1
|
Han X, Guo K. Pharmacophore-based virtual screening of commercial databases against β-secretase 1 for drug development against Alzheimer's disease. Front Chem 2024; 12:1412349. [PMID: 39045333 PMCID: PMC11263123 DOI: 10.3389/fchem.2024.1412349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 06/11/2024] [Indexed: 07/25/2024] Open
Abstract
β-secretase 1, one of the most important proteins, is an aspartate protease. This membrane-associated protein is used for treating Alzheimer's disease (AD). Several inhibitors have been pursued against β-secretase 1, but they still have not resulted effectively. Virtual screening based on pharmacophores has been shown to be useful for lead optimization and hit identification in the preliminary phase of developing a new drug. Here, we screen the commercially available databases to find the hits against β-secretase 1 for drug discovery against AD. Virtual screening for 200,000 compounds was done using the database from the Vitas-M Laboratory. The phase screen score was utilized to assess the screened hits. Molecular docking was performed on compounds with phase scores >1.9. According to the study, the 66H ligand of the crystal structure has the maximum performance against β-secretase 1. The redocking of the co-crystal ligand showed that the docked ligand was seamlessly united with the crystal structure. The reference complex had three hydrogen bonds with Asp93, Asp289, and Gly291; one van der Waals interaction with Gly74; and three hydrophobic interactions. After equilibration, the RMSD of the reference compound sustained a value of ∼1.5 Å until 30 ns and then boosted to 2.5 Å. On comparison, the RMSD of the S1 complex steadily increased to ∼2.5 Å at 15 ns, displayed slight aberrations at approximately ∼2.5-3 Å until 80 ns, and then achieved steadiness toward the end of the simulation. The arrangements of proteins stayed condensed during the mockup when bonded to these complexes as stable Rg values showed. Furthermore, the MM/GBSA technique was employed to analyze both compounds' total binding free energies (ΔGtotal). Our research study provides a new understanding of using 66H as anti-β-secretase 1 for drug development against AD.
Collapse
Affiliation(s)
- Xu Han
- The First Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Kaibo Guo
- Department of Oncology, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou, China
| |
Collapse
|
2
|
Zhao H, Ravn AK, Haibach MC, Engle KM, Johansson Seechurn CCC. Diversification of Pharmaceutical Manufacturing Processes: Taking the Plunge into the Non-PGM Catalyst Pool. ACS Catal 2024; 14:9708-9733. [PMID: 38988647 PMCID: PMC11232362 DOI: 10.1021/acscatal.4c01809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 07/12/2024]
Abstract
Recent global events have led to the cost of platinum group metals (PGMs) reaching unprecedented heights. Many chemical companies are therefore starting to seriously consider and evaluate if and where they can substitute PGMs for non-PGMs in their catalytic processes. This review covers recent highly relevant applications of non-PGM catalysts in the modern pharmaceutical industry. By highlighting these selected successful examples of non-PGM-catalyzed processes from the literature, we hope to emphasize the enormous potential of non-PGM catalysis and inspire further development within this field to enable this technology to progress toward manufacturing processes. We also present some historical contexts and review the perceived advantages and challenges of implementing non-PGM catalysts in the pharmaceutical manufacturing environment.
Collapse
Affiliation(s)
- Hui Zhao
- Sinocompound
Catalysts, Building C,
Bonded Area Technology Innovation Zone, Zhangjiagang, Jiangsu 215634, China
| | - Anne K. Ravn
- Department
of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | - Michael C. Haibach
- Process
Research and Development, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064, United States
| | - Keary M. Engle
- Department
of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, United States
| | | |
Collapse
|
3
|
Tanaka H, Yasui T, Uyanik M, Ishihara K. 1,3-Migrative Ring Expansion of Spiroindolenines to Azepino[3,4- b]indoles. Org Lett 2023; 25:2377-2381. [PMID: 36847223 DOI: 10.1021/acs.orglett.3c00207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2023]
Abstract
We serendipitously found an unprecedented 5-to-7-membered ring expansion of 2-alkylspiroindolenines to azepinoindoles mediated by n-tetrabutylammonium fluoride. The starting materials can be easily prepared by the hypoiodite-catalyzed oxidative dearomative spirocyclization of indole derivatives. Mildly basic conditions and electron-deficient protecting groups for the amines were found to be crucial to promoting chemoselective reactions. Moreover, the ring expansion of aniline-derived spiroindolenines proceeds smoothly under much milder conditions using only a catalytic amount of cesium carbonate.
