Current and Future Treatments in Alzheimer's Disease

Total Page:16

File Type:pdf, Size:1020Kb

Current and Future Treatments in Alzheimer's Disease 227 Current and Future Treatments in Alzheimer’sDisease Alireza Atri, MD, PhD1,2 1 Banner Sun Health Research Institute, Banner Health, Sun City, Address for correspondence Alireza Atri, MD, PhD, Banner Sun Health Arizona Research Institute, 10515 W Santa Fe Drive, Sun City, AZ 85351 2 Center for Brain/Mind Medicine, Department of Neurology, Brigham (e-mail: [email protected]). and Women’s Hospital, Harvard Medical School, Boston, Massachusetts Semin Neurol 2019;39:227–240. Abstract The foundation of current Alzheimer’s disease (AD) treatment involves pharmacolo- gical and nonpharmacological management and care planning predicated on patient- centered psychoeducation, shared goal-setting, and decision-making forged by a strong triadic relationship between clinician and the patient-caregiver dyad. Food and Drug Administration (FDA) approved AD medications, cholinesterase-inhibitors (ChEIs), and the N-methyl-d-aspartate (NMDA) antagonist memantine, when utilized as part of a comprehensive care plan, while generally considered symptomatic medica- tions, can provide modest “disease course-modifying” effects by enhancing cognition, and reducing loss of independence. When combined, pharmacologic and nonpharma- cologic treatments can meaningfully mitigate symptoms and reduce clinical progres- sion and care burden. AD pharmacotherapy first involves identification and elimination of potentially harmful medications and supplements. First line treatment for neurop- sychiatric symptoms and problem behaviors is nonpharmacological and involves psychoeducation, trigger identification, and implementation, iterative evaluation, and adjustment of behavioral and environmental interventions. Intensive research efforts are underway to develop more accurate and practical AD diagnostic biomarkers Keywords and clinical tools and better therapeutics. Ongoing research studies for primary ► cognitive impairment and secondary prevention of AD and clinical trials evaluating symptomatic and ► dementia disease-modifying treatments in symptomatic AD are directed at diverse therapeutic ► cholinesterase- targets including neurochemicals, amyloid and tau pathological processes, mitochon- inhibitor dria, inflammatory pathways, neuroglia, and multimodal lifestyle interventions. Alzheimer’s disease (AD), the most common cause of mid-to- The current treatment paradigm in Alzheimer’s disease late life cognitive impairment and dementia, is pathologi- (AD) involves a multipronged approach of combining pharma- cally defined by deposits of amyloid-β (Aβ42)-plaques and cological and nonpharmacological approaches to mitigate hyperphosphorylated-tau (p-tau) tangles. Complex, inter- progressive loss of cognitive and functional abilities. No new This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited. acting, and incompletely understood biopsychosocial and AD medications have been approved by Food and Drug Admin- pathological processes lead to heterogeneous AD clinical istration (FDA) since 2002 (other than a capsule preparation phenotypes (i.e., cognitive-behavioral syndromes related to that combines two available medications). However, over the AD). AD biopathological processes develop over one to two last decade multiple lines of evidence from randomized, “preclinical” decades before symptoms manifest insidiously; double-blind, and placebo-controlled trials (RCT), and pro- this preclinical period is increasingly targeted in research spective long-term observational cohort studies have emerged and may represent the best opportunities to potentially to support the clinical effectiveness of AD medications, in delay or prevent onset of dementia stages of AD.1,2 mono-, or add-on-dual combination therapy to at least Issue Theme Dementia; Guest Editor, Copyright © 2019 by Thieme Medical DOI https://doi.org/ Arash Salardini, MD Publishers, Inc., 333 Seventh Avenue, 10.1055/s-0039-1678581. New York, NY 10001, USA. ISSN 0271-8235. Tel: +1(212) 584-4662. 