1
|
Fawzy MA, Ibrahim KH, Aly AA, Mohamed AH, Naguib Abdel Hafez SM, Abdelzaher WY, Elkaeed EB, Alsfouk AA, Abdelhafez ESM. One-pot synthesis and pharmacological evaluation of new quinoline/pyrimido-diazepines as pulmonary antifibrotic agents. Future Med Chem 2024:1-20. [PMID: 39291539 DOI: 10.1080/17568919.2024.2394018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Aim: Pulmonary fibrosis is a life threating disease which requires an immediate treatment and due to the limited medications, this study focused on synthesizing a series of quinoline-based pyrimidodiazepines 4a-f as a novel antifibrotic hit.Materials & methods: The target compounds were synthesized via a one-pot reaction then investigated in a rat model of lung fibrosis induced by bleomycin (BLM).Results: Results revealed significant attenuation of the tested pro-inflammatory cytokines, fibrotic genes and apoptotic markers; however, Bcl-2 was upregulated, indicating a protective effect against fibrosis. Moreover, the molecular docking studies highlighted promising interactions between compounds 4b and 4c and specific amino acids within the protein pockets of caspase-3 (ARG341 and THR177), malondialdehyde (LYS195, LYS118 and ARG188) and TNF-α (SER99 and NME102).Conclusion: Compounds 4b and 4c emerge as promising candidates for further preclinical investigation as pulmonary antifibrotic agents.
Collapse
Affiliation(s)
- Michael Atef Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, 61519, Egypt
| | - Karim Hagag Ibrahim
- Department of Biochemistry, Faculty of Pharmacy, Minia University, 61519, Egypt
| | - Ashraf A Aly
- Chemistry Department, Faculty of Science, Minia University, El-Minia, 61519, Egypt
| | - Asmaa H Mohamed
- Chemistry Department, Faculty of Science, Minia University, El-Minia, 61519, Egypt
| | | | - Walaa Yehia Abdelzaher
- Department of Medical Pharmacology, Faculty of Medicine, Minia University, Minia, 61519, Egypt
| | - Eslam B Elkaeed
- Department of Pharmaceutical Sciences, College of Pharmacy, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Cairo, 11884, Egypt
| | - Aisha A Alsfouk
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - El-Shimaa Mn Abdelhafez
- Department of Medicinal Chemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| |
Collapse
|
2
|
Shu L, Du C, Zuo Y. Abnormal phosphorylation of protein tyrosine in neurodegenerative diseases. J Neuropathol Exp Neurol 2023; 82:826-835. [PMID: 37589710 DOI: 10.1093/jnen/nlad066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/18/2023] Open
Abstract
Neurodegenerative diseases, including Alzheimer disease, Parkinson disease, amyotrophic lateral sclerosis, and multiple sclerosis, are chronic disorders of the CNS that are characterized by progressive neuronal dysfunction. These diseases have diverse clinical and pathological features and their pathogenetic mechanisms are not yet fully understood. Currently, widely accepted hypotheses include the accumulation of misfolded proteins, oxidative stress from reactive oxygen species, mitochondrial dysfunction, DNA damage, neurotrophin dysfunction, and neuroinflammatory processes. In the CNS of patients with neurodegenerative diseases, a variety of abnormally phosphorylated proteins play important roles in pathological processes such as neuroinflammation and intracellular accumulation of β-amyloid plaques and tau. In recent years, the roles of abnormal tyrosine phosphorylation of intracellular signaling molecules regulated by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) in neurodegenerative diseases have attracted increasing attention. Here, we summarize the roles of signaling pathways related to protein tyrosine phosphorylation in the pathogenesis of neurodegenerative diseases and the progress of therapeutic studies targeting PTKs and PTPs that provide theoretical support for future studies on therapeutic strategies for these devastating and important neurodegenerative diseases.
Collapse
Affiliation(s)
- Lijuan Shu
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
- Department of Obstetrics and Gynecology Intensive Care Unit, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Chunfu Du
- Department of Neurosurgery, Ya'an People's Hospital, Ya'an, China
| | - Yunxia Zuo
- Department of Anesthesiology, West China Hospital, Sichuan University & The Research Units of West China (2018RU012), Chinese Academy of Medical Sciences, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Wolfe MS, Miao Y. Structure and mechanism of the γ-secretase intramembrane protease complex. Curr Opin Struct Biol 2022; 74:102373. [PMID: 35461161 DOI: 10.1016/j.sbi.2022.102373] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 12/25/2022]
Abstract
γ-Secretase is a membrane protein complex that proteolyzes within the transmembrane domain of >100 substrates, including those derived from the amyloid precursor protein and the Notch family of cell surface receptors. The nine-transmembrane presenilin is the catalytic component of this aspartyl protease complex that carries out hydrolysis in the lipid bilayer. Advances in cryoelectron microscopy have led to the elucidation of the structure of the γ-secretase complex at atomic resolution. Recently, structures of the enzyme have been determined with bound APP- or Notch-derived substrates, providing insight into the nature of substrate recognition and processing. Molecular dynamics simulations of substrate-bound enzymes suggest dynamic mechanisms of intramembrane proteolysis. Structures of the enzyme bound to small-molecule inhibitors and modulators have also been solved, setting the stage for rational structure-based drug discovery targeting γ-secretase.
Collapse
Affiliation(s)
- Michael S Wolfe
- Department of Medicinal Chemistry, University of Kansas, Lawrence, KS, 66045, USA.
| | - Yinglong Miao
- Center for Computational Biology, Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA. https://twitter.com/yinglongmiao
| |
Collapse
|
4
|
Hur JY. γ-Secretase in Alzheimer's disease. Exp Mol Med 2022; 54:433-446. [PMID: 35396575 PMCID: PMC9076685 DOI: 10.1038/s12276-022-00754-8] [Citation(s) in RCA: 96] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 01/05/2022] [Accepted: 01/20/2022] [Indexed: 12/16/2022] Open
Abstract
Alzheimer's disease (AD) is caused by synaptic and neuronal loss in the brain. One of the characteristic hallmarks of AD is senile plaques containing amyloid β-peptide (Aβ). Aβ is produced from amyloid precursor protein (APP) by sequential proteolytic cleavages by β-secretase and γ-secretase, and the polymerization of Aβ into amyloid plaques is thought to be a key pathogenic event in AD. Since γ-secretase mediates the final cleavage that liberates Aβ, γ-secretase has been widely studied as a potential drug target for the treatment of AD. γ-Secretase is a transmembrane protein complex containing presenilin, nicastrin, Aph-1, and Pen-2, which are sufficient for γ-secretase activity. γ-Secretase cleaves >140 substrates, including APP and Notch. Previously, γ-secretase inhibitors (GSIs) were shown to cause side effects in clinical trials due to the inhibition of Notch signaling. Therefore, more specific regulation or modulation of γ-secretase is needed. In recent years, γ-secretase modulators (GSMs) have been developed. To modulate γ-secretase and to understand its complex biology, finding the binding sites of GSIs and GSMs on γ-secretase as well as identifying transiently binding γ-secretase modulatory proteins have been of great interest. In this review, decades of findings on γ-secretase in AD are discussed.
Collapse
Affiliation(s)
- Ji-Yeun Hur
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
5
|
Small molecules targeting γ-secretase and their potential biological applications. Eur J Med Chem 2022; 232:114169. [DOI: 10.1016/j.ejmech.2022.114169] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 01/30/2022] [Accepted: 01/30/2022] [Indexed: 12/14/2022]
|
6
|
Luo JE, Li YM. Turning the tide on Alzheimer's disease: modulation of γ-secretase. Cell Biosci 2022; 12:2. [PMID: 34983641 PMCID: PMC8725520 DOI: 10.1186/s13578-021-00738-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 12/17/2021] [Indexed: 12/17/2022] Open
Abstract
Alzheimer's disease (AD) is the most common type of neurodegenerative disorder. Amyloid-beta (Aβ) plaques are integral to the "amyloid hypothesis," which states that the accumulation of Aβ peptides triggers a cascade of pathological events leading to neurodegeneration and ultimately AD. While the FDA approved aducanumab, the first Aβ-targeted therapy, multiple safe and effective treatments will be needed to target the complex pathologies of AD. γ-Secretase is an intramembrane aspartyl protease that is critical for the generation of Aβ peptides. Activity and specificity of γ-secretase are regulated by both obligatory subunits and modulatory proteins. Due to its complex structure and function and early clinical failures with pan inhibitors, γ-secretase has been a challenging drug target for AD. γ-secretase modulators, however, have dramatically shifted the approach to targeting γ-secretase. Here we review γ-secretase and small molecule modulators, from the initial characterization of a subset of NSAIDs to the most recent clinical candidates. We also discuss the chemical biology of γ-secretase, in which small molecule probes enabled structural and functional insights into γ-secretase before the emergence of high-resolution structural studies. Finally, we discuss the recent crystal structures of γ-secretase, which have provided valuable perspectives on substrate recognition and molecular mechanisms of small molecules. We conclude that modulation of γ-secretase will be part of a new wave of AD therapeutics.
Collapse
Affiliation(s)
- Joanna E Luo
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA.
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA. .,Program of Pharmacology, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, 10021, USA.
| |
Collapse
|
7
|
Weber TA, Lundkvist J, Wanngren J, Kvartsberg H, Jin S, Larssen P, Wu D, Oliveira DV, Minta K, Brinkmalm G, Zetterberg H, Blennow K, Nordvall G, Winblad B, Portelius E, Karlström H. γ-Secretase modulators show selectivity for γ-secretase-mediated amyloid precursor protein intramembrane processing. J Cell Mol Med 2021; 26:880-892. [PMID: 34931449 PMCID: PMC8817114 DOI: 10.1111/jcmm.17146] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 07/15/2021] [Accepted: 12/08/2021] [Indexed: 11/29/2022] Open
Abstract
The aggregation of β‐amyloid peptide 42 results in the formation of toxic oligomers and plaques, which plays a pivotal role in Alzheimer's disease pathogenesis. Aβ42 is one of several Aβ peptides, all of Aβ30 to Aβ43 that are produced as a result of γ‐secretase–mediated regulated intramembrane proteolysis of the amyloid precursor protein. γ‐Secretase modulators (GSMs) represent a promising class of Aβ42‐lowering anti‐amyloidogenic compounds for the treatment of AD. Gamma‐secretase modulators change the relative proportion of secreted Aβ peptides, while sparing the γ‐secretase–mediated processing event resulting in the release of the cytoplasmic APP intracellular domain. In this study, we have characterized how GSMs affect the γ‐secretase cleavage of three γ‐secretase substrates, E‐cadherin, ephrin type A receptor 4 (EphA4) and ephrin type B receptor 2 (EphB2), which all are implicated in important contexts of cell signalling. By using a reporter gene assay, we demonstrate that the γ‐secretase–dependent generation of EphA4 and EphB2 intracellular domains is unaffected by GSMs. We also show that γ‐secretase processing of EphA4 and EphB2 results in the release of several Aβ‐like peptides, but that only the production of Aβ‐like proteins from EphA4 is modulated by GSMs, but with an order of magnitude lower potency as compared to Aβ modulation. Collectively, these results suggest that GSMs are selective for γ‐secretase–mediated Aβ production.