Collapse
Affiliation(s)
- Hiroki Tanaka
- Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan
| | - Toshihiro Yasui
- Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan
| | - Muhammet Uyanik
- Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan
| | - Kazuaki Ishihara
- Graduate School of Engineering, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8603, Japan
| |
Collapse
|
4
|
Yang Q, Zhao Y, Ma D. Cu-Mediated Ullmann-Type Cross-Coupling and Industrial Applications in Route Design, Process Development, and Scale-up of Pharmaceutical and Agrochemical Processes. Org Process Res Dev 2022. [DOI: 10.1021/acs.oprd.2c00050] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Qiang Yang
- Synthetic Molecule Design and Development, Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | - Yinsong Zhao
- Department of Chemistry, Southern University of Science and Technology, Shenzhen, Guangdong 518055, China
| | - Dawei Ma
- State Key Laboratory of Bioorganic and Natural Products Chemistry, Center for Excellence in Molecular Synthesis, Shanghai Institute of Organic Chemistry, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200032, China
| |
Collapse
|
5
|
Ugbaja SC, Appiah-Kubi P, Lawal MM, Gumede NS, Kumalo HM. Unravelling the molecular basis of AM-6494 high potency at BACE1 in Alzheimer's disease: an integrated dynamic interaction investigation. J Biomol Struct Dyn 2021; 40:5253-5265. [PMID: 33410374 DOI: 10.1080/07391102.2020.1869099] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
β-amyloid precursor protein cleaving enzyme1 (BACE1) has prominently been an important drug design target implicated in Alzheimer's disease pathway. The failure rate of most of the already tested drugs at different clinical phases remains a major concern. Recently, AM-6494 was reported as a novel potent, highly selective, and orally effective inhibitor against BACE1. AM-6494 displayed no alteration of skin/fur colour in animal studies, an adverse effect common to previous BACE1 inhibitors. However, the atomistic molecular mechanism of BACE1 inhibition by AM-6494 remains unclear. To elucidate the binding mechanism of AM-6494 relative to umibecestat (CNP-520) as well as the structural changes when bound to BACE1, advanced computational techniques such as accelerated MD simulation and principal component analysis have been utilised. The results demonstrated higher binding affinity of AM-6494 at BACE1 with van der Waals as dominant energy contributor compared to umibecestat. Conformational monitoring of the β-hairpin flap covering the active site revealed an effective flap closure when bound with AM-6494 compared to CNP-520, which predominantly alternates between semi-open and closed conformations. The observed effective flap closure of AM-6494 explains its higher inhibitory power towards BACE1. Besides the catalytic Asp32/228 dyad, Tyr14, Leu30, Tyr71 and Gly230 represent critical residues in the potency of these inhibitors at BACE1 binding interface. The findings highlighted in this research provide a basis to explain AM-6494 high inhibitory potency and might assist in the design of new inhibitors with improved selectivity and potency for BACE1.
