Alzheimer’s pathology and that neurofibrillary tangles, cell loss, vascular damage, and dementia follow as a direct result of this deposition”.4 Given that the APP protein is the source of A found in plaques, and that it subsequently became clear that the PSEN genes encode the catalytic subunit of the - secretase enzyme that cleaves APP (along with -secretase) leading to the generation of A peptides, this led to the molecular basis of the “-amyloid hypothesis of AD”.4 How A peptides (be that soluble, oligomeric, or plaque) leads to cell death remains controversial. Similarly, how tau tangles affect neural function, the relationship between -amyloid and tau tangles, and the apparent inter-neuronal spread of tau, are all areas of intense research. The recent clinical failures (discussed below) of a number of therapeutics that attempt to test the “- amyloid hypothesis of AD” might lead to the suggestion that this hypothesis has been disproven. However, it is our view that the A hypothesis is still central to causation of the disease and has yet to be adequately tested either because of flaws in the various therapeutics tested or because the clinical trials need to be performed on subjects much earlier in the disease process. Whether A or tau are the better drug target is unknown at this moment, and perhaps a moot point; both appear to be central to the disease process and so it is likely that combination therapies modulating both pathologies will ultimately be most successful. 5 Digest for Bioorg. Med. Chem. Lett. As well as the identification of autosomal dominant mutations, extensive genome wide association studies (GWAS) in AD have led to the identification of numerous risk alleles.5 The most significant of these is apolipoprotein E4 (ApoE4) with homozygosity leading to an 8 fold increase in risk of AD. However, despite a significant effort, the biology that underpins this increased risk conferred by ApoE4 remains to be fully understood. In the absence of this knowledge, it remains unclear if positive or negative modulation of ApoE4 would be beneficial. Furthermore, ApoE is a lipid-binding protein and is likely to be a challenging drug target for a small molecule even when there is an understanding as to the required modulation of function. A group of other genes identified through GWAS all point towards a role for neuroinflammation and microglial cells in AD. This is becoming an important area of research. Therapeutic approaches to AD, like most chronic degenerative diseases, can be broadly divided into three categories: symptomatic, disease modifying, and regenerative. Current approved therapies (cholinesterase inhibitors and glutamate antagonists) are symptomatic having some effect on cognitive function. There remains a significant unmet medical need for symptomatic treatments that impact more effectively the cognitive domain and also the other distressing symptoms such as agitation, psychosis and sleep disturbance.6 Most ongoing efforts to find new therapies are focussing on disease modification/disease progression; the premise is that if the onset of the disease can be delayed, or the progression of the disease slowed, then people will live longer without the developing AD. To date approaches have largely focussed around intervention in the A cascade and tau biology. The third category, regeneration, remains very much a hypothetical approach. Regenerating a diseased brain, replacing lost neurons and circuitry, is arguably a step too far for AD at this time. Clinical trials for AD have proven challenging, largely because the rate of disease progression is many months (meaning the trials often last 18 months or more), and the registerable endpoints recognised by the regulatory agencies are measures of cognitive performance.7 These are generally considered less robust an endpoint than biochemical or physiological measures. Good strides have however been made with pharmacodynamic endpoints. For instance, cerebrospinal fluid (CSF) A concentrations are routinely used as a biomarker for assessing compounds whose mechanism is inhibition of the enzymes that generate A from APP.8 Similarly, there are now good positron emission tomography (PET) ligands available that support visualisation and quantification of both A plaques and tau tangles. This is beginning to enable the correlation of the change in these classical pathological markers of the disease with disease progression as measured by the cognitive scales.9 For example florbetaben (1), flutemetamol (2) and florbetapir (3) are diagnostic radiotracers developed for routine clinical application to visualize A plaques in the brain. 18F-Flutemetamol (2, Vizamyl) is indicated for PET imaging of A neuritic plaque 6 Digest for Bioorg. Med. Chem. Lett. density in the brains of adult patients with cognitive impairment who are being evaluated for AD (Figure 3). florbetaben F-18 (1) flutemetamol F-18 (2) florbetapir F-18 (3) Figure 3: Axial view of negative (left) and positive (right) Vizamyl scans. Frontal (f), lateral temporal (it) and striatal (s) regions of the brain. Image taken from Vizamyl prescribing information, revised 02/2017. http://www.gevizamyl.com/us/wp-content/uploads/2017/02/43-1067C-Vizamyl1.pdf. Copyright GE Healthcare. 2. Approved Therapies There are currently no approved therapies which directly target AD pathology. Existing treatments are symptomatic, aimed at ameliorating cognitive function through two different modes of action: agonism of the cholinergic system and antagonism of the N-methyl-D-aspartate receptor (NMDA-receptor).