Collapse
Affiliation(s)
- Tobias A Weber
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden.,Medical Scientific Affairs, Aesculap AG, Tuttlingen, Germany
| | - Johan Lundkvist
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden.,Alzecure Pharma, Huddinge, Sweden.,Sinfonia Biotherapeutics AB, Huddinge, Sweden
| | - Johanna Wanngren
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Hlin Kvartsberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - ShaoBo Jin
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Pia Larssen
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Dan Wu
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden.,Department of Obstetrics and Gynecology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Daniel V Oliveira
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden
| | - Karolina Minta
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Gunnar Brinkmalm
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden.,Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK.,UK Dementia Research Institute at UCL, London, UK
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Gunnar Nordvall
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden.,Alzecure Pharma, Huddinge, Sweden
| | - Bengt Winblad
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden
| | - Erik Portelius
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden.,Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
| | - Helena Karlström
- Division of Neurogeriatrics, Department of Neurobiology, Care Science and Society, Karolinska Institutet, Stockholm, Sweden.,Theme Aging, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden
| |
Collapse
|
8
|
Rynearson KD, Buckle RN, Herr RJ, Mayhew NJ, Chen X, Paquette WD, Sakwa SA, Yang J, Barnes KD, Nguyen P, Mobley WC, Johnson G, Lin JH, Tanzi RE, Wagner SL. Design and synthesis of novel methoxypyridine-derived gamma-secretase modulators. Bioorg Med Chem 2020; 28:115734. [PMID: 33007551 DOI: 10.1016/j.bmc.2020.115734] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 08/18/2020] [Accepted: 08/21/2020] [Indexed: 11/19/2022]
Abstract
The evolution of gamma-secretase modulators (GSMs) through the introduction of novel heterocycles with the goal of aligning activity for reducing the levels of Aβ42 and properties consistent with a drug-like molecule are described. The insertion of a methoxypyridine motif within the tetracyclic scaffold provided compounds with improved activity for arresting Aβ42 production as well as improved properties, including solubility. In vivo pharmacokinetic analysis demonstrated that several compounds within the novel series were capable of crossing the BBB and accessing the therapeutic target. Treatment with methoxypyridine-derived compound 64 reduced Aβ42 levels in the plasma of J20 mice, in addition to reducing Aβ42 levels in the plasma and brain of Tg2576 mice.
Collapse
Affiliation(s)
- Kevin D Rynearson
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093-0624, United States.
| | - Ronald N Buckle
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - R Jason Herr
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - Nicholas J Mayhew
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - Xinchao Chen
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - William D Paquette
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - Samuel A Sakwa
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - Jinhai Yang
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - Keith D Barnes
- Department of Medicinal Chemistry, AMRI, East Campus, 3 University Place, Rensselaer, NY 12144, United States
| | - Phuong Nguyen
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093-0624, United States
| | - William C Mobley
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093-0624, United States
| | - Graham Johnson
- NuPharmAdvise, 3 Lakeside Drive, Sanbornton, NH 03269, United States
| | - Juinn H Lin
- Biopharm Consulting Partners, 2 Willet Drive, Ambler, PA 19002, United States
| | - Rudolph E Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, United States
| | - Steven L Wagner
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093-0624, United States; Veterans Administrative San Diego Healthcare System, La Jolla, CA 92161, United States.
| |
Collapse
|
9
|
Robust approach leading to novel densely functionalized four-cyclic benzo[e]pyrazolo[5′,1′:2,3]pyrimido[4,5-b][1,4]diazepines with antibacterial activity toward resistant strains. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2020. [DOI: 10.1007/s13738-020-01875-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
10
|
Nie P, Vartak A, Li YM. γ-Secretase inhibitors and modulators: Mechanistic insights into the function and regulation of γ-Secretase. Semin Cell Dev Biol 2020; 105:43-53. [PMID: 32249070 DOI: 10.1016/j.semcdb.2020.03.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 03/10/2020] [Accepted: 03/11/2020] [Indexed: 02/08/2023]
Abstract
Over two decades, γ-secretase has been the target for extensive therapeutic development due to its pivotal role in pathogenesis of Alzheimer's disease and cancer. However, it has proven to be a challenging task owing to its large set of substrates and our limited understanding of the enzyme's structural and mechanistic features. The scientific community is taking bigger strides towards solving this puzzle with recent advancement in techniques like cryogenic electron microscopy (cryo-EM) and photo-affinity labelling (PAL). This review highlights the significance of the PAL technique with multiple examples of photo-probes developed from γ-secretase inhibitors and modulators. The binding of these probes into active and/or allosteric sites of the enzyme has provided crucial information on the γ-secretase complex and improved our mechanistic understanding of this protease. Combining the knowledge of function and regulation of γ-secretase will be a decisive factor in developing novel γ-secretase modulators and biological therapeutics.
Collapse
Affiliation(s)
- Pengju Nie
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology program, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA
| | - Abhishek Vartak
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Pharmacology program, Weill Graduate School of Medical Sciences of Cornell University, New York, NY 10021, USA.
| |
Collapse
|
11
|
Human-Induced Pluripotent Stem Cells and Herbal Small-Molecule Drugs for Treatment of Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21041327. [PMID: 32079110 PMCID: PMC7072986 DOI: 10.3390/ijms21041327] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Revised: 02/13/2020] [Accepted: 02/14/2020] [Indexed: 12/28/2022] Open
Abstract
Alzheimer’s disease (AD) is characterized by extracellular amyloid plaques composed of the β-amyloid peptides and intracellular neurofibrillary tangles and associates with progressive declines in memory and cognition. Several genes play important roles and regulate enzymes that produce a pathological accumulation of β-amyloid in the brain, such as gamma secretase (γ-secretase). Induced pluripotent stem cells from patients with Alzheimer’s disease with different underlying genetic mechanisms may help model different phenotypes of Alzheimer’s disease and facilitate personalized drug screening platforms for the identification of small molecules. We also discuss recent developments by γ-secretase inhibitors and modulators in the treatment of AD. In addition, small-molecule drugs isolated from Chinese herbal medicines have been shown effective in treating Alzheimer’s disease. We propose a mechanism of small-molecule drugs in treating Alzheimer’s disease. Combining therapy with different small-molecule drugs may increase the chance of symptomatic treatment. A customized strategy tailored to individuals and in combination with therapy may be a more suitable treatment option for Alzheimer’s disease in the future.
Collapse
|
12
|
Straightforward synthesis of pyrimido[4,5-e][1,4]diazepines via 6-aminopyrimidin-5-carbaldehydes. ARAB J CHEM 2019. [DOI: 10.1016/j.arabjc.2016.07.012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
13
|
Kounnas MZ, Durakoglugil MS, Herz J, Comer WT. NGP 555, a γ-secretase modulator, shows a beneficial shift in the ratio of amyloid biomarkers in human cerebrospinal fluid at safe doses. ALZHEIMER'S & DEMENTIA (NEW YORK, N. Y.) 2019; 5:458-467. [PMID: 31921961 PMCID: PMC6944734 DOI: 10.1016/j.trci.2019.06.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Currently, there is no cure for Alzheimer's disease (AD), and it is widely accepted that AD is a complex disease with multiple approaches necessary to prevent and treat the disease. METHODS Using amyloid biomarkers in human cerebrospinal fluid, pharmacokinetic, safety, and metabolism studies, we investigate the properties of NGP 555, γ-secretase modulator, for the first time in human clinical trials. RESULTS Our preclinical and clinical studies combined show beneficial effects with NGP 555 on synaptic response and amyloid cerebrospinal fluid biomarkers while avoiding negative side effects. Importantly, pharmacokinetic and pharmacodynamic parameters combined with safety outcomes indicate that NGP 555 penetrates the blood-brain barrier and increases the ratio of amyloid-β peptide Aβ37 and Aβ38 compared with that of Aβ42, establishing a proof of target engagement in humans in a 14 day, once-daily oral dosing trial. DISCUSSION In humans, NGP 555 has demonstrated a beneficial shift in the production of Aβ37 and Aβ38 versus Aβ42 biomarker levels in the cerebrospinal fluid while maintaining an adequate safety profile. The overall clinical goal is to achieve an optimal balance of efficacy for altering amyloid-β peptide (Aβ) biomarkers while maintaining a safe profile so that NGP 555 can be given early in AD to prevent production of Aβ42 and accumulation of amyloid plaques, in an effort to prevent aggregation of tau and destruction of neurons and synapses resulting in cognitive decline.
Collapse
Affiliation(s)
- Maria Z. Kounnas
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - Murat S. Durakoglugil
- Department of Molecular Genetics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX
| | - Joachim Herz
- Department of Molecular Genetics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX
| | | |
Collapse
|
14
|
The amyloid cascade and Alzheimer's disease therapeutics: theory versus observation. J Transl Med 2019; 99:958-970. [PMID: 30760863 DOI: 10.1038/s41374-019-0231-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/31/2022] Open
Abstract
The identification of amyloid-β precursor protein (APP) pathogenic mutations in familial early onset Alzheimer's disease (AD), along with knowledge that amyloid-β (Aβ) was the principle protein component of senile plaques, led to the establishment of the amyloid cascade hypothesis. Down syndrome substantiated the hypothesis, given an extra copy of the APP gene and invariable AD pathology hallmarks that occur by middle age. An abundance of support for the amyloid cascade hypothesis followed. Prion-like protein misfolding and non-Mendelian transmission of neurotoxicity are among recent areas of investigation. Aβ-targeted clinical trials have been disappointing, with negative results attributed to inadequacies in patient selection, challenges in pharmacology, and incomplete knowledge of the most appropriate target. There is evidence, however, that proof of concept has been achieved, i.e., clearance of Aβ during life, but with no significant changes in cognitive trajectory in AD. Whether the time, effort, and expense of Aβ-targeted therapy will prove valuable will be determined over time, as Aβ-centered clinical trials continue to dominate therapeutic strategies. It seems reasonable to hypothesize that the amyloid cascade is intimately involved in AD, in parallel with disease pathogenesis, but that removal of toxic Aβ is insufficient for an effective disease modification.
