1
|
Shaihutdinova ZM, Pashirova TN, Masson P. Slow-binding inhibitors of enzymes: kinetic characteristics and pharmacological interest. BIOMEDITSINSKAIA KHIMIIA 2025; 71:81-94. [PMID: 40326015 DOI: 10.18097/pbmcr1536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Currently, the search for new slow-binding inhibitors of enzymes (SBI) and their identification primary in vitro studies still attracts much attention in the context of their potential role as putative pharmacological agents for the treatment of various diseases. In contrast to their classical reversible analogues, SBI exhibit a slow enzyme binding kinetics, where the equilibrium steady-state is reached not in microseconds, but after longer time intervals. Such compounds could be promising drugs, because regardless of their pharmacokinetics in the bloodstream, they have such advantages as high affinity for the target enzyme, long residence time on the target, and therefore, prolonged action. These pharmacological properties ensure optimized dosage of drugs required to achieve high activity with less side effects. In this review we have considered mechanisms of SBI interaction with enzyme targets, the principles of their recognition at the level of in vitro studies and analysis of binding and kinetic parameters.
Collapse
Affiliation(s)
| | - T N Pashirova
- Kazan (Volga region) Federal University, Kazan, Russia; Arbuzov Institute of Organic and Physical Chemistry, FRC Kazan Scientific Center, Russian Academy of Sciences, Kazan, Russia
| | - P Masson
- Kazan (Volga region) Federal University, Kazan, Russia
| |
Collapse
|
2
|
Phillips AF, Zhang R, Jaffe M, Schulz R, Carty MC, Verma A, Feinberg TY, Arensman MD, Chiu A, Letso R, Bosco N, Queen KA, Racela AR, Stumpff J, Andreu-Agullo C, Bettigole SE, Depetris RS, Drutman S, Su SM, Cogan DA, Eng CH. Targeting chromosomally unstable tumors with a selective KIF18A inhibitor. Nat Commun 2025; 16:307. [PMID: 39747049 PMCID: PMC11697083 DOI: 10.1038/s41467-024-55300-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Chromosome instability is a prevalent vulnerability of cancer cells that has yet to be fully exploited therapeutically. To identify genes uniquely essential to chromosomally unstable cells, we mined the Cancer Dependency Map for genes essential in tumor cells with high levels of copy number aberrations. We identify and validate KIF18A, a mitotic kinesin, as a vulnerability of chromosomally unstable cancer cells. Knockdown of KIF18A leads to mitotic defects and reduction of tumor growth. Screening of a chemical library for inhibitors of KIF18A enzymatic activity identified a hit that was optimized to yield VLS-1272, which is orally bioavailable, potent, ATP non-competitive, microtubule-dependent, and highly selective for KIF18A versus other kinesins. Inhibition of KIF18A's ATPase activity prevents KIF18A translocation across the mitotic spindle, resulting in chromosome congression defects, mitotic cell accumulation, and cell death. Profiling VLS-1272 across >100 cancer cell lines demonstrates that the specificity towards cancer cells with chromosome instability differentiates KIF18A inhibition from other clinically tested anti-mitotic drugs. Treatment of tumor xenografts with VLS-1272 results in mitotic defects leading to substantial, dose-dependent inhibition of tumor growth. The strong biological rationale, robust preclinical data, and optimized compound properties enable the clinical development of a KIF18A inhibitor in cancers with high chromosomal instability.
Collapse
Affiliation(s)
| | | | - Mia Jaffe
- Volastra Therapeutics, New York, NY, USA
| | | | | | | | | | | | - Alan Chiu
- Volastra Therapeutics, New York, NY, USA
| | - Reka Letso
- Volastra Therapeutics, New York, NY, USA
| | | | - Katelyn A Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Allison R Racela
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Voget R, Breidenbach J, Claff T, Hingst A, Sylvester K, Steinebach C, Vu LP, Weiße RH, Bartz U, Sträter N, Müller CE, Gütschow M. Development of an active-site titrant for SARS-CoV-2 main protease as an indispensable tool for evaluating enzyme kinetics. Acta Pharm Sin B 2024; 14:2349-2357. [PMID: 38799620 PMCID: PMC11121168 DOI: 10.1016/j.apsb.2024.03.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/25/2024] [Accepted: 02/27/2024] [Indexed: 05/29/2024] Open
Abstract
A titrant for the SARS-CoV-2 main protease (Mpro) was developed that enables, for the first time, the exact determination of the concentration of the enzymatically active Mpro by active-site titration. The covalent binding mode of the tetrapeptidic titrant was elucidated by the determination of the crystal structure of the enzyme-titrant complex. Four fluorogenic substrates of Mpro, including a prototypical, internally quenched Dabcyl-EDANS peptide, were compared in terms of solubility under typical assay conditions. By exploiting the new titrant, key kinetic parameters for the Mpro-catalyzed cleavage of these substrates were determined.