Recommended publications
  • Upcoming Market Catalysts in Q4 2017
    NEWS & ANALYSIS BIOBUSINESS BRIEFS MARKET WATCH Upcoming market catalysts in Q4 2017 Market catalysts in the fourth quarter of disease, and further results from a pivotal trial 2017 include top-line clinical trial results in Alzheimer disease are anticipated in late for tenapanor (developed by Ardelyx) for September. In light of the failure of other constipation-predominant irritable bowel 5HT6 receptor antagonists — including the syndrome (IBS-C) and intepirdine (developed recent failure of Lundbeck’s idalopirdine in by Axovant Sciences) for the treatment of several phase III trials in Alzheimer disease dementia with Lewy bodies, as well as the — the upcoming phase III data in Alzheimer expected approval of axicabtagene disease, along with the phase II data in ciloleucel (developed by Kite Pharma) in the dementia, will give important guidance United States for the treatment of relapsed/ to the future market opportunities for refractory aggressive B cell non-Hodgkin intepirdine. lymphoma (NHL). The Biologics License Application for Early in the quarter, Ardelyx expects Kite Pharma’s axicabtagene ciloleucel for the top-line results from T3MPO-2, a 6-month treatment of relapsed/refractory aggressive phase III study of tenapanor. This orally B cell NHL is under priority review, with a administered small molecule inhibits the Prescription Drug User Fee Act target action sodium–proton exchanger NHE3 in the date of 29 November 2017. This gastrointestinal tract, which increases the FDA-designated breakthrough therapy amount of sodium and fluid in the gut, consists of a patient’s peripheral blood thereby loosening stools. Results from a T lymphocytes that have been genetically previous 12-week phase III study known as engineered in vitro with chimeric antigen T3MPO-1 were statistically significant for receptors (CAR), enabling them to recognize the primary composite end point of response the tumour-expressed molecule CD19 rate, and seven out of eight secondary end after infusion back into the patient.
    [Show full text]
  • Shexiang Baoxin Pill, a Traditional Chinese Herbal Formula, Rescues the Cognitive Impairments in APP/ PS1 Transgenic Mice
    ORIGINAL RESEARCH published: 14 July 2020 doi: 10.3389/fphar.2020.01045 Shexiang Baoxin Pill, a Traditional Chinese Herbal Formula, Rescues the Cognitive Impairments in APP/ PS1 Transgenic Mice † † Wei-Hui Hu 1,2,3 , Shing-Hung Mak 1,2 , Zhong-Yu Zheng 1,2, Ying-Jie Xia 1,2, Miranda Li Xu 1,2, Ran Duan 1,2,3, Tina Ting-Xia Dong 1,2,3, Shao-Ping Li 4, Chang-Sen Zhan 5,6, Xiao-Hui Shang 5,6 and Karl Wah-Keung Tsim 1,2,3* 1 Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Shenzhen, China, 2 Division of Life Science and Center for Chinese Medicine and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Hong Kong, Hong Kong, 3 Joint Laboratory of Guangdong Province and Hong Kong Region on Marine Bioresource Conservation and Exploitation, College of Marine Sciences, South China Agricultural University, Guangzhou, China, 4 Institute of Chinese Medical Sciences, University of Macau, Macau, Macau, 5 Shanghai Edited by: Engineering Research Center for Innovation of Solid Preparation of TCM, Shanghai, China, 6 Shanghai Hutchison Qiaobing Huang, Pharmaceuticals Ltd., Shanghai, China Southern Medical University, China Reviewed by: Background: Shexiang Baoxin Pill (SBP), a formulated traditional Chinese medicine Bing-Xing Pan, Nanchang University, China (TCM), has been widely used to treat cardiovascular diseases for years. This herbal Wenda Xue, mixture has been shown to promote differentiation of cultured neuronal cells. Here, we Nanjing University of Chinese aimed to investigate the effects of SBP in attenuating cognitive impairment in APP/PS1 Medicine, China *Correspondence: transgenic mice.