Collapse
|
15
|
Kuo YC, Rajesh R. Challenges in the treatment of Alzheimer’s disease: recent progress and treatment strategies of pharmaceuticals targeting notable pathological factors. Expert Rev Neurother 2019; 19:623-652. [DOI: 10.1080/14737175.2019.1621750] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Yung-Chih Kuo
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| | - Rajendiran Rajesh
- Department of Chemical Engineering, National Chung Cheng University, Chia-Yi, Taiwan, Republic of China
| |
Collapse
|
16
|
Morsy A, Trippier PC. Current and Emerging Pharmacological Targets for the Treatment of Alzheimer's Disease. J Alzheimers Dis 2019; 72:S145-S176. [PMID: 31594236 DOI: 10.3233/jad-190744] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
No cure or disease-modifying therapy for Alzheimer's disease (AD) has yet been realized. However, a multitude of pharmacological targets have been identified for possible engagement to enable drug discovery efforts for AD. Herein, we review these targets comprised around three main therapeutic strategies. First is an approach that targets the main pathological hallmarks of AD: amyloid-β (Aβ) oligomers and hyperphosphorylated tau tangles which primarily focuses on reducing formation and aggregation, and/or inducing their clearance. Second is a strategy that modulates neurotransmitter signaling. Comprising this strategy are the cholinesterase inhibitors and N-methyl-D-aspartate receptor blockade treatments that are clinically approved for the symptomatic treatment of AD. Additional targets that aim to stabilize neuron signaling through modulation of neurotransmitters and their receptors are also discussed. Finally, the third approach comprises a collection of 'sensitive targets' that indirectly influence Aβ or tau accumulation. These targets are proteins that upon Aβ accumulation in the brain or direct Aβ-target interaction, a modification in the target's function is induced. The process occurs early in disease progression, ultimately causing neuronal dysfunction. This strategy aims to restore normal target function to alleviate Aβ-induced toxicity in neurons. Overall, we generally limit our analysis to targets that have emerged in the last decade and targets that have been validated using small molecules in in vitro and/or in vivo models. This review is not an exhaustive list of all possible targets for AD but serves to highlight the most promising and critical targets suitable for small molecule drug intervention.
Collapse
Affiliation(s)
- Ahmed Morsy
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Paul C Trippier
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
- UNMC Center for Drug Discovery, University of Nebraska Medical Center, Omaha, NE, USA
| |
Collapse
|
17
|
Zeng X, Kirkpatrick R, Hofmann G, Grillot D, Linhart V, Viviani F, Marino J, Boyer J, Graham TL, Lu Q, Wu Z, Benowitz A, Cousins R. Screen for modulators of atonal homolog 1 gene expression using notch pathway-relevant gene transcription based cellular assays. PLoS One 2018; 13:e0207140. [PMID: 30540745 PMCID: PMC6291236 DOI: 10.1371/journal.pone.0207140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 10/25/2018] [Indexed: 12/11/2022] Open
Abstract
Atonal homolog 1 (Atoh1) is a basic helix-loop-helix 9 (bHLH) transcription factor acting downstream of Notch and is required for the differentiation of sensory hair cells in the inner ear and the specification of secretory cells during the intestinal crypt cell regeneration. Motivated by the observations that the upregulation of Atoh1 gene expression, through genetic manipulation or pharmacological inhibition of Notch signaling (e.g. γ-secretase inhibitors, GSIs), induces ectopic hair cell growth in the cochlea of the inner ear and partially restores hearing after injuries in experimental models, we decided to identify small molecule modulators of the Notch-Atoh1 pathway, which could potentially regenerate hair cells. However, the lack of cellular models of the inner ear has precluded the screening and characterization of such modulators. Here we report using a colon cancer cell line LS-174T, which displays Notch inhibition-dependent Atoh1 expression as a surrogate cellular model to screen for inducers of Atoh1 expression. We designed an Atoh1 promoter-driven luciferase assay to screen a target-annotated library of ~6000 compounds. We further developed a medium throughput, real-time quantitative RT-PCR assay measuring the endogenous Atoh1 gene expression to confirm the hits and eliminate false positives from the reporter-based screen. This strategy allowed us to successfully recover GSIs of known chemotypes. This LS-174T cell-based assay directly measures Atoh1 gene expression induced through Notch-Hes1 inhibition, and therefore offers an opportunity to identify novel cellular modulators along the Notch-Atoh1 pathway.
Collapse
Affiliation(s)
- Xin Zeng
- R&D Target Sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
- * E-mail: (XZ); (RC)
| | - Robert Kirkpatrick
- R&D Alternative Discovery and Development, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Glenn Hofmann
- R&D Platform Technology Sciences, Drug Design and Selection, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Didier Grillot
- R&D Flexible Discovery Unit, Villebon-sur-Yvette, Paris, France
| | - Valerie Linhart
- R&D Flexible Discovery Unit, Villebon-sur-Yvette, Paris, France
| | - Fabrice Viviani
- R&D Flexible Discovery Unit, Villebon-sur-Yvette, Paris, France
| | - Joseph Marino
- R&D Alternative Discovery and Development, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Joseph Boyer
- R&D Statistical sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Taylor L. Graham
- R&D Target Sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Quinn Lu
- R&D Target Sciences, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Zining Wu
- R&D Platform Technology Sciences, Drug Design and Selection, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Andrew Benowitz
- R&D Alternative Discovery and Development, GlaxoSmithKline, Upper Providence, Collegeville, United States of America
| | - Rick Cousins
- R&D Alternative Discovery and Development, GlaxoSmithKline, Stevenage, Hertfordshire, United Kingdom
- * E-mail: (XZ); (RC)
| |
Collapse
|
18
|
Allosteric Modulation of Intact γ-Secretase Structural Dynamics. Biophys J 2018; 113:2634-2649. [PMID: 29262358 DOI: 10.1016/j.bpj.2017.10.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/26/2017] [Accepted: 10/10/2017] [Indexed: 12/20/2022] Open
Abstract
As a protease complex involved in the cleavage of amyloid precursor proteins that lead to the formation of amyloid β fibrils implicated in Alzheimer's disease, γ-secretase is an important target for developing therapeutics against Alzheimer's disease. γ-secretase is composed of four subunits: nicastrin (NCT) in the extracellular (EC) domain, presenilin-1 (PS1), anterior pharynx defective 1, and presenilin enhancer 2 in the transmembrane (TM) domain. NCT and PS1 play important roles in binding amyloid β precursor proteins and modulating PS1 catalytic activity. Yet, the molecular mechanisms of coupling between substrate/modulator binding and catalytic activity remain to be elucidated. Recent determination of intact human γ-secretase cryo-electron microscopy structure has opened the way for a detailed investigation of the structural dynamics of this complex. Our analysis, based on a membrane-coupled anisotropic network model, reveals two types of NCT motions, bending and twisting, with respect to PS1. These underlie the fluctuations between the "open" and "closed" states of the lid-like NCT with respect to a hydrophilic loop 1 (HL1) on PS1, thus allowing or blocking access of the substrate peptide (EC portion) to HL1 and to the neighboring helix TM2. In addition to this alternating access mechanism, fluctuations in the volume of the PS1 central cavity facilitate the exposure of the catalytic site for substrate cleavage. Druggability simulations show that γ-secretase presents several hot spots for either orthosteric or allosteric inhibition of catalytic activity, consistent with experimental data. In particular, a hinge region at the interface between the EC and TM domains, near the interlobe groove of NCT, emerges as an allo-targeting site that would impact the coupling between HL1/TM2 and the catalytic pocket, opening, to our knowledge, new avenues for structure-based design of novel allosteric modulators of γ-secretase protease activity.
Collapse
|
19
|
Johnson DS, Li YM, Pettersson M, St George-Hyslop PH. Structural and Chemical Biology of Presenilin Complexes. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a024067. [PMID: 28320827 PMCID: PMC5710098 DOI: 10.1101/cshperspect.a024067] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The presenilin proteins are the catalytic subunits of a tetrameric complex containing presenilin 1 or 2, anterior pharynx defective 1 (APH1), nicastrin, and PEN-2. Other components such as TMP21 may exist in a subset of specialized complexes. The presenilin complex is the founding member of a unique class of aspartyl proteases that catalyze the γ, ɛ, ζ site cleavage of the transmembrane domains of Type I membrane proteins including amyloid precursor protein (APP) and Notch. Here, we detail the structural and chemical biology of this unusual enzyme. Taken together, these studies suggest that the complex exists in several conformations, and subtle long-range (allosteric) shifts in the conformation of the complex underpin substrate access to the catalytic site and the mechanism of action for allosteric inhibitors and modulators. Understanding the mechanics of these shifts will facilitate the design of γ-secretase modulator (GSM) compounds that modulate the relative efficiency of γ, ɛ, ζ site cleavage and/or substrate specificity.
Collapse
Affiliation(s)
- Douglas S. Johnson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Yue-Ming Li
- Chemical Biology Program, Memorial Sloan Kettering Cancer Center, New York, New York 10065
| | - Martin Pettersson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Peter H. St George-Hyslop
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Cambridge CB2 0XY, United Kingdom,Tanz Centre for Research in Neurodegenerative Diseases and Departments of Medicine, Laboratory Medicine and Pathobiology, and Medical Biophysics, University of Toronto, Toronto, Ontario M5T 2S8, Canada
| |
Collapse
|
20
|
Robertson AS, Iben LG, Wei C, Meredith JE, Drexler DM, Banks M, Vite GD, Olson RE, Thompson LA, Albright CF, Ahlijanian MK, Toyn JH. Synergistic inhibition of Aβ production by combinations of γ-secretase modulators. Eur J Pharmacol 2017; 812:104-112. [DOI: 10.1016/j.ejphar.2017.07.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 07/03/2017] [Accepted: 07/05/2017] [Indexed: 01/23/2023]
|
21
|
Mehta PD, Blain JF, Freeman EA, Patrick BA, Barshatzky M, Hrdlicka LA, Mehta SP, Frackowiak J, Mazur-Kolecka B, Wegiel J, Patzke H, Miller DL. Generation and Partial Characterization of Rabbit Monoclonal Antibody to Amyloid-β Peptide 1-37 (Aβ37). J Alzheimers Dis 2017; 57:135-145. [PMID: 28222530 DOI: 10.3233/jad-161207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Secreted soluble amyloid-β 1-37 (Aβ37) peptide is one of the prominent Aβ forms next to Aβ40, and is found in cerebrospinal fluid (CSF) and blood. Recent studies have shown the importance of quantitation of CSF Aβ37 levels in combination with Aβ38, Aβ40, and Aβ42 to support the diagnosis of patients with probable Alzheimer's disease (AD), and the value of antibody to Aβ37 to facilitate drug discovery studies. However, the availability of reliable and specific monoclonal antibody to Aβ37 is very limited. Our aims were: 1) to generate and partially characterize rabbit monoclonal antibody (RabmAb) to Aβ37, and 2) to determine whether the antibody detects changes in Aβ37 levels produced by a γ-secretase modulator (GSM). Our generated RabmAb to Aβ37 was found to be specific to Aβ37, since it did not react with Aβ36, Aβ38, Aβ39, Aβ40, and Aβ42 in an ELISA or immunoblotting. The epitope of the antibody was contained in the seven C-terminal residues of Aβ37. The antibody was sensitive enough to measure CSF and plasma Aβ37 levels in ELISA. Immunohistological studies showed the presence of Aβ37-positive deposits in the brain of AD, and Down syndrome persons diagnosed with AD. Our studies also showed that the antibody detected Aβ37 increases in CSF and brains of rodents following treatment with a GSM. Thus, our antibody can be widely applied to AD research, and in a panel based approach it may have potential to support the diagnosis of probable AD, and in testing the effect of GSMs to target AD.