228 Current and Future Treatments in Alzheimer’sDisease Atri Table 1 Risk factors for AD dementia Modifiable Nonmodifiable (in early life, midlife ages 45–65 y, or later life ages > 65 y) Vascular risks: Age Diabetes (later life) Gender (female > male) Hypertension (midlife) Family history (first- or second-degree relative or multiple generations) Dyslipidemia (midlife) Race Metabolic Syndrome and obesity (midlife) Down’sSyndrome Smoking tobacco (later life) ApoE-ε4 allele Low physical activity (later life) Cerebral amyloidosis–abiomarkeroftheADpathologicalprocessa Cerebral hypoperfusion Cerebrovascular injury or stroke Depression (later life) Severe head trauma or traumatic brain injury Hearing loss (midlife) Low cognitive reserve: low education, intelligence, professional or social attainment/occupation (early life and potentially midlife) Low social contact (later life) Abbreviations: AD, Alzheimer’s disease; ApoE, Apolipoprotein E. aIt is currently a nonmodifiable risk factor but primary and secondary prevention trials utilizing amyloid-modifying drugs are in progress and early results support that cerebral amyloid plaque burden can be lowered; whether this translates to clinical benefit or of lowering risk of progression to MCI or dementia, potentially making this be a modifiable risk factor, is to be determined. modestly (with small to medium effect sizes) mitigate symp- cellular, network, and systems. The current AD treatment toms and retard the expected trajectory of progressive paradigm is one of multifaceted management of symptoms decline.3 aimed at retaining quality of life, mitigating the burden of The last decade has brought several major advances illness and reducing long-term clinical decline. Successful involving explication of AD risk factors (see ►Table 1)and long-term pharmacotherapy with FDA-approved AD medica- AD diagnostics. In vivo AD biomarkers of the pathological tions, initially involving monotherapy with a cholinesterase- process are now available in the clinic. Measurements of inhibitor (ChEI) and ultimately involving add-on dual-combi- amyloid-Beta42 (Aβ42), tau (τ), and phospho-tau (p-tau) nation treatment with a ChEI and memantine, requires devel- proteins in cerebrospinal fluid (CSF), as well as Aβ42-plaque oping, implementing and sustaining a solid foundation of burden on amyloid-PET (positron emission tomography), can psychoeducation, nonpharmacological and behavioral care now aid clinical, as well are research, diagnosis of AD with a strategies, and clarity in care goals and expectations; this is high level of certainty. Though breakthroughs to affect newer predicated on a strong therapeutic alliance between the AD therapeutics have yet to materialize, learning from clinician and the patient–caregiver dyad. dozens of large and varied clinical trial programs have provided cornerstones of ongoing intensive research efforts Management of AD that aim at primary and secondary prevention, as well as ►Table 2 broadly outlines the multifactorial tailored man- symptomatic and disease-modifying treatments for AD. agement of AD. Evaluation and management of AD requires a This paper will (1) review current multifactorial manage- solid foundation of open, honest, and compassionate com- ment of AD, with a focus on practical aspects of psychoedu- munication; shared goal setting; and a triadic partnership cation and behavioral approaches, clinical evidence, and between the clinician, patient and care-partner(s). Evalua- This document was downloaded for personal use only. Unauthorized distribution is strictly prohibited. considerations for treatment with FDA-approved AD medi- tion and management should be a patient–caregiver dyad- cations; and (2) briefly summarize experimental pharmaco- centered and dynamic processes that involve multipronged logical approaches under research investigation that are and, ideally, integrated interdisciplinary team approaches. likely to form the bases of future combination therapies in Treatment of AD is not curative and successful management the AD clinical spectrum. to mitigate the burden of illness relies on four basic pillars: (1) timely and accurate syndromic and etiological diagnoses Current Treatments in AD combined with proactive educational and care planning that are customized to the patient–caregiver dyad (the dyad)– There is no cure for AD dementia. It is increasingly recognized without accurate diagnosis and tailored psychoeducation, that the complex and incompletely understood pathological appropriate dyad-centered treatment, and care can’tbe processes culminating in AD dementia can begin decades prior provided; (2) nonpharmacological interventions and beha- to the manifestation of clinical symptoms, by