    [Show full text]
  • The 1,3,5-Triazine Derivatives As Innovative Chemical Family of 5-HT6 Serotonin Receptor Agents with Therapeutic Perspectives for Cognitive Impairment
    International Journal of Molecular Sciences Article The 1,3,5-Triazine Derivatives as Innovative Chemical Family of 5-HT6 Serotonin Receptor Agents with Therapeutic Perspectives for Cognitive Impairment Gniewomir Latacz 1 , Annamaria Lubelska 1, Magdalena Jastrz˛ebska-Wi˛esek 2, Anna Partyka 2, Małgorzata Anna Mar´c 1, Grzegorz Satała 3, Daria Wilczy ´nska 2, Magdalena Kota ´nska 4, Małgorzata Wi˛ecek 1, Katarzyna Kami ´nska 1, Anna Wesołowska 2, Katarzyna Kie´c-Kononowicz 1 and Jadwiga Handzlik 1,* 1 Department of Technology and Biotechnology of Drugs, Medical College, Jagiellonian University, Medyczna 9, PL 30-688 Cracow, Poland 2 Department of Clinical Pharmacy, Medical College, Jagiellonian University, Medyczna 9, PL 30-688 Cracow, Poland 3 Department of Medicinal Chemistry Institute of Pharmacology, Polish Academy of Science, Sm˛etna12, PL 31-343 Cracow, Poland 4 Department of Pharmacodynamics, Faculty of Pharmacy, Medical College, Jagiellonian University, Medyczna 9, PL 30-688 Cracow, Poland * Correspondence: [email protected]; Tel.: +48-12-620-5580 Received: 27 May 2019; Accepted: 7 July 2019; Published: 12 July 2019 Abstract: Among serotonin receptors, the 5-HT6 subtype is the most controversial and the least known in the field of molecular mechanisms. The 5-HT6R ligands can be pivotal for innovative treatment of cognitive impairment, but none has reached pharmacological market, predominantly, due to insufficient “druglikeness” properties. Recently, 1,3,5-triazine-piperazine derivatives were identified as a new chemical family of potent 5-HT6R ligands. For the most active triazine 5-HT6R agents found (1–4), a wider binding profile and comprehensive in vitro evaluation of their drug-like parameters as well as behavioral studies and an influence on body mass in vivo were investigated within this work.
    [Show full text]
  • In Early Alzheimer's Disease De
    Protocol I8D-MC-AZFD(a) A Randomized, Double-Blind, Delayed-Start Study of LY3314814 (AZD3293) in Early Alzheimer’s Disease Dementia (Extension of Study AZES, The AMARANTH Study) NCT02972658 Approval Date: 06-Feb-2018 I8D-MC-AZFD(a) Clinical Protocol Page 1 Protocol I8D-MC-AZFD(a) A Randomized, Double-Blind, Delayed-Start Study of LY3314814 (AZD3293) in Early Alzheimer’s Disease Dementia (Extension of Study AZES, The AMARANTH Study) Confidential Information The information contained in this document is confidential and is intended for the use of clinical investigators. It is the property of Eli Lilly and Company or its subsidiaries and should not be copied by or distributed to persons not involved in the clinical investigation of LY3314814, unless such persons are bound by a confidentiality agreement with Eli Lilly and Company or its subsidiaries. Note to Regulatory Authorities: This document may contain protected personal data and/or commercially confidential information exempt from public disclosure. Eli Lilly and Company requests consultation regarding release/redaction prior to any public release. In the United States, this document is subject to Freedom of Information Act (FOIA) Exemption 4 and may not be reproduced or otherwise disseminated without the written approval of Eli Lilly and Company or its subsidiaries. LY3314814 (Lanabecestat) Study AZFD is a Phase 3 study designed to test whether LY3314814 will slow disease progression in patients with early Alzheimer’s Disease randomized in Study AZES. Eli Lilly and Company Indianapolis, Indiana USA 46285 Protocol Electronically Signed and Approved by Lilly on date provided below. Approval Date: 06-Feb-2018 GMT LY3314814 I8D-MC-AZFD(a) Clinical Protocol Page 2 Table of Contents Section Page 1.
