51
|
Brown DG. A Medicinal Chemistry Perspective on the Hit‐to‐Lead Phase in the Current Era of Drug Discovery. ACTA ACUST UNITED AC 2016. [DOI: 10.1002/9783527677047.ch12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
52
|
Inhibition of Large Neutral Amino Acid Transporters Suppresses Kynurenic Acid Production Via Inhibition of Kynurenine Uptake in Rodent Brain. Neurochem Res 2016; 41:2256-66. [DOI: 10.1007/s11064-016-1940-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 04/18/2016] [Accepted: 04/28/2016] [Indexed: 12/11/2022]
|
53
|
Lu H, Kopcho L, Ghosh K, Witmer M, Parker M, Gupta S, Paul M, Krishnamurthy P, Laksmaiah B, Xie D, Tredup J, Zhang L, Abell LM. Development of a RapidFire mass spectrometry assay and a fluorescence assay for the discovery of kynurenine aminotransferase II inhibitors to treat central nervous system disorders. Anal Biochem 2016; 501:56-65. [PMID: 26874021 DOI: 10.1016/j.ab.2016.02.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 01/15/2016] [Accepted: 02/02/2016] [Indexed: 12/16/2022]
Abstract
Kynurenine aminotransferases convert kynurenine to kynurenic acid and play an important role in the tryptophan degradation pathway. Kynurenic acid levels in brain have been hypothesized to be linked to a number of central nervous system (CNS) disorders. Kynurenine aminotransferase II (KATII) has proven to be a key modulator of kynurenic acid levels in brain and, thus, is an attractive target to treat CNS diseases. A sensitive, high-throughput, label-free RapidFire mass spectrometry assay has been developed for human KATII. Unlike other assays, this method is directly applicable to KATII enzymes from different animal species, which allows us to select proper animal model(s) to evaluate human KATII inhibitors. We also established a coupled fluorescence assay for human KATII. The short assay time and kinetic capability of the fluorescence assay provide a useful tool for orthogonal inhibitor validation and mechanistic studies.
Collapse
Affiliation(s)
- Hao Lu
- Lead Discovery and Optimization, Bristol-Myers Squibb R&D, Pennington, NJ 08534, USA.
| | - Lisa Kopcho
- Lead Discovery and Optimization, Bristol-Myers Squibb R&D, Pennington, NJ 08534, USA
| | - Kaushik Ghosh
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb R&D Centre, Bangalore, 560099, India
| | - Mark Witmer
- Protein Science, Bristol-Myers Squibb R&D, Princeton, NJ 08648, USA
| | - Michael Parker
- Discovery Chemistry, Bristol-Myers Squibb R&D, Wallingford, CT 06492, USA
| | - Sumit Gupta
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb R&D Centre, Bangalore, 560099, India
| | - Marilyn Paul
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb R&D Centre, Bangalore, 560099, India
| | - Prasad Krishnamurthy
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb R&D Centre, Bangalore, 560099, India
| | - Basanth Laksmaiah
- Disease Sciences and Technology, Biocon Bristol-Myers Squibb R&D Centre, Bangalore, 560099, India
| | - Dianlin Xie
- Protein Science, Bristol-Myers Squibb R&D, Princeton, NJ 08648, USA
| | - Jeffrey Tredup
- Protein Science, Bristol-Myers Squibb R&D, Princeton, NJ 08648, USA
| | - Litao Zhang
- Lead Discovery and Optimization, Bristol-Myers Squibb R&D, Pennington, NJ 08534, USA
| | - Lynn M Abell
- Lead Discovery and Optimization, Bristol-Myers Squibb R&D, Pennington, NJ 08534, USA.
| |
Collapse
|
54
|
Sun G, Nematollahi A, Nadvi NA, Kwan AH, Jeffries CM, Church WB. Expression, purification and crystallization of human kynurenine aminotransferase 2 exploiting a highly optimized codon set. Protein Expr Purif 2016; 121:41-5. [PMID: 26773745 DOI: 10.1016/j.pep.2016.01.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 01/05/2016] [Accepted: 01/06/2016] [Indexed: 10/22/2022]
Abstract
Kynurenine aminotransferase (KAT) is a pyridoxal-5'-phosphate (PLP) dependent enzyme that catalyses kynurenine (KYN) to kynurenic acid (KYNA), a neuroactive product in the tryptophan metabolic pathway. Evidence suggests that abnormal levels of KYNA are involved in many neurodegenerative diseases such as Parkinson's disease, Huntington's disease, Alzheimer's disease and schizophrenia. Reducing KYNA production through inhibiting kynurenine aminotransferase 2 (KAT2) would be a promising approach to understanding and treating the related neurological and mental disorders. In this study we used an optimized codon sequence to overexpress histidine-tagged human KAT2 (hKAT2) using an Escherichia coli expression system. After a single step of Ni-NTA based purification the purified protein (>95%) was confirmed to be active by an HPLC based activity assay and was crystallized using the hanging-drop vapour diffusion method. The crystal system represents a novel space group, and a complete X-ray diffraction data set was collected to 1.83 Å resolution, and higher resolution data than for any reported native human KAT2 structure. The optimised method of protein production provides a fast and reliable technique to generate large quantities of active human KAT2 suitable for future small-molecule lead compound screening and structural design work.