which time vioral approaches; (3) pharmacological interventions; and extensive and likely irreversible processes have wrought wide- (4) holistic care planning that is proactive, pragmatic and spread destruction on multiple levels: molecular, intracellular, dynamic, and includes monitoring and adjustment of the Seminars in Neurology Vol. 39 No. 2/2019 Current and Future Treatments in Alzheimer’sDisease Atri 229 Table 2 Key Elements of effective multifactorial management of AD Patient-caregiver dyad-centered evaluation, diagnosis and disclosure, and care planning processes • Timely detection of symptoms, accurate assessment and diagnosis, iterative psychoeduction, and appropriate disclosure. • Shared goal setting for diagnostic, disclosure and management processes; sustained psychoeducation and tailoring of a proactive
Recommended publications
  • Annual Report
    Zentralinstitut für Seelische Gesundheit Landesstiftung des öffentlichen Rechts 2017 2019 ANNUAL REPORT 2017 2019 ANNUAL REPORT EXECUTIVE BOARD RESEARCH REPORT BY THE EXECUTIVE BOARD, DEPARTMENTS, INSTITUTES DEVELOPMENT FIGURES AND RESEARCH GROUPS Report by the Executive Board 6 The Future of Therapy Research 42 Development Figures 8 Department of Neuropeptide Research 46 in Psychiatry Department of Molecular Neuroimaging 47 Department of Public Mental Health 48 Hector Institute for Translational 50 Brain Research RG Developmental Brain Pathologies 51 Department of Biostatistics 52 PATIENT CARE Institute of Cognitive and 53 Clinical Neuroscience CLINICAL DEPARTMENTS AND INSTITUTES RG Brain Stimulation, Neuroplasticity and 54 Learning RG Psychobiology of Risk Behavior 54 Clinic of Psychiatry and Psychotherapy 12 RG Body Plasticity and Memory Processes 55 Clinic of Child and Adolescent Psychiatry and 20 RG Psychobiology of Pain 56 Psychotherapy RG Psychobiology of Emotional Learning 57 Clinic of Psychosomatic Medicine and 24 Institute for Psychopharmacology 58 Psychotherapy RG Behavioral Genetics 59 RG Translational Addiction Research 60 Clinic of Addictive Behavior and 26 RG Physiology of Neuronal Networks 61 Addiction Medicine RG Molecular Psychopharmacology 62 Adolescent Center for Disorders 29 RG Neuroanatomy 63 of Emotional Regulation RG In Silico Psychopharmacology 64 Adolescent Center for 30 Institute for Psychiatric and 65 Psychotic Disorders – SOTERIA Psychosomatic Psychotherapy RG Experimental Psychotherapy 66 Central Outpatient
    [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]
  • La Place Des Composés Multi Target Directed Ligands Dans Le Traitement De La Maladie D’Alzheimer Katia Hamidouche
    La place des composés Multi Target Directed Ligands dans le traitement de la maladie d’Alzheimer Katia Hamidouche To cite this version: Katia Hamidouche. La place des composés Multi Target Directed Ligands dans le traitement de la maladie d’Alzheimer. Sciences pharmaceutiques. 2017. dumas-01556379 HAL Id: dumas-01556379 https://dumas.ccsd.cnrs.fr/dumas-01556379 Submitted on 5 Jul 2017 HAL is a multi-disciplinary open access L’archive ouverte pluridisciplinaire HAL, est archive for the deposit and dissemination of sci- destinée au dépôt et à la diffusion de documents entific research documents, whether they are pub- scientifiques de niveau recherche, publiés ou non, lished or not. The documents may come from émanant des établissements d’enseignement et de teaching and research institutions in France or recherche français ou étrangers, des laboratoires abroad, or from public or private research centers. publics ou privés. UNIVERSITE DE CAEN NORMANDIE ANNEE 2017 U.F.R. DES SCIENCES PHARMACEUTIQUES THESE POUR LE DIPLOME D’ETAT DE DOCTEUR EN PHARMACIE PRESENTEE PAR Katia HAMIDOUCHE SUJET : La place des composés "Multi Target Directed Ligands" dans le traitement de la maladie d'Alzheimer SOUTENUE PUBLIQUEMENT LE : 31/03/2017 JURY : Pr. Michel Boulouard PRESIDENT DU JURY Dr. Véronique Lelong Boulouard EXAMINATEUR Dr. Joanna Bourgine EXAMINATEUR Pr. Thomas Freret Remerciements Avant tout, je tiens à dédier ce travail à mes parents , que je remercie également profondément pour leurs longs encouragements et soutien, et à qui je présente toute ma reconnaissance et gratitude pour les sacrifices qu’ils ont choisis de faire afin de nous permettre, ma sœur, mes frères et moi -même de faire ces grandes études, et sans lesquels je n’aurai jamais découvert cet univers de savoir et de science « à la Française ».