Collapse
Affiliation(s)
- Samuel C Ugbaja
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Patrick Appiah-Kubi
- Discipline of Pharmaceutical Sciences, School of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Monsurat M Lawal
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Nelisiwe S Gumede
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| | - Hezekiel M Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
6
|
Ugbaja SC, Sanusi ZK, Appiah-Kubi P, Lawal MM, Kumalo HM. Computational modelling of potent β-secretase (BACE1) inhibitors towards Alzheimer's disease treatment. Biophys Chem 2020; 270:106536. [PMID: 33387910 DOI: 10.1016/j.bpc.2020.106536] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 12/20/2020] [Accepted: 12/21/2020] [Indexed: 12/28/2022]
Abstract
Researchers have identified the β-amyloid precursor protein cleaving enzyme 1 (BACE1) in the multifactorial pathway of Alzheimer's disease (AD) as a drug target. The design and development of molecules to inhibit BACE1 as a potential cure for AD thus remained significant. Herein, we simulated two potent BACE1 inhibitors (AM-6494 and CNP-520) to understand their binding affinity at the atomistic level. AM-6494 is a newly reported potent BACE1 inhibitor with an IC50 value of 0.4 nM in vivo and now picked for preclinical considerations. Umibecestat (CNP-520), which was discontinued at human trials lately, was considered to enable a reasonable evaluation of our results. Using density functional theory (DFT) and Our Own N-layered Integrated molecular Orbital and Molecular Mechanics (ONIOM), we achieved the aim of this investigation. These computational approaches enabled the prediction of the electronic properties of AM-6494 and CNP-520 plus their binding energies when complexed with BACE1. For AM-6494 and CNP-520 interaction with protonated BACE1, the ONIOM calculation gave binding free energy of -62.849 and -33.463 kcal/mol, respectively. In the unprotonated model, we observed binding free energy of -59.758 kcal/mol in AM-6494. Taken together thermochemistry of the process and molecular interaction plot, AM-6494 is more favourable than CNP-520 towards the inhibition of BACE1. The protonated model gave slightly better binding energy than the unprotonated form. However, both models could sufficiently describe ligand binding to BACE1 at the atomistic level. Understanding the detailed molecular interaction of these inhibitors could serve as a basis for pharmacophore exploration towards improved inhibitor design.
Collapse
Affiliation(s)
- Samuel C Ugbaja
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Zainab K Sanusi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa
| | - Patrick Appiah-Kubi
- Molecular Bio-computational and Drug Design Research Group, School of Health Sciences, University of KwaZulu-Natal, Westville Campus, Durban 4001, South Africa
| | - Monsurat M Lawal
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa.
| | - Hezekiel M Kumalo
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Science, University of KwaZulu-Natal, Durban 4001, South Africa.
| |
Collapse
|
7
|
Berlinck RGS, Bernardi DI, Fill T, Fernandes AAG, Jurberg ID. The chemistry and biology of guanidine secondary metabolites. Nat Prod Rep 2020; 38:586-667. [PMID: 33021301 DOI: 10.1039/d0np00051e] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Covering: 2017-2019Guanidine natural products isolated from microorganisms, marine invertebrates and terrestrial plants, amphibians and spiders, represented by non-ribosomal peptides, guanidine-bearing polyketides, alkaloids, terpenoids and shikimic acid derived, are the subject of this review. The topics include the discovery of new metabolites, total synthesis of natural guanidine compounds, biological activity and mechanism-of-action, biosynthesis and ecological functions.
Collapse
Affiliation(s)
- Roberto G S Berlinck
- Instituto de Química de São Carlos, Universidade de São Paulo, CP 780, CEP 13560-970, São Carlos, SP, Brazil.