Collapse
Affiliation(s)
- Guanchen Sun
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, NSW 2006, Australia
| | - Alireza Nematollahi
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, NSW 2006, Australia
| | - Naveed A Nadvi
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia
| | - Ann H Kwan
- School of Molecular Bioscience, University of Sydney, NSW 2006, Australia
| | - Cy M Jeffries
- Bragg Institute, Australian Nuclear Science and Technology Organisation, Lucas Heights, NSW 2234, Australia
| | - W Bret Church
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy, University of Sydney, NSW 2006, Australia.
| |
Collapse
|
55
|
Nasir N, Anant A, Vyas R, Biswal BK. Crystal structures of Mycobacterium tuberculosis HspAT and ArAT reveal structural basis of their distinct substrate specificities. Sci Rep 2016; 6:18880. [PMID: 26738801 PMCID: PMC4703992 DOI: 10.1038/srep18880] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 11/30/2015] [Indexed: 01/07/2023] Open
Abstract
Aminotransferases of subfamily Iβ, which include histidinol phosphate aminotransferases (HspATs) and aromatic amino acid aminotransferases (ArATs), are structurally similar but possess distinct substrate specificities. This study, encompassing structural and biochemical characterisation of HspAT and ArAT from Mycobacterium tuberculosis demonstrates that the residues lining the substrate binding pocket and N-terminal lid are the primary determinants of their substrate specificities. In mHspAT, hydrophilic residues in the substrate binding pocket and N-terminal lid allow the entry and binding of its preferential substrate, Hsp. On the other hand, the hydrophobic nature of both the substrate binding pocket and the N-terminal lid of mArAT is responsible for the discrimination of a polar substrate such as Hsp, while facilitating the binding of Phe and other aromatic residues such as Tyr and Trp. In addition, the present study delineates the ligand induced conformational rearrangements, providing insights into the plasticity of aminotransferases. Furthermore, the study also demonstrates that the adventitiously bound ligand 2-(N-morpholino)ethanesulfonic acid (MES) is indeed a specific inhibitor of HspAT. These results suggest that previously untapped morpholine-ring scaffold compounds could be explored for the design of new anti-TB agents.
Collapse
Affiliation(s)
- Nazia Nasir
- Protein Crystallography Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India
| | - Avishek Anant
- Protein Crystallography Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India
| | - Rajan Vyas
- Protein Crystallography Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India
| | - Bichitra Kumar Biswal
- Protein Crystallography Laboratory, National Institute of Immunology, Aruna Asaf Ali Marg, New Delhi, Delhi, 110067, India,
| |
Collapse
|
56
|
Astrocytes as Pharmacological Targets in the Treatment of Schizophrenia. HANDBOOK OF BEHAVIORAL NEUROSCIENCE 2016. [DOI: 10.1016/b978-0-12-800981-9.00025-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
57
|
O'Farrell K, Harkin A. Stress-related regulation of the kynurenine pathway: Relevance to neuropsychiatric and degenerative disorders. Neuropharmacology 2015; 112:307-323. [PMID: 26690895 DOI: 10.1016/j.neuropharm.2015.12.004] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 12/02/2015] [Accepted: 12/08/2015] [Indexed: 02/08/2023]
Abstract
The kynurenine pathway (KP), which is activated in times of stress and infection has been implicated in the pathophysiology of neurodegenerative and psychiatric disorders. Activation of this tryptophan metabolising pathway results in the production of neuroactive metabolites which have the potential to interfere with normal neuronal functioning which may contribute to altered neuronal transmission and the emergence of symptoms of these brain disorders. This review investigates the involvement of the KP in a range of neurological disorders, examining recent in vitro, in vivo and clinical discoveries highlights evidence to indicate that the KP is a potential therapeutic target in both neurodegenerative and stress-related neuropsychiatric disorders. Furthermore, this review identifies gaps in our knowledge with regard to this field which are yet to be examined to lead to a more comprehensive understanding of the role of KP activation in brain health and disease. This article is part of the Special Issue entitled 'The Kynurenine Pathway in Health and Disease'.