    [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]
  • Copyrighted Material
    Index Note: page numbers in italics refer to figures; those in bold to tables or boxes. abacavir 686 tolerability 536–537 children and adolescents 461 acamprosate vascular dementia 549 haematological 798, 805–807 alcohol dependence 397, 397, 402–403 see also donepezil; galantamine; hepatic impairment 636 eating disorders 669 rivastigmine HIV infection 680 re‐starting after non‐adherence 795 acetylcysteine (N‐acetylcysteine) learning disability 700 ACE inhibitors see angiotensin‐converting autism spectrum disorders 505 medication adherence and 788, 790 enzyme inhibitors obsessive compulsive disorder 364 Naranjo probability scale 811, 812 acetaldehyde 753 refractory schizophrenia 163 older people 525 acetaminophen, in dementia 564, 571 acetyl‐L‐carnitine 159 psychiatric see psychiatric adverse effects acetylcholinesterase (AChE) 529 activated partial thromboplastin time 805 renal impairment 647 acetylcholinesterase (AChE) acute intoxication see intoxication, acute see also teratogenicity inhibitors 529–543, 530–531 acute kidney injury 647 affective disorders adverse effects 537–538, 539 acutely disturbed behaviour 54–64 caffeine consumption 762 Alzheimer’s disease 529–543, 544, 576 intoxication with street drugs 56, 450 non‐psychotropics causing 808, atrial fibrillation 720 rapid tranquillisation 54–59 809, 810 clinical guidelines 544, 551, 551 acute mania see mania, acute stupor 107, 108, 109 combination therapy 536 addictions 385–457 see also bipolar disorder; depression; delirium 675 S‐adenosyl‐l‐methionine 275 mania dosing 535 ADHD
    [Show full text]
  • A [18F]Fluoroethoxybenzovesamicol Positron Emission Tomography Study
    Received: 24 May 2018 Revised: 7 September 2018 Accepted: 10 September 2018 DOI: 10.1002/cne.24541 RESEARCH ARTICLE Regional vesicular acetylcholine transporter distribution in human brain: A [18F]fluoroethoxybenzovesamicol positron emission tomography study Roger L. Albin1,2,3,4 | Nicolaas I. Bohnen1,2,3,5 | Martijn L. T. M. Muller3,5 | William T. Dauer1,2,3,6 | Martin Sarter3,7 | Kirk A. Frey2,5 | Robert A. Koeppe3,5 1Neurology Service & GRECC, VAAAHS, Ann Arbor, Michigan Abstract 2Department of Neurology, University of Prior efforts to image cholinergic projections in human brain in vivo had significant technical lim- Michigan, Ann Arbor, Michigan itations. We used the vesicular acetylcholine transporter (VAChT) ligand [18F]fluoroethoxyben- 3University of Michigan Morris K. Udall Center zovesamicol ([18F]FEOBV) and positron emission tomography to determine the regional of Excellence for Research in Parkinson's distribution of VAChT binding sites in normal human brain. We studied 29 subjects (mean age Disease, Ann Arbor, Michigan 47 [range 20–81] years; 18 men; 11 women). [18F]FEOBV binding was highest in striatum, inter- 4Michigan Alzheimer Disease Center, Ann Arbor, Michigan mediate in the amygdala, hippocampal formation, thalamus, rostral brainstem, some cerebellar 18 5Department of Radiology, University of regions, and lower in other regions. Neocortical [ F]FEOBV binding was inhomogeneous with Michigan, Ann Arbor, Michigan relatively high binding in insula, BA24, BA25, BA27, BA28, BA34, BA35, pericentral cortex, and 6Department of Cell and Developmental lowest in BA17–19. Thalamic [18F]FEOBV binding was inhomogeneous with greatest binding in Biology, University of Michigan, Ann Arbor, the lateral geniculate nuclei and relatively high binding in medial and posterior thalamus.