| | | | | | | | | |
Collapse
|
8
|
Rivera NR, Kassim B, Grigorov P, Wang H, Armenante M, Bu X, Lekhal A, Variankaval N. Investigation of a Flow Step Clogging Incident: A Precautionary Note on the Use of THF in Commercial-Scale Continuous Process. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
9
|
Burns MJ, Ott MA, Teasdale A, Stalford SA, Antonucci V, Baumann JC, Brown R, Covey-Crump EM, Elder D, Elliott E, Fennell JW, Gallou F, Ide ND, Itoh T, Jordine G, Kallemeyn JM, Lauwers D, Looker AR, Lovelle LE, Molzahn R, Schils D, Schulte Oestrich R, Sluggett GW, Stevenson N, Talavera P, Urquhart MW, Varie DL, Welch DS. New Semi-Automated Computer-Based System for Assessing the Purge of Mutagenic Impurities. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00358] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
| | | | | | | | - Vincent Antonucci
- Merck Sharp & Dohme Corporation, Rahway, New Jersey 07033, United States
| | | | | | | | - David Elder
- Consultant, Hertford SG14 2DE, Hertfordshire, U.K
| | - Eric Elliott
- Takeda Pharmaceuticals, Cambridge, Massachusetts 02139, United States
| | - Jared W. Fennell
- Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | | | - Nathan D. Ide
- AbbVie, North Chicago, Illinois 60064, United States
| | - Tetsuji Itoh
- Merck Sharp & Dohme Corporation, Rahway, New Jersey 07033, United States
| | | | | | | | - Adam R. Looker
- Vertex Pharmaceuticals, Boston, Massachusetts 02210, United States
| | | | | | | | | | | | | | | | | | - David L. Varie
- Eli Lilly and Company, Indianapolis, Indiana 46285, United States
| | | |
Collapse
|
10
|
Phillips EM, Reibarkh M, Limanto J, Kieu M, Lekhal A, Zewge D. Improved Process for a Copper-Catalyzed C–N Coupling in the Synthesis of Verubecestat. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00192] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Affiliation(s)
- Eric M. Phillips
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Mikhail Reibarkh
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - John Limanto
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Minh Kieu
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Azzeddine Lekhal
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Daniel Zewge
- Department of Process Research and Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
11
|
Snead DR, Lévesque F, Morris WJ, Naber JR. An improved Balz-Schiemann reaction enabled by ionic liquids and continuous processing. Tetrahedron 2019. [DOI: 10.1016/j.tet.2019.05.020] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
12
|
Beutner GL, Coombs JR, Green RA, Inankur B, Lin D, Qiu J, Roberts F, Simmons EM, Wisniewski SR. Palladium-Catalyzed Amidation and Amination of (Hetero)aryl Chlorides under Homogeneous Conditions Enabled by a Soluble DBU/NaTFA Dual-Base System. Org Process Res Dev 2019. [DOI: 10.1021/acs.oprd.9b00196] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Gregory L. Beutner
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - John R. Coombs
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Rebecca A. Green
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Bahar Inankur
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Dong Lin
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Jun Qiu
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Frederick Roberts
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Eric M. Simmons
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| | - Steven R. Wisniewski
- Chemical & Synthetic Development, Bristol-Myers Squibb Company, One Squibb Drive, New Brunswick, New Jersey 08903, United States
| |
Collapse
|
13
|
Fujimoto K, Matsuoka E, Asada N, Tadano G, Yamamoto T, Nakahara K, Fuchino K, Ito H, Kanegawa N, Moechars D, Gijsen HJM, Kusakabe KI. Structure-Based Design of Selective β-Site Amyloid Precursor Protein Cleaving Enzyme 1 (BACE1) Inhibitors: Targeting the Flap to Gain Selectivity over BACE2. J Med Chem 2019; 62:5080-5095. [DOI: 10.1021/acs.jmedchem.9b00309] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
14
|
Yang AJ, Hayward GC, MacPherson REK. Acute exercise and brain BACE1 protein content: a time course study. Physiol Rep 2019; 7:e14084. [PMID: 31033251 PMCID: PMC6487470 DOI: 10.14814/phy2.14084] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 12/23/2022] Open
Abstract
Obesity and insulin resistance are risk factors in the development of neurodegenerative disorders. Previous work suggests that one acute bout of exercise may have beneficial neuro-protective effects in obese mice. The rate limiting enzyme in the production of amyloid-beta peptides, BACE1, was reduced in the prefrontal cortex 2 h post-exercise, however if these effects remain over time is unknown. We aimed to determine how long exercise-induced alterations persist in the prefrontal cortex and hippocampus following a single exercise bout. Male C57BL/6J mice were fed either a low (LFD, 10% kcals from lard) or a high fat diet (HFD, 60% kcals from lard) for 7 weeks. HFD mice then underwent an acute bout of treadmill running (15 m/min, 5% incline, 120 min) followed by 2-, 8-, or 24-h of recovery. The HFD increased body mass (LFD 27.8 ± 1.05 vs. HFD 41.7 ± 0.60 g; P < 0.05) and glucose intolerance (AUC LFD 63.27 ± 4.5 vs. HFD 128.9 ± 4.6; P < 0.05). Prefrontal cortex BACE1 content was reduced 2- and 8-h post-exercise compared to sedentary HFD mice, however BACE1 protein content at 24 h was not different. Hippocampal BACE1 content was reduced 8- and 24-h post-exercise. Compared to the LFD, the HFD had higher prefrontal cortex phosphorylation of p38, JNK, and AMPK, indicative of increased neuronal stress. Post-exercise prefrontal cortex p38 and JNK phosphorylation were no different between the HFD or LFD groups, while ERK phosphorylation was significantly reduced by 24 h. The HFD increased JNK phosphorylation in the hippocampus. These results demonstrate the direct and potent effects of exercise on reducing BACE1 prefrontal cortex and hippocampal content. However the reduction in prefrontal cortex BACE1 content is short lived.