Collapse
Affiliation(s)
- Pankaj D Mehta
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | | | | | - Bruce A Patrick
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | - Marc Barshatzky
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | | | - Sangita P Mehta
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | - Janusz Frackowiak
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | - Bozena Mazur-Kolecka
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | - Jerzy Wegiel
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| | | | - David L Miller
- New York State Institute for Basic Research in Developmental Disabilities, Staten Island, New York, NY, USA
| |
Collapse
|
22
|
γ-Secretase Modulators as Aβ42-Lowering Pharmacological Agents to Treat Alzheimer’s Disease. TOPICS IN MEDICINAL CHEMISTRY 2017. [DOI: 10.1007/7355_2016_19] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
|
23
|
Kounnas MZ, Lane-Donovan C, Nowakowski DW, Herz J, Comer WT. NGP 555, a γ-Secretase Modulator, Lowers the Amyloid Biomarker, Aβ 42, in Cerebrospinal Fluid while Preventing Alzheimer's Disease Cognitive Decline in Rodents. ALZHEIMERS & DEMENTIA-TRANSLATIONAL RESEARCH & CLINICAL INTERVENTIONS 2016; 3:65-73. [PMID: 28497107 PMCID: PMC5421551 DOI: 10.1016/j.trci.2016.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Introduction Alzheimer's disease (AD) is defined by the progressive accumulation of amyloid plaques and neurofibrillary tangles in the brain which precedes cognitive decline by years. Methods Using amyloid biomarkers, chemical modeling, mouse behavioral models, and drug development techniques, we investigate the properties of NGP 555, a clinical-stage γ-secretase modulator. Results NGP 555 shifts amyloid peptide production to the smaller, nonaggregating forms of amyloid. Our preclinical studies show beneficial effects on amyloid biomarkers, pathology, and cognition. NGP 555 has successfully completed chemistry, pharmacology, toxicity, metabolism, and safety studies. Discussion Abundant data support Aβ42 as a target for prophylactic or early-stage intervention therapies in AD. The γ-secretase modulator, NGP 555 is being actively developed in human clinical trials for the prevention of Alzheimer's disease with the overall aim to achieve an appropriate balance of potency/efficacy on reducing the toxic forms of amyloid versus safety.
Collapse
Affiliation(s)
- Maria Z. Kounnas
- NeuroGenetic Pharmaceuticals, Inc., Del Mar, CA, USA
- Corresponding author.
| | - Courtney Lane-Donovan
- Department of Molecular Genetics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Joachim Herz
- Department of Molecular Genetics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | |
Collapse
|
24
|
Xu W, Weissmiller AM, White JA, Fang F, Wang X, Wu Y, Pearn ML, Zhao X, Sawa M, Chen S, Gunawardena S, Ding J, Mobley WC, Wu C. Amyloid precursor protein-mediated endocytic pathway disruption induces axonal dysfunction and neurodegeneration. J Clin Invest 2016; 126:1815-33. [PMID: 27064279 PMCID: PMC4855914 DOI: 10.1172/jci82409] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Accepted: 02/24/2016] [Indexed: 12/17/2022] Open
Abstract
The endosome/lysosome pathway is disrupted early in the course of both Alzheimer's disease (AD) and Down syndrome (DS); however, it is not clear how dysfunction in this pathway influences the development of these diseases. Herein, we explored the cellular and molecular mechanisms by which endosomal dysfunction contributes to the pathogenesis of AD and DS. We determined that full-length amyloid precursor protein (APP) and its β-C-terminal fragment (β-CTF) act though increased activation of Rab5 to cause enlargement of early endosomes and to disrupt retrograde axonal trafficking of nerve growth factor (NGF) signals. The functional impacts of APP and its various products were investigated in PC12 cells, cultured rat basal forebrain cholinergic neurons (BFCNs), and BFCNs from a mouse model of DS. We found that the full-length wild-type APP (APPWT) and β-CTF both induced endosomal enlargement and disrupted NGF signaling and axonal trafficking. β-CTF alone induced atrophy of BFCNs that was rescued by the dominant-negative Rab5 mutant, Rab5S34N. Moreover, expression of a dominant-negative Rab5 construct markedly reduced APP-induced axonal blockage in Drosophila. Therefore, increased APP and/or β-CTF impact the endocytic pathway to disrupt NGF trafficking and signaling, resulting in trophic deficits in BFCNs. Our data strongly support the emerging concept that dysregulation of Rab5 activity contributes importantly to early pathogenesis of AD and DS.
Collapse
Affiliation(s)
- Wei Xu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurosciences, UCSD, La Jolla, California, USA
| | | | - Joseph A. White
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Fang Fang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Neurosciences, UCSD, La Jolla, California, USA
| | - Xinyi Wang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Wu
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Matthew L. Pearn
- Department of Anesthesiology, UCSD, La Jolla, California, USA
- VA San Diego Healthcare System, San Diego, California, USA
| | - Xiaobei Zhao
- Department of Neurosciences, UCSD, La Jolla, California, USA
| | - Mariko Sawa
- Department of Neurosciences, UCSD, La Jolla, California, USA
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shermali Gunawardena
- Department of Biological Sciences, The State University of New York at Buffalo, Buffalo, New York, USA
| | - Jianqing Ding
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | - Chengbiao Wu
- Department of Neurosciences, UCSD, La Jolla, California, USA
| |
Collapse
|
25
|
Toyn JH, Boy KM, Raybon J, Meredith JE, Robertson AS, Guss V, Hoque N, Sweeney F, Zhuo X, Clarke W, Snow K, Denton RR, Zuev D, Thompson LA, Morrison J, Grace J, Berisha F, Furlong M, Wang JS, Lentz KA, Padmanabha R, Cook L, Wei C, Drexler DM, Macor JE, Albright CF, Gasior M, Olson RE, Hong Q, Soares HD, AbuTarif M, Ahlijanian MK. Robust Translation of γ-Secretase Modulator Pharmacology across Preclinical Species and Human Subjects. J Pharmacol Exp Ther 2016; 358:125-37. [PMID: 27189974 PMCID: PMC4931879 DOI: 10.1124/jpet.116.232249] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Accepted: 04/19/2016] [Indexed: 12/20/2022] Open
Abstract
The amyloid-β peptide (Aβ)—in particular, the 42–amino acid form, Aβ1-42—is thought to play a key role in the pathogenesis of Alzheimer’s disease (AD). Thus, several therapeutic modalities aiming to inhibit Aβ synthesis or increase the clearance of Aβ have entered clinical trials, including γ-secretase inhibitors, anti-Aβ antibodies, and amyloid-β precursor protein cleaving enzyme inhibitors. A unique class of small molecules, γ-secretase modulators (GSMs), selectively reduce Aβ1-42 production, and may also decrease Aβ1-40 while simultaneously increasing one or more shorter Aβ peptides, such as Aβ1-38 and Aβ1-37. GSMs are particularly attractive because they do not alter the total amount of Aβ peptides produced by γ-secretase activity; they spare the processing of other γ-secretase substrates, such as Notch; and they do not cause accumulation of the potentially toxic processing intermediate, β-C-terminal fragment. This report describes the translation of pharmacological activity across species for two novel GSMs, (S)-7-(4-fluorophenyl)-N2-(3-methoxy-4-(3-methyl-1H-1,2,4-triazol-1-yl)phenyl)-N4-methyl-6,7-dihydro-5H-cyclopenta[d]pyrimidine-2,4-diamine (BMS-932481) and (S,Z)-17-(4-chloro-2-fluorophenyl)-34-(3-methyl-1H-1,2,4-triazol-1-yl)-16,17-dihydro-15H-4-oxa-2,9-diaza-1(2,4)-cyclopenta[d]pyrimidina-3(1,3)-benzenacyclononaphan-6-ene (BMS-986133). These GSMs are highly potent in vitro, exhibit dose- and time-dependent activity in vivo, and have consistent levels of pharmacological effect across rats, dogs, monkeys, and human subjects. In rats, the two GSMs exhibit similar pharmacokinetics/pharmacodynamics between the brain and cerebrospinal fluid. In all species, GSM treatment decreased Aβ1-42 and Aβ1-40 levels while increasing Aβ1-38 and Aβ1-37 by a corresponding amount. Thus, the GSM mechanism and central activity translate across preclinical species and humans, thereby validating this therapeutic modality for potential utility in AD.
Collapse
Affiliation(s)
- Jeremy H Toyn
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Kenneth M Boy
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Joseph Raybon
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Jere E Meredith
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Alan S Robertson
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Valerie Guss
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Nina Hoque
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Francis Sweeney
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Xiaoliang Zhuo
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Wendy Clarke
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Kimberly Snow
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - R Rex Denton
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Dmitry Zuev
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Lorin A Thompson
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - John Morrison
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - James Grace
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Flora Berisha
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Michael Furlong
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Jun-Sheng Wang
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Kimberly A Lentz
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Ramesh Padmanabha
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Lynda Cook
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Cong Wei
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Dieter M Drexler
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - John E Macor
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Charlie F Albright
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Maciej Gasior
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Richard E Olson
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Quan Hong
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Holly D Soares
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Malaz AbuTarif
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| | - Michael K Ahlijanian
- Yale University, New Haven, Connecticut (J.H.T.); Bristol-Myers Squibb, Wallingford, Connecticut (K.M.B, J.R., Je.E.M., A.S.R., V.G., N.H., F.S., X.Z., W.C., K.S., R.R.D., L.A.T., J.M., J.G., K.A.L., R.P., L.C., D.M.D., C.F.A., R.E.O., M.K.A.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S., C.W.); Cantor Colburn LLP, Hartford, Connecticut (D.Z.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.F.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Bristol-Myers Squibb, Pennington, New Jersey (Jo.E.M., H.D.S., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); and Eisai, Woodcliff Lake, New Jersey (Q.H.)
| |
Collapse
|
26
|
Soares HD, Gasior M, Toyn JH, Wang JS, Hong Q, Berisha F, Furlong MT, Raybon J, Lentz KA, Sweeney F, Zheng N, Akinsanya B, Berman RM, Thompson LA, Olson RE, Morrison J, Drexler DM, Macor JE, Albright CF, Ahlijanian MK, AbuTarif M. The γ-Secretase Modulator, BMS-932481, Modulates Aβ Peptides in the Plasma and Cerebrospinal Fluid of Healthy Volunteers. J Pharmacol Exp Ther 2016; 358:138-50. [PMID: 27189973 PMCID: PMC4931877 DOI: 10.1124/jpet.116.232256] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Accepted: 04/05/2016] [Indexed: 11/22/2022] Open
Abstract
The pharmacokinetics, pharmacodynamics, safety, and tolerability of BMS-932481, a γ-secretase modulator (GSM), were tested in healthy young and elderly volunteers after single and multiple doses. BMS-932481 was orally absorbed, showed dose proportionality after a single dose administration, and had approximately 3-fold accumulation after multiple dosing. High-fat/caloric meals doubled the Cmax and area under the curve and prolonged Tmax by 1.5 hours. Consistent with the preclinical pharmacology of GSMs, BMS-932481 decreased cerebrospinal fluid (CSF) Aβ39, Aβ40, and Aβ42 while increasing Aβ37 and Aβ38, thereby providing evidence of γ-secretase enzyme modulation rather than inhibition. In plasma, reductions in Aβ40 and Aβ42 were observed with no change in total Aβ; in CSF, modest decreases in total Aβ were observed at higher dose levels. Increases in liver enzymes were observed at exposures associated with greater than 70% CSF Aβ42 lowering after multiple dosing. Although further development was halted due to an insufficient safety margin to test the hypothesis for efficacy of Aβ lowering in Alzheimer’s disease, this study demonstrates that γ-secretase modulation is achievable in healthy human volunteers and supports further efforts to discover well tolerated GSMs for testing in Alzheimer’s disease and other indications.