    [Show full text]
  • The “Rights” of Precision Drug Development for Alzheimer's Disease
    Cummings et al. Alzheimer's Research & Therapy (2019) 11:76 https://doi.org/10.1186/s13195-019-0529-5 REVIEW Open Access The “rights” of precision drug development for Alzheimer’s disease Jeffrey Cummings1*, Howard H. Feldman2 and Philip Scheltens3 Abstract There is a high rate of failure in Alzheimer’s disease (AD) drug development with 99% of trials showing no drug- placebo difference. This low rate of success delays new treatments for patients and discourages investment in AD drug development. Studies across drug development programs in multiple disorders have identified important strategies for decreasing the risk and increasing the likelihood of success in drug development programs. These experiences provide guidance for the optimization of AD drug development. The “rights” of AD drug development include the right target, right drug, right biomarker, right participant, and right trial. The right target identifies the appropriate biologic process for an AD therapeutic intervention. The right drug must have well-understood pharmacokinetic and pharmacodynamic features, ability to penetrate the blood-brain barrier, efficacy demonstrated in animals, maximum tolerated dose established in phase I, and acceptable toxicity. The right biomarkers include participant selection biomarkers, target engagement biomarkers, biomarkers supportive of disease modification, and biomarkers for side effect monitoring. The right participant hinges on the identification of the phase of AD (preclinical, prodromal, dementia). Severity of disease and drug mechanism both have a role in defining the right participant. The right trial is a well-conducted trial with appropriate clinical and biomarker outcomes collected over an appropriate period of time, powered to detect a clinically meaningful drug-placebo difference, and anticipating variability introduced by globalization.
    [Show full text]
  • Treatment of Alzheimer's Disease and Blood–Brain Barrier Drug Delivery
    pharmaceuticals Review Treatment of Alzheimer’s Disease and Blood–Brain Barrier Drug Delivery William M. Pardridge Department of Medicine, University of California, Los Angeles, CA 90024, USA; [email protected] Received: 24 October 2020; Accepted: 13 November 2020; Published: 16 November 2020 Abstract: Despite the enormity of the societal and health burdens caused by Alzheimer’s disease (AD), there have been no FDA approvals for new therapeutics for AD since 2003. This profound lack of progress in treatment of AD is due to dual problems, both related to the blood–brain barrier (BBB). First, 98% of small molecule drugs do not cross the BBB, and ~100% of biologic drugs do not cross the BBB, so BBB drug delivery technology is needed in AD drug development. Second, the pharmaceutical industry has not developed BBB drug delivery technology, which would enable industry to invent new therapeutics for AD that actually penetrate into brain parenchyma from blood. In 2020, less than 1% of all AD drug development projects use a BBB drug delivery technology. The pathogenesis of AD involves chronic neuro-inflammation, the progressive deposition of insoluble amyloid-beta or tau aggregates, and neural degeneration. New drugs that both attack these multiple sites in AD, and that have been coupled with BBB drug delivery technology, can lead to new and effective treatments of this serious disorder. Keywords: blood–brain barrier; brain drug delivery; drug targeting; endothelium; Alzheimer’s disease; therapeutic antibodies; neurotrophins; TNF inhibitors 1. Introduction Alzheimer’s Disease (AD) afflicts over 50 million people world-wide, and this health burden costs over 1% of global GDP [1].