Collapse
Affiliation(s)
- Alex J. Yang
- Department of Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | - Grant C. Hayward
- Department of Health SciencesBrock UniversitySt. CatharinesOntarioCanada
| | | |
Collapse
|
15
|
Kuhl N, Raval S, Cohen RD. Synthesis of Cyanamides via a One-Pot Oxidation–Cyanation of Primary and Secondary Amines. Org Lett 2019; 21:1268-1272. [DOI: 10.1021/acs.orglett.8b04007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Nadine Kuhl
- Department of Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Saurin Raval
- Department of Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ryan D. Cohen
- Department of Process Research & Development, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
16
|
Dong Y, Li X, Cheng J, Hou L. Drug Development for Alzheimer's Disease: Microglia Induced Neuroinflammation as a Target? Int J Mol Sci 2019; 20:E558. [PMID: 30696107 PMCID: PMC6386861 DOI: 10.3390/ijms20030558] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/09/2019] [Accepted: 01/13/2019] [Indexed: 12/14/2022] Open
Abstract
Alzheimer's disease (AD) is one of the most common causes of dementia. Its pathogenesis is characterized by the aggregation of the amyloid-β (Aβ) protein in senile plaques and the hyperphosphorylated tau protein in neurofibrillary tangles in the brain. Current medications for AD can provide temporary help with the memory symptoms and other cognitive changes of patients, however, they are not able to stop or reverse the progression of AD. New medication discovery and the development of a cure for AD is urgently in need. In this review, we summarized drugs for AD treatments and their recent updates, and discussed the potential of microglia induced neuroinflammation as a target for anti-AD drug development.
Collapse
Affiliation(s)
- Yuan Dong
- Department of Biochemistry, Medical College, Qingdao University, Qingdao 266071, China.
| | - Xiaoheng Li
- Beijing Institute for Brain Disorders, Capital Medical University, Beijing 100069, China.
| | - Jinbo Cheng
- The Brain Science Center, Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | - Lin Hou
- Department of Biochemistry, Medical College, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
17
|
Chen W, Meng D, N'Zemba B, Morris WJ. Palladium-Catalyzed Enantioselective Synthesis of Cyclic Sulfamidates and Application to a Synthesis of Verubecestat. Org Lett 2018; 20:1265-1268. [PMID: 29461065 DOI: 10.1021/acs.orglett.7b03639] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
An enantioselective arylation reaction catalyzed by palladium complexed with substituted phosphinooxazoline (PHOX) ligands is described. Aza-quaternary stereocenters are readily accessible through the arylation reaction between cyclic iminosulfates and a wide variety of arylboronic acids, including electron-poor and ortho-substituted arylboronic acids. This reaction was applied to the preparation of verubecestat, which is currently undergoing clinical evaluation for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Wenyong Chen
- Process Research & Development, Merck & Co., Inc. , Rahway, New Jersey 07065, United States
| | - Dongfang Meng
- Process Research & Development, Merck & Co., Inc. , Rahway, New Jersey 07065, United States
| | - Blaise N'Zemba
- Process Research & Development, Merck & Co., Inc. , Rahway, New Jersey 07065, United States
| | - William J Morris
- Process Research & Development, Merck & Co., Inc. , Rahway, New Jersey 07065, United States
| |
Collapse
|