Collapse
Affiliation(s)
- Rabea Voget
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Julian Breidenbach
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Tobias Claff
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Alexandra Hingst
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Katharina Sylvester
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Christian Steinebach
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Lan Phuong Vu
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Renato H. Weiße
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig 04103, Germany
| | - Ulrike Bartz
- Department of Natural Sciences, University of Applied Sciences Bonn-Rhein-Sieg, Rheinbach 53359, Germany
| | - Norbert Sträter
- Institute of Bioanalytical Chemistry, Center for Biotechnology and Biomedicine, Leipzig University, Leipzig 04103, Germany
| | - Christa E. Müller
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| | - Michael Gütschow
- Pharmaceutical Institute, Pharmaceutical & Medicinal Chemistry, University of Bonn, Bonn 53121, Germany
| |
Collapse
|
4
|
Characterization of Recombinantly-Expressed Hydrolytic Enzymes from Chinese Hamster Ovary Cells: Identification of Host Cell Proteins that Degrade Polysorbate. J Pharm Sci 2023; 112:1351-1363. [PMID: 36646283 DOI: 10.1016/j.xphs.2023.01.003] [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: 09/30/2022] [Revised: 01/03/2023] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Enzymatic hydrolysis of polysorbate in drug products is a major challenge for the biopharmaceutical industry. Polysorbate hydrolysis caused by host cell proteins (HCPs) co-purified during bioprocessing can reduce the protective effects of the surfactant for the active pharmaceutical ingredient and cause the accumulation of low-solubility degradation products over the long-term storage. The identities of such HCPs are elusive due to their extremely low concentrations after the efficient purification processes of most biopharmaceuticals. In this work, 20 enzymes-selected for their known or putative hydrolytic activity and potential to degrade polysorbate-were recombinantly expressed, purified, and characterized via orthogonal methods. First, these recombinant HCPs were assessed for hydrolytic activity against a fluorogenic esterase substrate in a recently-developed, high-throughput assay. Second, these HCPs were screened for hydrolytic activity against polysorbate in a representative mAb formulation. Third, HCPs that displayed hydrolytic activities in the first two assays were subjected to more detailed characterization of their enzyme kinetics against polysorbates. Finally, these HCPs were evaluated for substrate specificity towards different sub-species of polysorbates. This work provides critical new insights for targeted LC-MS/MS approaches for identification of relevant polysorbate-degrading enzymes and supports improvements to remove such HCPs, including knockouts or targeted removal during purification.
Collapse
|
5
|
Huang Y, Cui Y, Deng H, Wang J, Hong R, Hu S, Hou H, Dong Y, Wang H, Chen J, Li L, Xie Y, Sun P, Fu X, Yin L, Xiong W, Shi SH, Luo M, Wang S, Li X, Sheng X. Bioresorbable thin-film silicon diodes for the optoelectronic excitation and inhibition of neural activities. Nat Biomed Eng 2022; 7:486-498. [PMID: 36065014 DOI: 10.1038/s41551-022-00931-0] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 07/25/2022] [Indexed: 11/09/2022]
Abstract
Neural activities can be modulated by leveraging light-responsive nanomaterials as interfaces for exerting photothermal, photoelectrochemical or photocapacitive effects on neurons or neural tissues. Here we show that bioresorbable thin-film monocrystalline silicon pn diodes can be used to optoelectronically excite or inhibit neural activities by establishing polarity-dependent positive or negative photovoltages at the semiconductor/solution interface. Under laser illumination, the silicon-diode optoelectronic interfaces allowed for the deterministic depolarization or hyperpolarization of cultured neurons as well as the upregulated or downregulated intracellular calcium dynamics. The optoelectronic interfaces can also be mounted on nerve tissue to activate or silence neural activities in peripheral and central nervous tissues, as we show in mice with exposed sciatic nerves and somatosensory cortices. Bioresorbable silicon-based optoelectronic thin films that selectively excite or inhibit neural tissue may find advantageous biomedical applicability.
Collapse
Affiliation(s)
- Yunxiang Huang
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China.,School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China.,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China
| | - Yuting Cui
- Chinese Institute for Brain Research, Beijing, China.,National Institute of Biological Sciences, Beijing, China
| | - Hanjie Deng
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Jingjing Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Rongqi Hong
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China
| | - Shuhan Hu
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Hanqing Hou
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Yuanrui Dong
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, China
| | - Huachun Wang
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Junyu Chen
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Lizhu Li
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Yang Xie
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Pengcheng Sun
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Xin Fu
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China
| | - Wei Xiong
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Song-Hai Shi
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Minmin Luo
- IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.,Chinese Institute for Brain Research, Beijing, China.,National Institute of Biological Sciences, Beijing, China.,School of Life Sciences, Tsinghua University, Beijing, China
| | - Shirong Wang
- Beijing Advanced Innovation Center for Intelligent Robots and Systems, Beijing Institute of Technology, Beijing, China.
| | - Xiaojian Li
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, China.