    [Show full text]
  • Current Research Efforts in the Prevention and Treatment of Alzheimer’S Disease (AD) and Related Dementias
    Current Research efforts in the prevention and treatment of Alzheimer’s Disease (AD) and Related Dementias What’s new? 1. New and More effective Biomarkers 2. New Diagnostic Framework : • New “A/T/N” Framework • New Framework conceptualizes AD as a continuum from pathophysiological, biomarker and clinical perspectives. 3. LATE (Limbic-predominant, Age-related, TDP-43 Encephalopathy) looks like Alzheimer’s disease 4. Alzheimer’s pathophysiology starts decades prior to clinical disease! 1. New and More Effective Biomarkers AD Pathology • Alzheimer’s Disease consists of two main features: • Senile plaques (β-amyloid)- earliest pathological event! • Neurofibrillary tangles (hyperphosphorylated tau)- primary culprit in cells death • Mechanism: • β-amyloid hyperphosphorylated tau tau spreads throughout the brain from neuron to neuron (Pooler et al, 2013). Cell Death Biomarkers Budson & Solomon, Practical Neurology 2012;12:88–96 PET Amyloid Imaging was approved since 2012 • Use when knowing that AD pathology is present in symptomatic patient would change management. • May detect amyloid plaques in asymptomatic patients who may not develop disease for 10-15+ years • Not paid for by Medicare or other insurance companies • Can obtain through Veterans Affairs hospitals, clinical trials/research studies, and self-pay. • Will have broader use when disease modifying therapies are available. Alzheimer’s Disease Non-AD dementia 65 year old MoCA 21 From Budson & Solomon, 2016 PET tau imaging : FDA approved May 2020 • Use when knowing that AD pathology is present in symptomatic patient would change management. • Will detect AD tau tangles in symptomatic patients and should correlate with symptoms • May detect other types of tau tangles in other dementias (not yet clear) • Not paid for by Medicare or other insurance companies • Can obtain through Veterans Affairs hospitals, clinical trials/research studies, and self-pay.
    [Show full text]
  • Natural Antioxidant Anthocyanins—A Hidden Therapeutic Candidate in Metabolic Disorders with Major Focus in Neurodegeneration
    Review Natural Antioxidant Anthocyanins—A Hidden Therapeutic Candidate in Metabolic Disorders with Major Focus in Neurodegeneration Rahat Ullah 1,†, Mehtab Khan 1,†, Shahid Ali Shah 1,2, Kamran Saeed 1 and Myeong Ok Kim 1,* 1 Division of Applied Life Science (BK 21), College of Natural Sciences, Gyeongsang National University, Jinju 52828, Korea; [email protected] (R.U.); [email protected] (M.K.); [email protected] (S.A.S.); [email protected] (K.S.) 2 Department of Chemistry, Sarhad University of Science & Information Technology (SUIT), Peshawar Khyber Pakhtunkhwa 25000, Pakistan * Correspondence: [email protected]; Tel.: +82-55-772-1345; Fax: +82-55-772-2656 † These authors contributed equally to this paper. Received: 7 May 2019; Accepted: 24 May 2019; Published: 28 May 2019 Abstract: All over the world, metabolic syndrome constitutes severe health problems. Multiple factors have been reported in the pathogenesis of metabolic syndrome. Metabolic disorders result in reactive oxygen species (ROS) induced oxidative stress, playing a vital role in the development and pathogenesis of major health issues, including neurological disorders Alzheimer’s disease (AD) Parkinson's disease (PD). Considerable increasing evidence indicates the substantial contribution of ROS-induced oxidative stress in neurodegenerative diseases. An imbalanced metabolism results in a defective antioxidant defense system, free radicals causing inflammation, cellular apoptosis, and tissue damage. Due to the annual increase in financial and social burdens, in addition to the adverse effects associated with available synthetic agents, treatment diversion from synthetic to natural approaches has occurred. Antioxidants are now being considered as convincing therapeutic agents against various neurodegenerative disorders.