    [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]
  • In Alzheimer Disease: Never Change a Winning Team and Do Not Build Exclusively on Surrogates
    Journal of Pharmacology & Clinical Research ISSN: 2473-5574 Review Article J of Pharmacol & Clin Res Volume 2 Issue 1 - February 2017 Copyright © All rights are reserved by Jan M Keppel Hesselink DOI: 10.19080/JPCR.2017.02.555580 Idalopirdine (LY483518, SGS518, Lu AE 58054) in Alzheimer disease: never change a winning team and do not build exclusively on surrogates. Lessons Learned from Drug Development Trials Jan M Keppel Hesselink* Department of Molecular Pharmacology, University Witten/Herdecke, Germany Submission: January 11, 2017; Published: February 07, 2017 *Corresponding author: Jan M Keppel Hesselink, Department of Molecular Pharmacology, University Witten/Herdecke, Germany, Email: Abstract The effect of Acetylcholinesterase inhibition on Alzheimer’s disease is modest. Augmentation strategies are thus whished for and explored. acetylcholinesterase inhibitors in animal pharmacology. A phase Idalopirdine is a 5HT6 antagonist, and was found to augment the efficacy of II study supported the concept, however the study did not follow a dose-finding design, but focused on one dose only, 90 mg/daily. Currently Wea phase will discussIII program the phase further II andevaluates phase IIIthe program value of ofsuch idalopirdine augmentation in Alzheimer strategy. disease The first and phase outline III thetrial lessons however learned missed for the drug target. development: This first phase III trial was underdosed; maximum dose was 60 mg/daily, possibly based on an overly firm belief in surrogate parameters, a PET study. mg/day). Subsequently do not change the dose-regime from t.i.d. in phase II to once daily in phase III, even if surrogate parameters support always use fixed dose range studies in phase II, first define the lowest effective dose and the no-effect dose, as well as the effective dose (90 conservative drug development pattern and avoid cutting corners seems the lesson of this case of idalopirdine in Alzheimer disease.
    [Show full text]
  • Dietary Restriction of Amino Acids for Cancer Therapy Jian-Sheng Kang
    Kang Nutrition & Metabolism (2020) 17:20 https://doi.org/10.1186/s12986-020-00439-x REVIEW Open Access Dietary restriction of amino acids for Cancer therapy Jian-Sheng Kang Abstract Biosyntheses of proteins, nucleotides and fatty acids, are essential for the malignant proliferation and survival of cancer cells. Cumulating research findings show that amino acid restrictions are potential strategies for cancer interventions. Meanwhile, dietary strategies are popular among cancer patients. However, there is still lacking solid rationale to clarify what is the best strategy, why and how it is. Here, integrated analyses and comprehensive summaries for the abundances, signalling and functions of amino acids in proteomes, metabolism, immunity and food compositions, suggest that, intermittent dietary lysine restriction with normal maize as an intermittent staple food for days or weeks, might have the value and potential for cancer prevention or therapy. Moreover, dietary supplements were also discussed for cancer cachexia including dietary immunomodulatory. Keywords: Amino acid restriction, Cancer, Lysine, Kwashiorkor, Tryptophan, Arginine, Cachexia Introduction common and effective metabolic intervention for cancer? Cancer is a complex disease. There are more than 100 For amino acids (AAs), which is the most heavily used distinct types of cancer but sharing common hallmarks, AA in vivo? Which AA restriction is cell proliferation including sustaining proliferative signaling and evading the most sensitive to? What kind of dietary strategies are growth suppressors [1, 2]. The anabolic and catabolic practically available for cancer control? metabolisms of cancer cells must be reprogrammed to maintain their proliferation and survival, and may even hijack normal cells to create tumor microenvironment AA metabolism is the leading energy-consuming process (TME) for tumorigenesis and avoiding immune destruc- The consumption and release profiles of 219 metabolites tion [2].