Collapse
Affiliation(s)
- Katherine O'Farrell
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland
| | - Andrew Harkin
- Neuropsychopharmacology Research Group, School of Pharmacy and Pharmaceutical Sciences & Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland; Neuroimmunology Research Group, Department of Physiology, School of Medicine & Trinity College Institute of Neuroscience, Trinity College Dublin, Ireland.
| |
Collapse
|
58
|
Dounay AB, Tuttle JB, Verhoest PR. Challenges and Opportunities in the Discovery of New Therapeutics Targeting the Kynurenine Pathway. J Med Chem 2015. [DOI: 10.1021/acs.jmedchem.5b00461] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Amy B. Dounay
- Department
of Chemistry and Biochemistry, Colorado College, 14 E. Cache
La Poudre Street, Colorado Springs, Colorado 80903, United States
| | - Jamison B. Tuttle
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research & Development, Cambridge, Massachusetts 02139, United States
| | - Patrick R. Verhoest
- Worldwide Medicinal Chemistry, Pfizer Worldwide Research & Development, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
59
|
Dhole S, Selvaraju M, Maiti B, Chanda K, Sun CM. Microwave Controlled Reductive Cyclization: A Selective Synthesis of Novel Benzimidazole-alkyloxypyrrolo[1,2-a]quinoxalinones. ACS COMBINATORIAL SCIENCE 2015; 17:310-6. [PMID: 25897944 DOI: 10.1021/acscombsci.5b00010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
An efficient cascade synthesis of novel benzimidazole linked alkyloxypyrrolo[1,2-a]quinoxalinones was explored on soluble polymer support under microwave irradiation. Two exclusive protocols have been developed for the partial and full reductive cyclization by controlling the microwave energy. Commencing from the same substrate, ortho nitro pyrrol carboxylates, N-hydroxy pyrroloquinoxalinones were obtained by partial reductive cyclization (60 °C, 7 min), and the synthesis of pyrroloquinoxalinones was accomplished by full reductive cyclization (85 °C, 12 min). This method represents the first synthesis of N-hydroxy pyrroloquinoxalinones using Pd/C and ammonium formate as reducing agents. Further employing a variety of alkyl bromides, the obtained pyrroloquinoxalinones were transformed to their corresponding O- and N-alkylated analogues to deliver the diversified, novel molecular entities.
Collapse
Affiliation(s)
- Sandip Dhole
- Department
of Applied Chemistry, National Chiao Tung University, 1001 Ta-Hseuh
Road, Hsinchu 300-10, Taiwan
| | - Manikandan Selvaraju
- Department
of Applied Chemistry, National Chiao Tung University, 1001 Ta-Hseuh
Road, Hsinchu 300-10, Taiwan
| | - Barnali Maiti
- Department
of Applied Chemistry, National Chiao Tung University, 1001 Ta-Hseuh
Road, Hsinchu 300-10, Taiwan
| | - Kaushik Chanda
- Department
of Applied Chemistry, National Chiao Tung University, 1001 Ta-Hseuh
Road, Hsinchu 300-10, Taiwan
| | - Chung-Ming Sun
- Department
of Applied Chemistry, National Chiao Tung University, 1001 Ta-Hseuh
Road, Hsinchu 300-10, Taiwan
- Department
of Medicinal and Applied Chemistry, Kaohsiung Medical University, 100,
Shih-Chuan first Road, Kaohsiung 807-08, Taiwan
| |
Collapse
|
60
|
Chen G, Shigenari T, Jain P, Zhang Z, Jin Z, He J, Li S, Mapelli C, Miller MM, Poss MA, Scola PM, Yeung KS, Yu JQ. Ligand-enabled β-C-H arylation of α-amino acids using a simple and practical auxiliary. J Am Chem Soc 2015; 137:3338-51. [PMID: 25697780 PMCID: PMC4432912 DOI: 10.1021/ja512690x] [Citation(s) in RCA: 136] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pd-catalyzed β-C-H functionalizations of carboxylic acid derivatives using an auxiliary as a directing group have been extensively explored in the past decade. In comparison to the most widely used auxiliaries in asymmetric synthesis, the simplicity and practicality of the auxiliaries developed for C-H activation remains to be improved. We previously developed a simple N-methoxyamide auxiliary to direct β-C-H activation, albeit this system was not compatible with carboxylic acids containing α-hydrogen atoms. Herein we report the development of a pyridine-type ligand that overcomes this limitation of the N-methoxyamide auxiliary, leading to a significant improvement of β-arylation of carboxylic acid derivatives, especially α-amino acids. The arylation using this practical auxiliary is applied to the gram-scale syntheses of unnatural amino acids, bioactive molecules, and chiral bis(oxazoline) ligands.