    [Show full text]
  • Drug Candidates in Clinical Trials for Alzheimer's Disease
    Hung and Fu Journal of Biomedical Science (2017) 24:47 DOI 10.1186/s12929-017-0355-7 REVIEW Open Access Drug candidates in clinical trials for Alzheimer’s disease Shih-Ya Hung1,2 and Wen-Mei Fu3* Abstract Alzheimer’s disease (AD) is a major form of senile dementia, characterized by progressive memory and neuronal loss combined with cognitive impairment. AD is the most common neurodegenerative disease worldwide, affecting one-fifth of those aged over 85 years. Recent therapeutic approaches have been strongly influenced by five neuropathological hallmarks of AD: acetylcholine deficiency, glutamate excitotoxicity, extracellular deposition of amyloid-β (Aβ plague), formation of intraneuronal neurofibrillary tangles (NTFs), and neuroinflammation. The lowered concentrations of acetylcholine (ACh) in AD result in a progressive and significant loss of cognitive and behavioral function. Current AD medications, memantine and acetylcholinesterase inhibitors (AChEIs) alleviate some of these symptoms by enhancing cholinergic signaling, but they are not curative. Since 2003, no new drugs have been approved for the treatment of AD. This article focuses on the current research in clinical trials targeting the neuropathological findings of AD including acetylcholine response, glutamate transmission, Aβ clearance, tau protein deposits, and neuroinflammation. These investigations include acetylcholinesterase inhibitors, agonists and antagonists of neurotransmitter receptors, β-secretase (BACE) or γ-secretase inhibitors, vaccines or antibodies targeting Aβ clearance or tau protein, as well as anti-inflammation compounds. Ongoing Phase III clinical trials via passive immunotherapy against Aβ peptides (crenezumab, gantenerumab, and aducanumab) seem to be promising. Using small molecules blocking 5-HT6 serotonin receptor (intepirdine), inhibiting BACE activity (E2609, AZD3293, and verubecestat), or reducing tau aggregation (TRx0237) are also currently in Phase III clinical trials.
    [Show full text]
  • 2017 Medicines in Development for Alzheimer's Disease
    2017 Medicines in Development for Alzheimer's Disease Alzheimer's Disease Product Name Sponsor Indication Development Phase ABBV-8E12 AbbVie Alzheimer's disease Phase II (anti-tau antibody) North Chicago, IL www.abbvie.com AC-1204 Accera mild to moderate Alzheimer's disease Phase III (glucose stimulant) Broomfield, CO www.accerapharma.com ACI-24 AC Immune Alzheimer's disease in Down Phase I (anti-Abeta vaccine) Lausanne, Switzerland syndrome patients www.acimmune.com ACI-35 AC Immune mild to moderate Alzheimer's disease Phase I (anti-pTau vaccine) Lausanne, Switzerland www.acimmune.com Janssen Research & Development www.janssen.com Raritan, NJ aducanumab (BIIB037) Biogen Alzheimer's disease (Fast Track) Phase III (amyloid beta mAb) Cambridge, MA www.biogen.com Neurimmune Zurich, Switzerland AGB101 AgenBio amnestic mild cognitive impairment Phase II completed (levetiracetam low-dose) Baltimore, MD in Alzheimer's disease www.agenebio.com Medicines in Development: Alzheimer's Disease | 2017 Update 1 Alzheimer's Disease Product Name Sponsor Indication Development Phase ALZ-801 Alzheon mild Alzheimer's disease Phase II (amyloid beta-protein inhibitor) Framingham, MA (homozygous APOE4/4 genotype) www.alzheon.com mild Alzheimer's disease Phase II (heterozygous APOE4 genotype) www.alzheon.com ALZT-OP1 AZTherapies Alzheimer's disease Phase III (amyloid beta-protein inhibitor/ Boston, MA www.aztherapies.com inflammation mediator inhibitor) AMG520/CNP520 Amgen Alzheimer's disease (Fast Track) Phase II/III (BACE1 protein inhibitor) Thousand Oaks,
    [Show full text]