Collapse
Affiliation(s)
- Holly D Soares
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Maciej Gasior
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Jeremy H Toyn
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Jun-Sheng Wang
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Quan Hong
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Flora Berisha
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Michael T Furlong
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Joseph Raybon
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Kimberley A Lentz
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Francis Sweeney
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Naiyu Zheng
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Billy Akinsanya
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Robert M Berman
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Lorin A Thompson
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Richard E Olson
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - John Morrison
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Dieter M Drexler
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - John E Macor
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Charlie F Albright
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Michael K Ahlijanian
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| | - Malaz AbuTarif
- Bristol-Myers Squibb, Lawrence Township, New Jersey (H.D.S., J.R., K.A.L., N.Z., B.A., J.M., J.E.M., M.A.); Teva Pharmaceuticals, Frazer, Pennsylvania (M.G.); Bristol-Myers Squibb, Wallingford, Connecticut (J.H.T., L.A.T., R.E.O., D.M.D., C.F.A., M.K.A.); GSK Consumer Healthcare, Parsippany, New Jersey (J.-S.W.); Eisai, Woodcliff Lake, New Jersey (Q.H.); Kyowa Hakko Kirin Pharma, Princeton, New Jersey (F.B.); FORUM Pharmaceuticals, Waltham, Massachusetts (M.T.F.); Pfizer Worldwide Research and Development, Groton, Connecticut (F.S.); and Biohaven Medical Services, New Haven, Connecticut (R.M.B.)
| |
Collapse
|
27
|
Kumar R, Juillerat-Jeanneret L, Golshayan D. Notch Antagonists: Potential Modulators of Cancer and Inflammatory Diseases. J Med Chem 2016; 59:7719-37. [DOI: 10.1021/acs.jmedchem.5b01516] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Rajesh Kumar
- Transplantation
Center and Transplantation Immunopathology Laboratory, Department
of Medicine and ‡University Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| | - Lucienne Juillerat-Jeanneret
- Transplantation
Center and Transplantation Immunopathology Laboratory, Department
of Medicine and ‡University Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| | - Dela Golshayan
- Transplantation
Center and Transplantation Immunopathology Laboratory, Department
of Medicine and ‡University Institute of Pathology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), CH-1011 Lausanne, Switzerland
| |
Collapse
|
28
|
Canalis E, Schilling L, Yee SP, Lee SK, Zanotti S. Hajdu Cheney Mouse Mutants Exhibit Osteopenia, Increased Osteoclastogenesis, and Bone Resorption. J Biol Chem 2015; 291:1538-1551. [PMID: 26627824 DOI: 10.1074/jbc.m115.685453] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Indexed: 11/06/2022] Open
Abstract
Notch receptors are determinants of cell fate and function and play a central role in skeletal development and bone remodeling. Hajdu Cheney syndrome, a disease characterized by osteoporosis and fractures, is associated with NOTCH2 mutations resulting in a truncated stable protein and gain-of-function. We created a mouse model reproducing the Hajdu Cheney syndrome by introducing a 6955C→T mutation in the Notch2 locus leading to a Q2319X change at the amino acid level. Notch2(Q2319X) heterozygous mutants were smaller and had shorter femurs than controls; and at 1 month of age they exhibited cancellous and cortical bone osteopenia. As the mice matured, cancellous bone volume was restored partially in male but not female mice, whereas cortical osteopenia persisted in both sexes. Cancellous bone histomorphometry revealed an increased number of osteoclasts and bone resorption, without a decrease in osteoblast number or bone formation. Osteoblast differentiation and function were not affected in Notch2(Q2319X) cells. The pre-osteoclast cell pool, osteoclast differentiation, and bone resorption in response to receptor activator of nuclear factor κB ligand in vitro were increased in Notch2(Q2319X) mutants. These effects were suppressed by the γ-secretase inhibitor LY450139. In conclusion, Notch2(Q2319X) mice exhibit cancellous and cortical bone osteopenia, enhanced osteoclastogenesis, and increased bone resorption.
Collapse
Affiliation(s)
| | | | - Siu-Pok Yee
- Cell Biology, Genetics, and; Genome Sciences Biology
| | - Sun-Kyeong Lee
- Medicine,; Center on Aging, University of Connecticut Health Center, Farmington, Connecticut 06030
| | | |
Collapse
|
29
|
Abstract
Electron microscopy has enlarged the visual horizons of the morphological alterations in Alzheimer's disease (AD). Study of the mitochondria and Golgi apparatus in early cases of AD revealed the principal role that these important organelles play in the drama of pathogenic dialog of AD, substantially affecting energy production and supply, and protein trafficking in neurons and glia. In addition, study of the morphological alterations of the dendritic arbor, dendritic spines and neuronal synapses, which are associated with mitochondrial damage, may reasonably interpret the clinical phenomena of the irreversible decline of the mental faculties and an individual's personality changes. Electron microscopy also reveals the involvement of microvascular alterations in the etiopathogenic background of AD.
Collapse
|
30
|
Potent benzoazepinone γ-secretase modulators with improved bioavailability. Bioorg Med Chem Lett 2015; 25:3495-500. [DOI: 10.1016/j.bmcl.2015.06.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2015] [Revised: 06/04/2015] [Accepted: 06/08/2015] [Indexed: 11/22/2022]
|
31
|
Cleavage of amyloid precursor protein by an archaeal presenilin homologue PSH. Proc Natl Acad Sci U S A 2015; 112:3344-9. [PMID: 25733893 DOI: 10.1073/pnas.1502150112] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Aberrant cleavage of amyloid precursor protein (APP) by γ-secretase contributes to the development of Alzheimer's disease. More than 200 disease-derived mutations have been identified in presenilin (the catalytic subunit of γ-secretase), making modulation of γ-secretase activity a potentially attractive therapeutic opportunity. Unfortunately, the technical challenges in dealing with intact γ-secretase have hindered discovery of modulators and demand a convenient substitute approach. Here we report that, similar to γ-secretase, the archaeal presenilin homolog PSH faithfully processes the substrate APP C99 into Aβ42, Aβ40, and Aβ38. The molar ratio of the cleavage products Aβ42 over Aβ40 by PSH is nearly identical to that by γ-secretase. The proteolytic activity of PSH is specifically suppressed by presenilin-specific inhibitors. Known modulators of γ-secretase also modulate PSH similarly in terms of the Aβ42/Aβ40 ratio. Structural analysis reveals association of a known γ-secretase inhibitor with PSH between its two catalytic aspartate residues. These findings identify PSH as a surrogate protease for the screening of agents that may regulate the protease activity and the cleavage preference of γ-secretase.
Collapse
|
32
|
Weissmiller AM, Natera-Naranjo O, Reyna SM, Pearn ML, Zhao X, Nguyen P, Cheng S, Goldstein LSB, Tanzi RE, Wagner SL, Mobley WC, Wu C. A γ-secretase inhibitor, but not a γ-secretase modulator, induced defects in BDNF axonal trafficking and signaling: evidence for a role for APP. PLoS One 2015; 10:e0118379. [PMID: 25710492 PMCID: PMC4339551 DOI: 10.1371/journal.pone.0118379] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/14/2015] [Indexed: 11/19/2022] Open
Abstract
Clues to Alzheimer disease (AD) pathogenesis come from a variety of different sources including studies of clinical and neuropathological features, biomarkers, genomics and animal and cellular models. An important role for amyloid precursor protein (APP) and its processing has emerged and considerable interest has been directed at the hypothesis that Aβ peptides induce changes central to pathogenesis. Accordingly, molecules that reduce the levels of Aβ peptides have been discovered such as γ-secretase inhibitors (GSIs) and modulators (GSMs). GSIs and GSMs reduce Aβ levels through very different mechanisms. However, GSIs, but not GSMs, markedly increase the levels of APP CTFs that are increasingly viewed as disrupting neuronal function. Here, we evaluated the effects of GSIs and GSMs on a number of neuronal phenotypes possibly relevant to their use in treatment of AD. We report that GSI disrupted retrograde axonal trafficking of brain-derived neurotrophic factor (BDNF), suppressed BDNF-induced downstream signaling pathways and induced changes in the distribution within neuronal processes of mitochondria and synaptic vesicles. In contrast, treatment with a novel class of GSMs had no significant effect on these measures. Since knockdown of APP by specific siRNA prevented GSI-induced changes in BDNF axonal trafficking and signaling, we concluded that GSI effects on APP processing were responsible, at least in part, for BDNF trafficking and signaling deficits. Our findings argue that with respect to anti-amyloid treatments, even an APP-specific GSI may have deleterious effects and GSMs may serve as a better alternative.
Collapse
Affiliation(s)
- April M. Weissmiller
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - Orlangie Natera-Naranjo
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - Sol M. Reyna
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, United States of America
| | - Matthew L. Pearn
- Department of Anesthesiology, University of California San Diego, San Diego, California, United States of America
- V.A. San Diego Healthcare System, San Diego, California, United States of America
| | - Xiaobei Zhao
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - Phuong Nguyen
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - Soan Cheng
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - Lawrence S. B. Goldstein
- Department of Cellular and Molecular Medicine, University of California San Diego, San Diego, California, United States of America
| | - Rudolph E. Tanzi
- Genetics and Aging Research Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, Massachusetts, United States of America
| | - Steven L. Wagner
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - William C. Mobley
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| | - Chengbiao Wu
- Department of Neurosciences, University of California San Diego, San Diego, California, United States of America
| |
Collapse
|
33
|
Zhang X, Li Y, Xu H, Zhang YW. The γ-secretase complex: from structure to function. Front Cell Neurosci 2014; 8:427. [PMID: 25565961 PMCID: PMC4263104 DOI: 10.3389/fncel.2014.00427] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 11/27/2014] [Indexed: 12/21/2022] Open
Abstract
One of the most critical pathological features of Alzheimer’s disease (AD) is the accumulation of β-amyloid (Aβ) peptides that form extracellular senile plaques in the brain. Aβ is derived from β-amyloid precursor protein (APP) through sequential cleavage by β- and γ-secretases. γ-secretase is a high molecular weight complex minimally composed of four components: presenilins (PS), nicastrin, anterior pharynx defective 1 (APH-1), and presenilin enhancer 2 (PEN-2). In addition to APP, γ-secretase also cleaves many other type I transmembrane (TM) protein substrates. As a crucial enzyme for Aβ production, γ-secretase is an appealing therapeutic target for AD. Here, we summarize current knowledge on the structure and function of γ-secretase, as well as recent progress in developing γ-secretase targeting drugs for AD treatment.