Collapse
Affiliation(s)
- Gang Chen
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Toshihiko Shigenari
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Pankaj Jain
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Zhipeng Zhang
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Zhong Jin
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Jian He
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Suhua Li
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| | - Claudio Mapelli
- Bristol-Myers Squibb Co, Route 206, Province Line Road, Princeton, New Jersey 08543, United States
| | - Michael M. Miller
- Bristol-Myers Squibb Co, Route 206, Province Line Road, Princeton, New Jersey 08543, United States
| | - Michael A. Poss
- Bristol-Myers Squibb Co, Route 206, Province Line Road, Princeton, New Jersey 08543, United States
| | - Paul M. Scola
- Bristol-Myers Squibb Co, Route 206, Province Line Road, Princeton, New Jersey 08543, United States
| | - Kap-Sun Yeung
- Bristol-Myers Squibb Co, Route 206, Province Line Road, Princeton, New Jersey 08543, United States
| | - Jin-Quan Yu
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Road, La Jolla, California 92037, United States
| |
Collapse
|
61
|
Sekine A, Okamoto M, Kanatani Y, Sano M, Shibata K, Fukuwatari T. Amino acids inhibit kynurenic acid formation via suppression of kynurenine uptake or kynurenic acid synthesis in rat brain in vitro. SPRINGERPLUS 2015; 4:48. [PMID: 25674503 PMCID: PMC4318830 DOI: 10.1186/s40064-015-0826-9] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 01/16/2015] [Indexed: 12/12/2022]
Abstract
The tryptophan metabolite, kynurenic acid (KYNA), is a preferential antagonist of the α7 nicotinic acetylcholine receptor at endogenous brain concentrations. Recent studies have suggested that increase of brain KYNA levels is involved in psychiatric disorders such as schizophrenia and depression. KYNA-producing enzymes have broad substrate specificity for amino acids, and brain uptake of kynurenine (KYN), the immediate precursor of KYNA, is via large neutral amino acid transporters (LAT). In the present study, to find out amino acids with the potential to suppress KYNA production, we comprehensively investigated the effects of proteinogenic amino acids on KYNA formation and KYN uptake in rat brain in vitro. Cortical slices of rat brain were incubated for 2 h in Krebs-Ringer buffer containing a physiological concentration of KYN with individual amino acids. Ten out of 19 amino acids (specifically, leucine, isoleucine, phenylalanine, methionine, tyrosine, alanine, cysteine, glutamine, glutamate, and aspartate) significantly reduced KYNA formation at 1 mmol/L. These amino acids showed inhibitory effects in a dose-dependent manner, and partially inhibited KYNA production at physiological concentrations. Leucine, isoleucine, methionine, phenylalanine, and tyrosine, all LAT substrates, also reduced tissue KYN concentrations in a dose-dependent manner, with their inhibitory rates for KYN uptake significantly correlated with KYNA formation. These results suggest that five LAT substrates inhibit KYNA formation via blockade of KYN transport, while the other amino acids act via blockade of the KYNA synthesis reaction in brain. Amino acids can be a good tool to modulate brain function by manipulation of KYNA formation in the brain. This approach may be useful in the treatment and prevention of neurological and psychiatric diseases associated with increased KYNA levels.
Collapse
Affiliation(s)
- Airi Sekine
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga 522-8533 Japan
| | - Misaki Okamoto
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga 522-8533 Japan
| | - Yuka Kanatani
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga 522-8533 Japan
| | - Mitsue Sano
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga 522-8533 Japan
| | - Katsumi Shibata
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga 522-8533 Japan
| | - Tsutomu Fukuwatari
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, 2500 Hassaka, Hikone, Shiga 522-8533 Japan
| |
Collapse
|
62
|
Rankovic Z. CNS Drug Design: Balancing Physicochemical Properties for Optimal Brain Exposure. J Med Chem 2015; 58:2584-608. [DOI: 10.1021/jm501535r] [Citation(s) in RCA: 342] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Zoran Rankovic
- Eli Lilly and Company, 893 South
Delaware Street, Indianapolis, Indiana 46285, United States
| |
Collapse
|
63
|
Rani R, Granchi C. Bioactive heterocycles containing endocyclic N-hydroxy groups. Eur J Med Chem 2014; 97:505-24. [PMID: 25466924 DOI: 10.1016/j.ejmech.2014.11.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/13/2014] [Accepted: 11/15/2014] [Indexed: 10/24/2022]
Abstract
Drug-likeness rules consider N-O single bonds as "structural alerts" which should not be present in a perspective drug candidate. In most cases this concern is correct, since it is known that N-hydroxy metabolites of branded drugs produce reactive species that cause serious side effects. However, this dangerous reactivity of the N-OH species generally takes place when the nitrogen atom is not comprised in a cyclic moiety. In fact, the same type of metabolic behavior should not be expected when the nitrogen atom is included in the ring of an aromatic heterocyclic scaffold. Nevertheless, heterocycles bearing endocyclic N-hydroxy portions have so far been poorly studied as chemical classes that may provide new therapeutic agents. This review provides an overview of N-OH-containing heterocycles with reported bioactivities that may be considered as therapeutically relevant and, therefore, may extend the chemical space available for the future development of novel pharmaceuticals. A systematic treatment of the various chemical classes belonging to this particular family of molecules is described along with a discussion of the biological activities associated to the most important examples.