    [Show full text]
  • Zebrafish Behavioural Profiling Identifies GABA and Serotonin
    ARTICLE https://doi.org/10.1038/s41467-019-11936-w OPEN Zebrafish behavioural profiling identifies GABA and serotonin receptor ligands related to sedation and paradoxical excitation Matthew N. McCarroll1,11, Leo Gendelev1,11, Reid Kinser1, Jack Taylor 1, Giancarlo Bruni 2,3, Douglas Myers-Turnbull 1, Cole Helsell1, Amanda Carbajal4, Capria Rinaldi1, Hye Jin Kang5, Jung Ho Gong6, Jason K. Sello6, Susumu Tomita7, Randall T. Peterson2,10, Michael J. Keiser 1,8 & David Kokel1,9 1234567890():,; Anesthetics are generally associated with sedation, but some anesthetics can also increase brain and motor activity—a phenomenon known as paradoxical excitation. Previous studies have identified GABAA receptors as the primary targets of most anesthetic drugs, but how these compounds produce paradoxical excitation is poorly understood. To identify and understand such compounds, we applied a behavior-based drug profiling approach. Here, we show that a subset of central nervous system depressants cause paradoxical excitation in zebrafish. Using this behavior as a readout, we screened thousands of compounds and identified dozens of hits that caused paradoxical excitation. Many hit compounds modulated human GABAA receptors, while others appeared to modulate different neuronal targets, including the human serotonin-6 receptor. Ligands at these receptors generally decreased neuronal activity, but paradoxically increased activity in the caudal hindbrain. Together, these studies identify ligands, targets, and neurons affecting sedation and paradoxical excitation in vivo in zebrafish. 1 Institute for Neurodegenerative Diseases, University of California, San Francisco, CA 94143, USA. 2 Cardiovascular Research Center and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA.
    [Show full text]
  • GPCR/G Protein
    Inhibitors, Agonists, Screening Libraries www.MedChemExpress.com GPCR/G Protein G Protein Coupled Receptors (GPCRs) perceive many extracellular signals and transduce them to heterotrimeric G proteins, which further transduce these signals intracellular to appropriate downstream effectors and thereby play an important role in various signaling pathways. G proteins are specialized proteins with the ability to bind the nucleotides guanosine triphosphate (GTP) and guanosine diphosphate (GDP). In unstimulated cells, the state of G alpha is defined by its interaction with GDP, G beta-gamma, and a GPCR. Upon receptor stimulation by a ligand, G alpha dissociates from the receptor and G beta-gamma, and GTP is exchanged for the bound GDP, which leads to G alpha activation. G alpha then goes on to activate other molecules in the cell. These effects include activating the MAPK and PI3K pathways, as well as inhibition of the Na+/H+ exchanger in the plasma membrane, and the lowering of intracellular Ca2+ levels. Most human GPCRs can be grouped into five main families named; Glutamate, Rhodopsin, Adhesion, Frizzled/Taste2, and Secretin, forming the GRAFS classification system. A series of studies showed that aberrant GPCR Signaling including those for GPCR-PCa, PSGR2, CaSR, GPR30, and GPR39 are associated with tumorigenesis or metastasis, thus interfering with these receptors and their downstream targets might provide an opportunity for the development of new strategies for cancer diagnosis, prevention and treatment. At present, modulators of GPCRs form a key area for the pharmaceutical industry, representing approximately 27% of all FDA-approved drugs. References: [1] Moreira IS. Biochim Biophys Acta. 2014 Jan;1840(1):16-33.
    [Show full text]