Collapse
Affiliation(s)
- Xian Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| | - Yanfang Li
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| | - Huaxi Xu
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China ; Degenerative Disease Research Program, Sanford-Burnham Medical Research Institute La Jolla, CA, USA
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, College of Medicine, Xiamen University Xiamen, FJ, China
| |
Collapse
|
34
|
Strömberg K, Eketjäll S, Georgievska B, Tunblad K, Eliason K, Olsson F, Radesäter AC, Klintenberg R, Arvidsson PI, von Berg S, Fälting J, Cowburn RF, Dabrowski M. Combining an amyloid-beta (Aβ) cleaving enzyme inhibitor with a γ-secretase modulator results in an additive reduction of Aβ production. FEBS J 2014; 282:65-73. [DOI: 10.1111/febs.13103] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 07/31/2014] [Accepted: 10/06/2014] [Indexed: 12/27/2022]
Affiliation(s)
- Kia Strömberg
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Susanna Eketjäll
- AstraZeneca Translational Science Centre; Science for Life Laboratory; Solna Sweden
| | - Biljana Georgievska
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Karin Tunblad
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Kristina Eliason
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Fredrik Olsson
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | | | - Rebecka Klintenberg
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Per I. Arvidsson
- Project Management, AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Stefan von Berg
- Department of Medicinal Chemistry; R&I iMed; AstraZeneca R&D Mölndal; Sweden
| | - Johanna Fälting
- Project Management, AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Richard F. Cowburn
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| | - Michael Dabrowski
- Department of Neuroscience; AstraZeneca R&D; CNS/Pain iMED; Södertälje Sweden
| |
Collapse
|
35
|
Toyn JH, Thompson LA, Lentz KA, Meredith JE, Burton CR, Sankaranararyanan S, Guss V, Hall T, Iben LG, Krause CM, Krause R, Lin XA, Pierdomenico M, Polson C, Robertson AS, Denton RR, Grace JE, Morrison J, Raybon J, Zhuo X, Snow K, Padmanabha R, Agler M, Esposito K, Harden D, Prack M, Varma S, Wong V, Zhu Y, Zvyaga T, Gerritz S, Marcin LR, Higgins MA, Shi J, Wei C, Cantone JL, Drexler DM, Macor JE, Olson RE, Ahlijanian MK, Albright CF. Identification and Preclinical Pharmacology of the γ-Secretase Modulator BMS-869780. Int J Alzheimers Dis 2014; 2014:431858. [PMID: 25097793 PMCID: PMC4109680 DOI: 10.1155/2014/431858] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2014] [Accepted: 05/18/2014] [Indexed: 01/13/2023] Open
Abstract
Alzheimer's disease is the most prevalent cause of dementia and is associated with accumulation of amyloid-β peptide (Aβ), particularly the 42-amino acid Aβ1-42, in the brain. Aβ1-42 levels can be decreased by γ-secretase modulators (GSM), which are small molecules that modulate γ-secretase, an enzyme essential for Aβ production. BMS-869780 is a potent GSM that decreased Aβ1-42 and Aβ1-40 and increased Aβ1-37 and Aβ1-38, without inhibiting overall levels of Aβ peptides or other APP processing intermediates. BMS-869780 also did not inhibit Notch processing by γ-secretase and lowered brain Aβ1-42 without evidence of Notch-related side effects in rats. Human pharmacokinetic (PK) parameters were predicted through allometric scaling of PK in rat, dog, and monkey and were combined with the rat pharmacodynamic (PD) parameters to predict the relationship between BMS-869780 dose, exposure and Aβ1-42 levels in human. Off-target and safety margins were then based on comparisons to the predicted exposure required for robust Aβ1-42 lowering. Because of insufficient safety predictions and the relatively high predicted human daily dose of 700 mg, further evaluation of BMS-869780 as a potential clinical candidate was discontinued. Nevertheless, BMS-869780 demonstrates the potential of the GSM approach for robust lowering of brain Aβ1-42 without Notch-related side effects.
Collapse
Affiliation(s)
- Jeremy H. Toyn
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Lorin A. Thompson
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Kimberley A. Lentz
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Jere E. Meredith
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Catherine R. Burton
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Sethu Sankaranararyanan
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Valerie Guss
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Tracey Hall
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
- Preclinical Sciences, Alexion Pharmaceuticals, Inc 352 Knotter Drive, Cheshire, CT 06410, USA
| | - Lawrence G. Iben
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Carol M. Krause
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Rudy Krause
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Xu-Alan Lin
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Maria Pierdomenico
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Craig Polson
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Alan S. Robertson
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - R. Rex Denton
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - James E. Grace
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - John Morrison
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Joseph Raybon
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Xiaoliang Zhuo
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Kimberly Snow
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Ramesh Padmanabha
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Michele Agler
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
- High Throughput Biology, Boehringer Ingelheim, 900 Ridgebury Road, Ridgefield, CT 06877, USA
| | - Kim Esposito
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - David Harden
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Margaret Prack
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Sam Varma
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
- Stratford High School, 45 North Parade, Stratford, CT 06615, USA
| | - Victoria Wong
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
- External Research Solutions, WWMC, Pfizer World Wide Research & Development, Eastern Point Road, Groton, CT 06340, USA
| | - Yingjie Zhu
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
- Arvinas Inc, 5 Science Park, New Haven, CT 06511, USA
| | - Tatyana Zvyaga
- Lead Discovery and Lead Profiling, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Samuel Gerritz
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Lawrence R. Marcin
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Mendi A. Higgins
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Jianliang Shi
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Cong Wei
- Discovery Analytical Sciences, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer World Wide Research & Development, Eastern Point Road, Groton, CT 06340, USA
| | - Joseph L. Cantone
- Discovery Analytical Sciences, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Dieter M. Drexler
- Discovery Analytical Sciences, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - John E. Macor
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Richard E. Olson
- Discovery Chemistry, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Michael K. Ahlijanian
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| | - Charles F. Albright
- Exploratory Biology and Genomics, Bristol-Myers Squibb Research and Development, 5 Research Parkway, Wallingford, CT 06492, USA
| |
Collapse
|
36
|
Karran E, Hardy J. A critique of the drug discovery and phase 3 clinical programs targeting the amyloid hypothesis for Alzheimer disease. Ann Neurol 2014; 76:185-205. [PMID: 24853080 PMCID: PMC4204160 DOI: 10.1002/ana.24188] [Citation(s) in RCA: 196] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 05/19/2014] [Accepted: 05/19/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Eric Karran
- Alzheimer's Research UK, Cambridge; Reta Lila Weston Laboratories, London; Department of Molecular Neuroscience, University College London, London, United Kingdom
| | | |
Collapse
|
37
|
Loss of presenilin 2 is associated with increased iPLA2 activity and lung tumor development. Oncogene 2014; 33:5193-200. [PMID: 24858037 PMCID: PMC4287650 DOI: 10.1038/onc.2014.128] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/11/2014] [Accepted: 04/14/2014] [Indexed: 12/17/2022]
Abstract
Presenilins are the enzymatic components of γ-secretase complex that cleaves amyloid precursor protein, Notch and β-catenin, which has critical roles in the development of Alzheimer's disease and cancer cell growth. Therefore, in the present study, we studied the effects and mechanisms of PS2 knockout on lung cancer development and possible mechanisms as a key regulator of lung tumor development. We compared carcinogen-induced tumor growth between PS2 knockout mice and wild-type mice. PS2 knockout mice showed increased urethane (1 mg/g)-induced lung tumor incidence when compared with that of wild-type mice with decreased activity of γ-secretase in the lung tumor tissues. Consequently, iPLA2 activities in lung tumor tissues of PS2 knockout mice were much higher than in tumor tissues of wild-type mice. Furthermore, knockdown of PS2 using PS2 siRNA decreased γ-secretase activity with increased iPLA2 activity in the lung cancer cells (A549 and NCI-H460), leading to increased lung cancer cell growth. PS2 knockout mice and PS2 knockdown lung cancer cells showed increased DNA-binding activities of nuclear factor kappa-beta, signal transducer and activator of transcription 3 (STAT3) and AP-1 which are critical transcriptional factors of iPLA2 than those of PS2 wild-type mice and control lung cancer cells. Taken together, these results suggest that the loss of PS2 could have a critical role in lung tumor development through the upregulation of iPLA2 activity by reducing γ-secretase.
Collapse
|
38
|
Pettersson M, Johnson DS, Subramanyam C, Bales KR, am Ende CW, Fish BA, Green ME, Kauffman GW, Mullins PB, Navaratnam T, Sakya SM, Stiff CM, Tran TP, Xie L, Zhang L, Pustilnik LR, Vetelino BC, Wood KM, Pozdnyakov N, Verhoest PR, O’Donnell CJ. Design, Synthesis, and Pharmacological Evaluation of a Novel Series of Pyridopyrazine-1,6-dione γ-Secretase Modulators. J Med Chem 2014; 57:1046-62. [DOI: 10.1021/jm401782h] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Martin Pettersson
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Douglas S. Johnson
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Chakrapani Subramanyam
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kelly R. Bales
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Christopher W. am Ende
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Benjamin A. Fish
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Michael E. Green
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Gregory W. Kauffman
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Patrick B. Mullins
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Thayalan Navaratnam
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Subas M. Sakya
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Cory M. Stiff
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Tuan P. Tran
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Longfei Xie
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Liming Zhang
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Leslie R. Pustilnik
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Beth C. Vetelino
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Kathleen M. Wood
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Nikolay Pozdnyakov
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Patrick R. Verhoest
- Pfizer Worldwide Research & Development, 610 Main Street, Cambridge, Massachusetts 02139, United States
| | - Christopher J. O’Donnell
- Pfizer Worldwide Research & Development, Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
39
|
Evin G, Barakat A. Critical analysis of the use of β-site amyloid precursor protein-cleaving enzyme 1 inhibitors in the treatment of Alzheimer's disease. Degener Neurol Neuromuscul Dis 2014; 4:1-19. [PMID: 32669897 PMCID: PMC7337240 DOI: 10.2147/dnnd.s41056] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 03/06/2014] [Indexed: 01/18/2023] Open
Abstract
Alzheimer’s disease (AD) is the major cause of dementia in the elderly and an unmet clinical challenge. A variety of therapies that are currently under development are directed to the amyloid cascade. Indeed, the accumulation and toxicity of amyloid-β (Aβ) is believed to play a central role in the etiology of the disease, and thus rational interventions are aimed at reducing the levels of Aβ in the brain. Targeting β-site amyloid precursor protein-cleaving enzyme (BACE)-1 represents an attractive strategy, as this enzyme catalyzes the initial and rate-limiting step in Aβ production. Observation of increased levels of BACE1 and enzymatic activity in the brain, cerebrospinal fluid, and platelets of patients with AD and mild cognitive impairment supports the potential benefits of BACE1 inhibition. Numerous potent inhibitors have been generated, and many of these have been proved to lower Aβ levels in the brain of animal models. Over 10 years of intensive research on BACE1 inhibitors has now culminated in advancing half a dozen of these drugs into human trials, yet translating the in vitro and cellular efficacy of BACE1 inhibitors into preclinical and clinical trials represents a challenge. This review addresses the promises and also the potential problems associated with BACE1 inhibitors for AD therapy, as the complex biological function of BACE1 in the brain is becoming unraveled.