Collapse
Affiliation(s)
- Reshma Rani
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Carlotta Granchi
- Dipartimento di Farmacia, Università di Pisa, Via Bonanno 6, 56126 Pisa, Italy.
| |
Collapse
|
64
|
Pauli GF, Chen SN, Simmler C, Lankin DC, Gödecke T, Jaki BU, Friesen JB, McAlpine JB, Napolitano JG. Importance of purity evaluation and the potential of quantitative ¹H NMR as a purity assay. J Med Chem 2014; 57:9220-31. [PMID: 25295852 PMCID: PMC4255677 DOI: 10.1021/jm500734a] [Citation(s) in RCA: 266] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
![]()
In any biomedical and chemical context,
a truthful description of chemical constitution requires coverage
of both structure and purity. This qualification affects all drug
molecules, regardless of development stage (early discovery to approved
drug) and source (natural product or synthetic). Purity assessment
is particularly critical in discovery programs and whenever
chemistry is linked with biological and/or therapeutic outcome. Compared
with chromatography and elemental analysis, quantitative NMR (qNMR)
uses nearly universal detection and provides a versatile and orthogonal
means of purity evaluation. Absolute qNMR with flexible calibration
captures analytes that frequently escape detection (water, sorbents).
Widely accepted structural NMR workflows require minimal or no adjustments
to become practical 1H qNMR (qHNMR) procedures with simultaneous
qualitative and (absolute) quantitative capability. This study reviews
underlying concepts, provides a framework for standard qHNMR purity
assays, and shows how adequate accuracy and precision are achieved
for the intended use of the material.
Collapse
Affiliation(s)
- Guido F Pauli
- Department of Medicinal Chemistry and Pharmacognosy and ‡Institute for Tuberculosis Research, College of Pharmacy, University of Illinois at Chicago , 833 S. Wood Street, Chicago, Illinois 60612, United States
| | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Stone TW, Darlington LG. The kynurenine pathway as a therapeutic target in cognitive and neurodegenerative disorders. Br J Pharmacol 2014; 169:1211-27. [PMID: 23647169 DOI: 10.1111/bph.12230] [Citation(s) in RCA: 195] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 04/16/2013] [Accepted: 04/24/2013] [Indexed: 02/06/2023] Open
Abstract
Understanding the neurochemical basis for cognitive function is one of the major goals of neuroscience, with a potential impact on the diagnosis, prevention and treatment of a range of psychiatric and neurological disorders. In this review, the focus will be on a biochemical pathway that remains under-recognized in its implications for brain function, even though it can be responsible for moderating the activity of two neurotransmitters fundamentally involved in cognition - glutamate and acetylcholine. Since this pathway - the kynurenine pathway of tryptophan metabolism - is induced by immunological activation and stress, it also stands in a unique position to mediate the effects of environmental factors on cognition and behaviour. Targeting the pathway for new drug development could, therefore, be of value not only for the treatment of existing psychiatric conditions, but also for preventing the development of cognitive disorders in response to environmental pressures.
Collapse
Affiliation(s)
- Trevor W Stone
- Institute of Neuroscience & Psychology, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, UK.
| | | |
Collapse
|
66
|
Kozak R, Campbell BM, Strick CA, Horner W, Hoffmann WE, Kiss T, Chapin DS, McGinnis D, Abbott AL, Roberts BM, Fonseca K, Guanowsky V, Young DA, Seymour PA, Dounay A, Hajos M, Williams GV, Castner SA. Reduction of brain kynurenic acid improves cognitive function. J Neurosci 2014; 34:10592-602. [PMID: 25100593 PMCID: PMC6802596 DOI: 10.1523/jneurosci.1107-14.2014] [Citation(s) in RCA: 129] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 06/18/2014] [Accepted: 06/24/2014] [Indexed: 11/21/2022] Open
Abstract
The elevation of kynurenic acid (KYNA) observed in schizophrenic patients may contribute to core symptoms arising from glutamate hypofunction, including cognitive impairments. Although increased KYNA levels reduce excitatory neurotransmission, KYNA has been proposed to act as an endogenous antagonist at the glycine site of the glutamate NMDA receptor (NMDAR) and as a negative allosteric modulator at the α7 nicotinic acetylcholine receptor. Levels of KYNA are elevated in CSF and the postmortem brain of schizophrenia patients, and these elevated levels of KYNA could contribute to NMDAR hypofunction and the cognitive deficits and negative symptoms associated with this disease. However, the impact of endogenously produced KYNA on brain function and behavior is less well understood due to a paucity of pharmacological tools. To address this issue, we identified PF-04859989, a brain-penetrable inhibitor of kynurenine aminotransferase II (KAT II), the enzyme responsible for most brain KYNA synthesis. In rats, systemic administration of PF-04859989 dose-dependently reduced brain KYNA to as little as 28% of basal levels, and prevented amphetamine- and ketamine-induced disruption of auditory gating and improved performance in a sustained attention task. It also prevented ketamine-induced disruption of performance in a working memory task and a spatial memory task in rodents and nonhuman primates, respectively. Together, these findings support the hypotheses that endogenous KYNA impacts cognitive function and that inhibition of KAT II, and consequent lowering of endogenous brain KYNA levels, improves cognitive performance under conditions considered relevant for schizophrenia.