  • UNITED STATES SECURITIES and EXCHANGE COMMISSION Washington, D.C
    UNITED STATES SECURITIES AND EXCHANGE COMMISSION Washington, D.C. 20549 FORM 8-K CURRENT REPORT Pursuant to Section 13 or 15(d) of the Securities Exchange Act of 1934 Date of Report (Date of Earliest Event Reported): January 8, 2018 Axovant Sciences Ltd. (Exact Name of Registrant as specified in its charter) Bermuda 001-37418 98-1333697 (State or Other Jurisdiction (Commission (IRS Employer of Incorporation) File Number) Identification No.) Suite 1, 3rd Floor 11-12 St. James’s Square London SW1Y 4LB, United Kingdom (Address of principal executive offices) Registrant’s telephone number, including area code: +44 203 318 9708 Not Applicable (Registrant’s name or former address, if change since last report) Check the appropriate box below if the Form 8-K filing is intended to simultaneously satisfy the filing obligation of the registrant under any of the following provisions: o Written communications pursuant to Rule 425 under the Securities Act (17 CFR 230.425) o Soliciting material pursuant to Rule 14a-12 under the Exchange Act (17 CFR 240.14a-12) o Pre-commencement communications pursuant to Rule 14d-2(b) under the Exchange Act (17 CFR 240.14d-2(b)) o Pre-commencement communications pursuant to Rule 13e-4(c) under the Exchange Act (17 CFR 240.13e-4(c)) Indicate by check mark whether the registrant is an emerging growth company as defined in Rule 405 of the Securities Act of 1933 (§230.405 of this chapter) or Rule 12b-2 of the Securities Exchange Act of 1934 (§240.12b-2 of this chapter). Emerging growth company x If an emerging growth company, indicate by check mark if the registrant has elected not to use the extended transition period for complying with any new or revised financial accounting standards provided pursuant to Section 13(a) of the Exchange Act.
    [Show full text]
  • A New Analysis of the Phase II Alzheimer's
    Will the Next Five Years Witness an Innovation Wave in Medicines for Alzheimer’s? The Healthcare System Is Not Ready for New Alzheimer’s Drugs Success in drug development for Alzheimer’s disease has been elusive. Recent analysis has shown a near 100% failure rate in the past decade. Yet, each failed trial advances the science and presents the opportunity to apply new insights. As a result, there are a significant number of compounds in Phase 3, the final stage of drug development. Seventeen drugs in this final stage are likely to finish testing and could be available to people with the disease in the next five years. These findings are based on the analysis led by ResearchersAgainstAlzhei- mer’s, a global network of leading Alzheimer’s researchers at top academic institutions and corporations. Our analysis leads us to be cautiously optimistic that innovative treatments are within our reach. We need to make sure the healthcare system is ready. Understanding the Alzheimer’s Drug Development Pipeline Robust Alzheimer’s Phase 3 Drugs in Development That Could Launch in the Next Five Years Pipeline Offers Promise for Treatment – Despite 2016 2017 2018 2019 2020 2021 Recent Track Record Brexpiprazole Otsuka Pharmaceuticals, H. Lundbeck Aripiprazole Otsuka Pharmaceuticals Solanezumab Eli Lilly MORE THAN A DECADE SINCE A NOVEL Masitinib ALZHEIMER’S DRUG WAS APPROVED AB Science Namenda was approved by TRx0237 the FDA in 2003, marking the last TauRx Therapeutics time a novel Alzheimer’s therapy Idalopirdine reached the market.1 Otsuka Pharmaceuticals, H. Lundbeck RVT-101 Axovant Sodium Oligo Shanghai Greenvalley Pharmaceuticals Azeliragon vTv Therapeutics PIPELINE OUTLOOK Nilvadipine There are approximately 50 drugs Astellas Pharma, Archer Pharmaceuticals in Phase 2 trials and about a dozen 2 ALZT-0P1 drugs in Phase 2/3 trials.
    [Show full text]