Collapse
Affiliation(s)
- Genevieve Evin
- Oxidation Biology Laboratory, Mental Health Research Institute, Florey Institute of Neuroscience and Mental Health, University of Melbourne.,Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| | - Adel Barakat
- Department of Pathology, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
40
|
Cudaback E, Jorstad NL, Yang Y, Montine TJ, Keene CD. Therapeutic implications of the prostaglandin pathway in Alzheimer's disease. Biochem Pharmacol 2014; 88:565-72. [PMID: 24434190 DOI: 10.1016/j.bcp.2013.12.014] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Revised: 12/18/2013] [Accepted: 12/18/2013] [Indexed: 11/19/2022]
Abstract
An important pathologic hallmark of Alzheimer's disease (AD) is neuroinflammation, a process characterized in AD by disproportionate activation of cells (microglia and astrocytes, primarily) of the non-specific innate immune system within the CNS. While inflammation itself is not intrinsically detrimental, a delicate balance of pro- and anti-inflammatory signals must be maintained to ensure that long-term exaggerated responses do not damage the brain over time. Non-steroidal anti-inflammatory drugs (NSAIDs) represent a broad class of powerful therapeutics that temper inflammation by inhibiting cyclooxygenase-mediated signaling pathways including prostaglandins, which are the principal mediators of CNS neuroinflammation. While historically used to treat discrete or systemic inflammatory conditions, epidemiologic evidence suggests that protracted NSAID use may delay AD onset, as well as decrease disease severity and rate of progression. Unfortunately, clinical trials with NSAIDs have thus far yielded disappointing results, including premature discontinuation of a large-scale prevention trial due to unexpected cardiovascular side effects. Here we review the literature and make the argument that more targeted exploitation of downstream prostaglandin signaling pathways may offer significant therapeutic benefits for AD while minimizing adverse side effects. Directed strategies such as these may ultimately help to delay the deleterious consequences of brain aging and might someday lead to new therapies for AD and other chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Eiron Cudaback
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - Nikolas L Jorstad
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - Yue Yang
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - Thomas J Montine
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA
| | - C Dirk Keene
- University of Washington Harborview Medical Center, Department of Pathology, Box 359791, 325 Ninth Ave, Seattle, WA 98104, USA.
| |
Collapse
|
41
|
Yu Y, Logovinsky V, Schuck E, Kaplow J, Chang MK, Miyagawa T, Wong N, Ferry J. Safety, tolerability, pharmacokinetics, and pharmacodynamics of the novel γ-secretase modulator, E2212, in healthy human subjects. J Clin Pharmacol 2014; 54:528-36. [PMID: 24343761 DOI: 10.1002/jcph.249] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 12/09/2013] [Indexed: 01/19/2023]
Abstract
E2212, a novel γ-secretase modulator, is under development for the treatment of Alzheimer's disease. The safety, tolerability, pharmacokinetics, and pharmacodynamics of single ascending oral doses (10-250 mg, double-blind, placebo-controlled, randomized) of E2212 were evaluated. In this phase I clinical trial, E2212 was found to be well tolerated in single doses. Maximum tolerated dose was not achieved up to 250 mg. Most AEs were mild to moderate in severity with no identifiable dose related pattern. There were no clinically significant findings on physical and ophthalmologic examinations as well as vital signs, laboratory, ECG and C-SSRS assessments. E2212 was rapidly absorbed, with median tmax values ranging from 0.5 to 1.0 h. E2212 exhibited biphasic disposition with the terminal t1/2 of 12.5-19.0 h. Renal excretion was the minor pathway for E2212 elimination. Increased PD response (reduction in plasma concentrations of Aβ(x-42)) was observed with increasing doses. The maximum PD response was observed in the highest dose 250 mg cohort, with ΔAUAC(0-24 h) of 44.1% and Amax of 53.6%. These results support further clinical development of E2212.
Collapse
Affiliation(s)
- Yanke Yu
- Drug Metabolism and Pharmacokinetics, Eisai Inc., Andover, MA, USA
| | | | - Edgar Schuck
- Drug Metabolism and Pharmacokinetics, Eisai Inc., Andover, MA, USA
| | - June Kaplow
- Department of Biostatistics, Eisai Inc., Woodcliff Lake, NJ, USA
| | - Min-Kun Chang
- Department of Biostatistics, Eisai Inc., Woodcliff Lake, NJ, USA
| | | | - Nancy Wong
- Drug Metabolism and Pharmacokinetics, Eisai Inc., Andover, MA, USA
| | - Jim Ferry
- Department of Clinical Pharmacology, Eisai Inc., Woodcliff Lake, NJ, USA
| |
Collapse
|
42
|
Olsson F, Schmidt S, Althoff V, Munter LM, Jin S, Rosqvist S, Lendahl U, Multhaup G, Lundkvist J. Characterization of intermediate steps in amyloid beta (Aβ) production under near-native conditions. J Biol Chem 2013; 289:1540-50. [PMID: 24225948 DOI: 10.1074/jbc.m113.498246] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Processing of the amyloid precursor protein (APP) by γ-secretase results in generation of Aβ peptides of different lengths ranging from 51 to 30 residues. Accumulation of Aβ and in particular Aβ42 is enhanced by familial Alzheimer disease (FAD) causing mutations in APP and is believed to play a pivotal role. The molecular mechanism underlying normal Aβ production, the impact of FAD mutations on this process and how anti-amyloidogenic γ-secretase modulators (GSMs) cause a selective decrease in Aβ40 and Aβ42 and an increase in shorter Aβ peptides, however, is poorly understood. By using a combined immuno- and LC-MS-based assay we identify several major intermediates, i.e. 3- and 4-peptides that line up head to head across the entire APP transmembrane sequence from Aβ51 to Aβ31/Aβ30 and from Aβ49 to Aβ30/31. FAD APP mutations displayed a relative increase in 3- and 4-peptides from Aβ48 to Aβ38 compared with Aβ49 to Aβ37. These findings correlate with an increase in the Aβ42/40 ratio. GSMs caused a decrease in Aβ40 and Aβ42 and an increase in Aβ37 and Aβ38 paralleled by an increase of the intermediates Aβ40-38 and Aβ42-39. Collectively, these data provide a thorough characterization of all intermediate steps in Aβ production in native cell membranes and provide key mechanistic insights to genetic and pharmacological modulation of Aβ generation.
Collapse
Affiliation(s)
- Fredrik Olsson
- From the AstraZeneca iMED CNS/Pain, 15185 Södertälje, Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ginman T, Viklund J, Malmström J, Blid J, Emond R, Forsblom R, Johansson A, Kers A, Lake F, Sehgelmeble F, Sterky KJ, Bergh M, Lindgren A, Johansson P, Jeppsson F, Fälting J, Gravenfors Y, Rahm F. Core refinement toward permeable β-secretase (BACE-1) inhibitors with low hERG activity. J Med Chem 2013; 56:4181-205. [PMID: 23126626 DOI: 10.1021/jm3011349] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
By use of iterative design aided by predictive models for target affinity, brain permeability, and hERG activity, novel and diverse compounds based on cyclic amidine and guanidine cores were synthesized with the goal of finding BACE-1 inhibitors as a treatment for Alzheimer's disease. Since synthesis feasibility had low priority in the design of the cores, an extensive synthesis effort was needed to make the relevant compounds. Syntheses of these compounds are reported, together with physicochemical properties and structure-activity relationships based on in vitro data. Four crystal structures of diverse amidines binding in the active site are deposited and discussed. Inhibitors of BACE-1 with 3 μM to 32 nM potencies in cells are shown, together with data on in vivo brain exposure levels for four compounds. The results presented show the importance of the core structure for the profile of the final compounds.
Collapse
Affiliation(s)
- Tobias Ginman
- Department of Medicinal Chemistry, AstraZeneca R&D Södertälje, SE-151 85, Södertälje, Sweden
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Crump CJ, Johnson DS, Li YM. Development and mechanism of γ-secretase modulators for Alzheimer's disease. Biochemistry 2013; 52:3197-216. [PMID: 23614767 DOI: 10.1021/bi400377p] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
γ-Secretase is an aspartyl intramembranal protease composed of presenilin, Nicastrin, Aph1, and Pen2 with 19 transmembrane domains. γ-Secretase cleaves the amyloid precursor proteins (APP) to release Aβ peptides that likely play a causative role in the pathogenesis of Alzheimer's disease (AD). In addition, γ-secretase cleaves Notch and other type I membrane proteins. γ-Secretase inhibitors (GSIs) have been developed and used for clinical studies. However, clinical trials have shown adverse effects of GSIs that are potentially linked with nondiscriminatory inhibition of Notch signaling, overall APP processing, and other substrate cleavages. Therefore, these findings call for the development of disease-modifying agents that target γ-secretase activity to lower levels of Aβ42 production without blocking the overall processing of γ-secretase substrates. γ-Secretase modulators (GSMs) originally derived from nonsteroidal anti-inflammatory drugs (NSAIDs) display such characteristics and are the focus of this review. However, first-generation GSMs have limited potential because of the low potency and undesired neuropharmacokinetic properties. This generation of GSMs has been suggested to interact with the APP substrate, γ-secretase, or both. To improve the potency and brain availability, second-generation GSMs, including NSAID-derived carboxylic acid and non-NSAID-derived heterocyclic chemotypes, as well as natural product-derived GSMs have been developed. Animal studies of this generation of GSMs have shown encouraging preclinical profiles. Moreover, using potent GSM photoaffinity probes, multiple studies unambiguously have showed that both carboxylic acid and heterocyclic GSMs specifically target presenilin, the catalytic subunit of γ-secretase. In addition, two types of GSMs have distinct binding sites within the γ-secretase complex and exhibit different Aβ profiles. GSMs induce a conformational change of γ-secretase to achieve modulation. Various models are proposed and discussed. Despite the progress of GSM research, many outstanding issues remain to be investigated to achieve the ultimate goal of developing GSMs as effective AD therapies.