Collapse
Affiliation(s)
- Rouba Kozak
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139,
| | | | - Christine A Strick
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Weldon Horner
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - William E Hoffmann
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Tamas Kiss
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Douglas S Chapin
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Dina McGinnis
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Amanda L Abbott
- Departments of Psychiatry and VA Connecticut Healthcare System, West Haven, Connecticut 06519
| | - Brooke M Roberts
- Departments of Psychiatry and VA Connecticut Healthcare System, West Haven, Connecticut 06519
| | - Kari Fonseca
- Department of Pharmacokinetics, Pharmacodynamics and Metabolism, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Victor Guanowsky
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Damon A Young
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Patricia A Seymour
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Amy Dounay
- Neuroscience Research Unit, Worldwide Research and Development, Pfizer Inc., Cambridge, Massachusetts 02139
| | - Mihaly Hajos
- Comparative Medicine, Yale School of Medicine, New Haven, Connecticut 06510
| | - Graham V Williams
- Departments of Psychiatry and VA Connecticut Healthcare System, West Haven, Connecticut 06519
| | - Stacy A Castner
- Departments of Psychiatry and VA Connecticut Healthcare System, West Haven, Connecticut 06519
| |
Collapse
|
67
|
Lee M, Sousa MC. Structural basis for substrate specificity in ArnB. A key enzyme in the polymyxin resistance pathway of Gram-negative bacteria. Biochemistry 2014; 53:796-805. [PMID: 24460375 PMCID: PMC3985747 DOI: 10.1021/bi4015677] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
![]()
Cationic Antimicrobial Peptides (CAMPs)
represent a first line
of defense against bacterial colonization. When fighting Gram-negative
bacteria, CAMPs initially interact electrostatically with the negatively
charged phosphate groups in lipid A and are thought to kill bacteria
by disrupting their membrane integrity. However, many human pathogens,
including Salmonella and Pseudomonas, have evolved lipid A modification mechanisms
that result in resistance to CAMPs and related antibiotics such as
Colistin. The addition of 4-amino-4-deoxy-l-Arabinose (Ara4N)
to a phosphate group in lipid A is one such modification, frequently
found in Pseudomonas isolated from
cystic fibrosis patients. The pathway for biosynthesis of Ara4N-lipid
A requires conversion of UDP-Glucuronic acid into UDP-Ara4N and subsequent
transfer of the amino-sugar to lipid A. ArnB is a pyridoxal-phosphate
(PLP) dependent transaminase that catalyzes a crucial step in the
pathway: synthesis of UDP-Ara4N from UDP-4-keto-pentose. Here we present
the 2.3 Å resolution crystal structure of an active site mutant
of ArnB (K188A) in complex with the reaction intermediate aldimine
formed by UDP-Ara4N and PLP. The sugar–nucleotide binding site
is in a cleft between the subunits of the ArnB dimer with the uracil
buried at the interface and the UDP ribose and phosphate groups exposed
to the solvent. The Ara4N moiety is found in the 4C1 conformation and its positioning, stabilized by interactions
with both the protein and cofactor, is compatible with catalysis.
The structure suggests strategies for the development of specific
inhibitors that may prove useful in the treatment of resistant bacteria
such as Pseudomonas found in cystic
fibrosis patients.
Collapse
Affiliation(s)
- Myeongseon Lee
- Department of Chemistry and Biochemistry, University of Colorado at Boulder , Boulder, Colorado 80309-0596, United States
| | | |
Collapse
|
68
|
Li X, Yang L, Zhang X, Zhang-Negrerie D, Du Y, Zhao K. Construction of 1,4-Benzodiazepine Skeleton from 2-(Arylamino)benzamides through PhI(OAc)2-Mediated Oxidative C–N Bond Formation. J Org Chem 2014; 79:955-62. [DOI: 10.1021/jo402413g] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Xuming Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Liu Yang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Xiang Zhang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Daisy Zhang-Negrerie
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| | - Yunfei Du
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
- Collaborative
Innovation Center of Chemical Science and Engineering (Tianjin), Tianjin 300072, China
| | - Kang Zhao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin 300072, China
| |
Collapse
|
69
|
Kim YS, Keyser SGL, Schneekloth JS. Synthesis of 2',3',4'-trihydroxyflavone (2-D08), an inhibitor of protein sumoylation. Bioorg Med Chem Lett 2014; 24:1094-7. [PMID: 24468414 DOI: 10.1016/j.bmcl.2014.01.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Revised: 01/02/2014] [Accepted: 01/06/2014] [Indexed: 01/22/2023]
Abstract
Protein sumoylation is a dynamic posttranslational modification involved in diverse biological processes during cellular homeostasis and development. Recently sumoylation has been shown to play a critical role in cancer, although to date there are few small molecule probes available to inhibit enzymes involved in the SUMO conjugation process. As part of a program to identify and study inhibitors of sumoylation we recently reported the discovery that 2',3',4'-trihydroxyflavone (2-D08) is a cell permeable, mechanistically unique inhibitor of protein sumoylation. The work reported herein describes an efficient synthesis of 2-D08 as well as a structurally related but inactive isomer. We also report an unanticipated Wessely-Moser rearrangement that occurs under vigorous methyl ether deprotection conditions. This rearrangement likely gave rise to 2-D08 during a deprotection step, resulting in 2-D08 appearing as a contaminant in a screening well from a commercial supplier.
Collapse
Affiliation(s)
- Yeong Sang Kim
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, 376 Boyles St., Frederick, MD 21702, USA
| | - Samantha G L Keyser
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, 376 Boyles St., Frederick, MD 21702, USA
| | - John S Schneekloth
- Chemical Biology Laboratory, Center for Cancer Research, National Cancer Institute, 376 Boyles St., Frederick, MD 21702, USA.
| |
Collapse
|
70
|
Mah R, Thomas JR, Shafer CM. Drug discovery considerations in the development of covalent inhibitors. Bioorg Med Chem Lett 2013; 24:33-9. [PMID: 24314671 DOI: 10.1016/j.bmcl.2013.10.003] [Citation(s) in RCA: 153] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 09/27/2013] [Accepted: 10/01/2013] [Indexed: 12/31/2022]
Abstract
In recent years, the number of drug candidates with a covalent mechanism of action progressing through clinical trials or being approved by the FDA has increased significantly. And as interest in covalent inhibitors has increased, the technical challenges for characterizing and optimizing these inhibitors have become evident. A number of new tools have been developed to aid this process, but these have not gained wide-spread use. This review will highlight a number of methods and tools useful for prosecuting covalent inhibitor drug discovery programs.
Collapse
Affiliation(s)
- Robert Mah
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, Klybeckstrasse 141, CH-4057 Basel, Switzerland
| | - Jason R Thomas
- Developmental and Molecular Pathways, Novartis Institutes for Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Cynthia M Shafer
- Global Discovery Chemistry/Oncology & Exploratory Chemistry, Novartis Institutes for Biomedical Research, 4560 Horton Street, Emeryville, CA 94608, USA.
| |
Collapse
|
71
|
Cioffi CL. Modulation of NMDA receptor function as a treatment for schizophrenia. Bioorg Med Chem Lett 2013; 23:5034-44. [PMID: 23916256 DOI: 10.1016/j.bmcl.2013.07.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2013] [Revised: 07/03/2013] [Accepted: 07/13/2013] [Indexed: 11/30/2022]
Abstract
Schizophrenia is a devastating mental illness that afflicts nearly 1% of the world's population. Currently available antipsychotics treat positive symptoms, but are largely ineffective at addressing negative symptoms and cognitive dysfunction. Thus, improved pharmacotherapies that treat all aspects of the disease remain a critical unmet need. There is mounting evidence that links NMDA receptor hypofunction and the expression of schizophrenia, and numerous drug discovery programs have developed agents that directly or indirectly potentiate NMDA receptor-mediated neurotransmission. Several compounds have emerged that show promise for treating all symptom sub-domains in both preclinical models and clinical studies, and we will review recent developments in many of these areas.
Collapse
|
72
|
PF-04859989 as a template for structure-based drug design: identification of new pyrazole series of irreversible KAT II inhibitors with improved lipophilic efficiency. Bioorg Med Chem Lett 2013; 23:1961-6. [PMID: 23466229 DOI: 10.1016/j.bmcl.2013.02.039] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 02/07/2013] [Indexed: 11/24/2022]
Abstract
The structure-based design, synthesis, and biological evaluation of a new pyrazole series of irreversible KAT II inhibitors are described herein. The modification of the inhibitor scaffold of 1 and 2 from a dihydroquinolinone core to a tetrahydropyrazolopyridinone core led to discovery of a new series of potent KAT II inhibitors with excellent physicochemical properties. Compound 20 is the most potent and lipophilically efficient of these new pyrazole analogs, with a k(inact)/K(i) value of 112,000 M(-1)s(-1) and lipophilic efficiency (LipE) of 8.53. The X-ray crystal structure of 20 with KAT II demonstrates key features that contribute to this remarkable potency and binding efficiency.