Collapse
Affiliation(s)
- Christina J Crump
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center , 1275 York Avenue, New York, New York 10065, United States
| | | | | |
Collapse
|
45
|
Suspected limited efficacy of γ-secretase modulators. Neurobiol Aging 2013; 34:1101-4. [DOI: 10.1016/j.neurobiolaging.2012.08.017] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2012] [Accepted: 08/25/2012] [Indexed: 11/20/2022]
|
46
|
Alzheimer's disease: presenilin 2-sparing γ-secretase inhibition is a tolerable Aβ peptide-lowering strategy. J Neurosci 2013. [PMID: 23197721 DOI: 10.1523/jneurosci.1451-12.2012] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
γ-Secretase inhibition represents a major therapeutic strategy for lowering amyloid β (Aβ) peptide production in Alzheimer's disease (AD). Progress toward clinical use of γ-secretase inhibitors has, however, been hampered due to mechanism-based adverse events, primarily related to impairment of Notch signaling. The γ-secretase inhibitor MRK-560 represents an exception as it is largely tolerable in vivo despite displaying only a small selectivity between Aβ production and Notch signaling in vitro. In exploring the molecular basis for the observed tolerability, we show that MRK-560 displays a strong preference for the presenilin 1 (PS1) over PS2 subclass of γ-secretases and is tolerable in wild-type mice but causes dose-dependent Notch-related side effect in PS2-deficient mice at drug exposure levels resulting in a substantial decrease in brain Aβ levels. This demonstrates that PS2 plays an important role in mediating essential Notch signaling in several peripheral organs during pharmacological inhibition of PS1 and provide preclinical in vivo proof of concept for PS2-sparing inhibition as a novel, tolerable and efficacious γ-secretase targeting strategy for AD.
Collapse
|
47
|
Pozdnyakov N, Murrey HE, Crump CJ, Pettersson M, Ballard TE, Am Ende CW, Ahn K, Li YM, Bales KR, Johnson DS. γ-Secretase modulator (GSM) photoaffinity probes reveal distinct allosteric binding sites on presenilin. J Biol Chem 2013; 288:9710-9720. [PMID: 23396974 DOI: 10.1074/jbc.m112.398602] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
γ-Secretase is an intramembrane aspartyl protease that cleaves the amyloid precursor protein to produce neurotoxic β-amyloid peptides (i.e. Aβ42) that have been implicated in the pathogenesis of Alzheimer disease. Small molecule γ-secretase modulators (GSMs) have emerged as potential disease-modifying treatments for Alzheimer disease because they reduce the formation of Aβ42 while not blocking the processing of γ-secretase substrates. We developed clickable GSM photoaffinity probes with the goal of identifying the target of various classes of GSMs and to better understand their mechanism of action. Here, we demonstrate that the photoaffinity probe E2012-BPyne specifically labels the N-terminal fragment of presenilin-1 (PS1-NTF) in cell membranes as well as in live cells and primary neuronal cultures. The labeling is competed in the presence of the parent imidazole GSM E2012, but not with acid GSM-1, allosteric GSI BMS-708163, or substrate docking site peptide inhibitor pep11, providing evidence that these compounds have distinct binding sites. Surprisingly, we found that the cross-linking of E2012-BPyne to PS1-NTF is significantly enhanced in the presence of the active site-directed GSI L-685,458 (L458). In contrast, L458 does not affect the labeling of the acid GSM photoprobe GSM-5. We also observed that E2012-BPyne specifically labels PS1-NTF (active γ-secretase) but not full-length PS1 (inactive γ-secretase) in ANP.24 cells. Taken together, our results support the hypothesis that multiple binding sites within the γ-secretase complex exist, each of which may contribute to different modes of modulatory action. Furthermore, the enhancement of PS1-NTF labeling by E2012-BPyne in the presence of L458 suggests a degree of cooperativity between the active site of γ-secretase and the modulatory binding site of certain GSMs.
Collapse
Affiliation(s)
- Nikolay Pozdnyakov
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Heather E Murrey
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - Christina J Crump
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; Department of Pharmacology, Weill Graduate School of Medical Science of Cornell University, New York, New York 10021
| | - Martin Pettersson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | - T Eric Ballard
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139
| | | | - Kwangwook Ahn
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; Department of Pharmacology, Weill Graduate School of Medical Science of Cornell University, New York, New York 10021
| | - Yue-Ming Li
- Molecular Pharmacology and Chemistry Program, Memorial Sloan-Kettering Cancer Center, New York, New York 10065; Department of Pharmacology, Weill Graduate School of Medical Science of Cornell University, New York, New York 10021
| | - Kelly R Bales
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139.
| | - Douglas S Johnson
- Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139.
| |
Collapse
|
48
|
Ousson S, Saric A, Baguet A, Losberger C, Genoud S, Vilbois F, Permanne B, Hussain I, Beher D. Substrate determinants in the C99 juxtamembrane domains differentially affect γ-secretase cleavage specificity and modulator pharmacology. J Neurochem 2013; 125:610-9. [PMID: 23253155 DOI: 10.1111/jnc.12129] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2012] [Revised: 12/12/2012] [Accepted: 12/13/2012] [Indexed: 11/29/2022]
Abstract
The molecular mechanisms governing γ-secretase cleavage specificity are not fully understood. Herein, we demonstrate that extending the transmembrane domain of the amyloid precursor protein-derived C99 substrate in proximity to the cytosolic face strongly influences γ-secretase cleavage specificity. Sequential insertion of leucines or replacement of membrane-anchoring lysines by leucines elevated the production of Aβ42, whilst lowering production of Aβ40. A single insertion or replacement was sufficient to produce this phenotype, suggesting that the helical length distal to the ε-site is a critical determinant of γ-secretase cleavage specificity. Replacing the lysine at the luminal membrane border (K28) with glutamic acid (K28E) increased Aβ37 and reduced Aβ42 production. Maintaining a positive charge with an arginine replacement, however, did not alter cleavage specificity. Using two potent and structurally distinct γ-secretase modulators (GSMs), we elucidated the contribution of K28 to the modulatory mechanism. Surprisingly, whilst lowering the potency of the non-steroidal anti-inflammatory drug-type GSM, the K28E mutation converted a heteroaryl-type GSM to an inverse GSM. This result implies the proximal lysine is critical for the GSM mechanism and pharmacology. This region is likely a major determinant for substrate binding and we speculate that modulation of substrate binding is the fundamental mechanism by which GSMs exert their action.
Collapse
Affiliation(s)
- Solenne Ousson
- Global Research and Early Development, Merck Serono SA, Geneva, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Hochard A, Oumata N, Bettayeb K, Gloulou O, Fant X, Durieu E, Buron N, Porceddu M, Borgne-Sanchez A, Galons H, Flajolet M, Meijer L. Aftins increase amyloid-β42, lower amyloid-β38, and do not alter amyloid-β40 extracellular production in vitro: toward a chemical model of Alzheimer's disease? J Alzheimers Dis 2013; 35:107-20. [PMID: 23364140 PMCID: PMC5039020 DOI: 10.3233/jad-121777] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Increased production of amyloid-β (Aβ)42 peptide, derived from the amyloid-β protein precursor, and its subsequent aggregation into oligomers and plaques constitutes a hallmark of Alzheimer's disease (AD). We here report on a family of low molecular weight molecules, the Aftins (Amyloid-β Forty-Two Inducers), which, in cultured cells, dramatically affect the production of extracellular/secreted amyloid peptides. Aftins trigger β-secretase inhibitor and γ-secretase inhibitors (GSIs) sensitive, robust upregulation of Aβ42, and parallel down-regulation of Aβ38, while Aβ40 levels remain stable. In contrast, intracellular levels of these amyloids appear to remain stable. In terms of their effects on Aβ38/Aβ40/Aβ42 relative abundance, Aftins act opposite to γ-secretase modulators (GSMs). Aβ42 upregulation induced by Aftin-5 is unlikely to originate from reduced proteolytic degradation or diminished autophagy. Aftin-5 has little effects on mitochondrial functional parameters (swelling, transmembrane potential loss, cytochrome c release, oxygen consumption) but reversibly alters the ultrastructure of mitochondria. Aftins thus alter the Aβ levels in a fashion similar to that described in the brain of AD patients. Aftins therefore constitute new pharmacological tools to investigate this essential aspect of AD, in cell cultures, allowing (1) the detection of inhibitors of Aftin induced action (potential 'anti-AD compounds', including GSIs and GSMs) but also (2) the identification, in the human chemical exposome, of compounds that, like Aftins, might trigger sustained Aβ42 production and Aβ38 down-regulation (potential 'pro-AD compounds').
Collapse
Affiliation(s)
- Arnaud Hochard
- ManRos Therapeutics, Centre de Perharidy, Roscoff, Bretagne, France
- CNRS, USR3151, Station Biologique, Roscoff, Bretagne, France
| | - Nassima Oumata
- ManRos Therapeutics, Centre de Perharidy, Roscoff, Bretagne, France
| | - Karima Bettayeb
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Olfa Gloulou
- Laboratoire de Chimie Organique 2, CNRS, UMR 8601, Université Paris-Descartes, Paris, France
| | - Xavier Fant
- CNRS, USR3151, Station Biologique, Roscoff, Bretagne, France
| | - Emilie Durieu
- ManRos Therapeutics, Centre de Perharidy, Roscoff, Bretagne, France
- CNRS, USR3151, Station Biologique, Roscoff, Bretagne, France
| | - Nelly Buron
- Mitologics SAS, Hôpital Robert Debré, 48, Boulevard Sérurier, Paris, France
| | - Mathieu Porceddu
- Mitologics SAS, Hôpital Robert Debré, 48, Boulevard Sérurier, Paris, France
| | | | - Hervé Galons
- ManRos Therapeutics, Centre de Perharidy, Roscoff, Bretagne, France
- Laboratoire de Chimie Organique 2, CNRS, UMR 8601, Université Paris-Descartes, Paris, France
| | - Marc Flajolet
- Laboratory of Molecular and Cellular Neuroscience, The Rockefeller University, New York, NY, USA
| | - Laurent Meijer
- ManRos Therapeutics, Centre de Perharidy, Roscoff, Bretagne, France
| |
Collapse
|
50
|
Yngve U, Paulsen K, Macsari I, Sundström M, Santangelo E, Linde C, Bogar K, Lake F, Besidski Y, Malmborg J, Strömberg K, Appelkvist P, Radesäter AC, Olsson F, Bergström D, Klintenberg R, Arvidsson PI. Triazolopyrimidinones as γ-secretase modulators: structure–activity relationship, modulator profile, and in vivo profiling. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20312j] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|