Collapse
|
73
|
Tuttle JB, Anderson M, Bechle BM, Campbell BM, Chang C, Dounay AB, Evrard E, Fonseca KR, Gan X, Ghosh S, Horner W, James LC, Kim JY, McAllister LA, Pandit J, Parikh VD, Rago BJ, Salafia MA, Strick CA, Zawadzke LE, Verhoest PR. Structure-Based Design of Irreversible Human KAT II Inhibitors: Discovery of New Potency-Enhancing Interactions. ACS Med Chem Lett 2013; 4:37-40. [PMID: 24900560 DOI: 10.1021/ml300237v] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Accepted: 10/24/2012] [Indexed: 11/29/2022] Open
Abstract
A series of aryl hydroxamates recently have been disclosed as irreversible inhibitors of kynurenine amino transferase II (KAT II), an enzyme that may play a role in schizophrenia and other psychiatric and neurological disorders. The utilization of structure-activity relationships (SAR) in conjunction with X-ray crystallography led to the discovery of hydroxamate 4, a disubstituted analogue that has a significant potency enhancement due to a novel interaction with KAT II. The use of k inact/K i to assess potency was critical for understanding the SAR in this series and for identifying compounds with improved pharmacodynamic profiles.
Collapse
Affiliation(s)
- Jamison B. Tuttle
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Marie Anderson
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Bruce M. Bechle
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Brian M. Campbell
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Cheng Chang
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Amy B. Dounay
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Edelweiss Evrard
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Kari R. Fonseca
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Xinmin Gan
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Somraj Ghosh
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Weldon Horner
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Larry C. James
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Ji-Young Kim
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Laura A. McAllister
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Jayvardhan Pandit
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Vinod D. Parikh
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Brian J. Rago
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Michelle A. Salafia
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Christine A. Strick
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Laura E. Zawadzke
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Patrick R. Verhoest
- Pfizer Worldwide Research and Development, Neuroscience Medicinal Chemistry, Eastern
Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
74
|
Henderson JL, Sawant-Basak A, Tuttle JB, Dounay AB, McAllister LA, Pandit J, Rong S, Hou X, Bechle BM, Kim JY, Parikh V, Ghosh S, Evrard E, Zawadzke LE, Salafia MA, Rago B, Obach RS, Clark A, Fonseca KR, Chang C, Verhoest PR. Discovery of hydroxamate bioisosteres as KAT II inhibitors with improved oral bioavailability and pharmacokinetics. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20166f] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A series of kynurenine aminotransferase II (KAT II) inhibitors has been developed replacing the hydroxamate motif with a bioisostere.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Suobao Rong
- Neuroscience Medicinal Chemistry
- Cambridge
- USA
| | - Xinjun Hou
- Neuroscience Medicinal Chemistry
- Cambridge
- USA
| | | | | | | | | | | | | | | | - Brian Rago
- Neuroscience Medicinal Chemistry
- Cambridge
- USA
| | | | - Alan Clark
- Neuroscience Medicinal Chemistry
- Cambridge
- USA
| | | | - Cheng Chang
- Neuroscience Medicinal Chemistry
- Cambridge
- USA
| | | |
Collapse
|
75
|
Quiclet-Sire B, Tran NDM, Zard SZ. An Unusual Synthesis of N-Unsubstituted Benzazepinones. Org Lett 2012; 14:5514-7. [DOI: 10.1021/ol3026044] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Béatrice Quiclet-Sire
- Laboratoire de Synthèse Organique, CNRS UMR 7652 Ecole Polytechnique, 91128 Palaiseau Cedex, France
| | - Ngoc Diem My Tran
- Laboratoire de Synthèse Organique, CNRS UMR 7652 Ecole Polytechnique, 91128 Palaiseau Cedex, France
| | - Samir Z. Zard
- Laboratoire de Synthèse Organique, CNRS UMR 7652 Ecole Polytechnique, 91128 Palaiseau Cedex, France
| |
Collapse
|
76
|
Hopper AT, Campbell BM, Kao H, Pintchovski SA, Staal RG. Recent Developments in Targeting Neuroinflammation in Disease. ANNUAL REPORTS IN MEDICINAL CHEMISTRY VOLUME 47 2012. [DOI: 10.1016/b978-0-12-396492-2.00004-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|