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, Tsinghua University, Beijing, China. .,IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
| |
Collapse
|
6
|
Tawa P, Zhang L, Metwally E, Hou Y, McCoy MA, Seganish WM, Zhang R, Frank E, Sheth P, Hanisak J, Sondey C, Bauman D, Soriano A. Mechanistic insights on novel small molecule allosteric activators of cGMP-dependent protein kinase PKG1α. J Biol Chem 2022; 298:102284. [PMID: 35868561 PMCID: PMC9425037 DOI: 10.1016/j.jbc.2022.102284] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 07/14/2022] [Accepted: 07/16/2022] [Indexed: 11/30/2022] Open
Abstract
cGMP-dependent protein kinase (PKG) represents a compelling drug target for treatment of cardiovascular diseases. PKG1 is the major effector of beneficial cGMP signaling which is involved in smooth muscle relaxation and vascular tone, inhibition of platelet aggregation and signaling that leads to cardioprotection. In this study, a novel piperidine series of activators previously identified from an ultrahigh-throughput screen were validated to directly bind partially activated PKG1α and subsequently enhance its kinase activity in a concentration-dependent manner. Compounds from initial optimization efforts showed an ability to activate PKG1α independent of the endogenous activator, cGMP. We demonstrate these small molecule activators mimic the effect of cGMP on the kinetic parameters of PKG1α by positively modulating the KM of the peptide substrate and negatively modulating the apparent KM for ATP with increase in catalytic efficiency, kcat. In addition, these compounds also allosterically modulate the binding affinity of cGMP for PKG1α by increasing the affinity of cGMP for the high-affinity binding site (CNB-A) and decreasing the affinity of cGMP for the low-affinity binding site (CNB-B). We show the mode of action of these activators involves binding to an allosteric site within the regulatory domain, near the CNB-B binding site. To the best of our knowledge, these are the first reported non-cGMP mimetic small molecules shown to directly activate PKG1α. Insights into the mechanism of action of these compounds will enable future development of cardioprotective compounds that function through novel modes of action for the treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Paul Tawa
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Lei Zhang
- Biologics AR&D Immunoassay Group, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Essam Metwally
- Computational & Structural Chemistry, Merck & Co., Inc., South San Francisco, CA, USA
| | - Yan Hou
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Mark A McCoy
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | - Rumin Zhang
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Emily Frank
- Quantitative Biosciences, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Payal Sheth
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA
| | | | | | - David Bauman
- Discovery Biology, Merck & Co., Inc., Kenilworth, NJ, USA
| | - Aileen Soriano
- Mass Spectrometry & Biophysics, Computational & Structural Chemistry, Merck & Co., Inc., Kenilworth, NJ, USA.
| |
Collapse
|
7
|
Xu H, Zhang Y, Wang P, Zhang J, Chen H, Zhang L, Du X, Zhao C, Wu D, Liu F, Yang H, Liu C. A comprehensive review of integrative pharmacology-based investigation: A paradigm shift in traditional Chinese medicine. Acta Pharm Sin B 2021; 11:1379-1399. [PMID: 34221858 PMCID: PMC8245857 DOI: 10.1016/j.apsb.2021.03.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/12/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
Over the past decade, traditional Chinese medicine (TCM) has widely embraced systems biology and its various data integration approaches to promote its modernization. Thus, integrative pharmacology-based traditional Chinese medicine (TCMIP) was proposed as a paradigm shift in TCM. This review focuses on the presentation of this novel concept and the main research contents, methodologies and applications of TCMIP. First, TCMIP is an interdisciplinary science that can establish qualitative and quantitative pharmacokinetics-pharmacodynamics (PK-PD) correlations through the integration of knowledge from multiple disciplines and techniques and from different PK-PD processes in vivo. Then, the main research contents of TCMIP are introduced as follows: chemical and ADME/PK profiles of TCM formulas; confirming the three forms of active substances and the three action modes; establishing the qualitative PK-PD correlation; and building the quantitative PK-PD correlations, etc. After that, we summarize the existing data resources, computational models and experimental methods of TCMIP and highlight the urgent establishment of mathematical modeling and experimental methods. Finally, we further discuss the applications of TCMIP for the improvement of TCM quality control, clarification of the molecular mechanisms underlying the actions of TCMs and discovery of potential new drugs, especially TCM-related combination drug discovery.
Collapse
|
8
|
Chaudhry C, Tebben A, Tokarski JS, Borzilleri R, Pitts WJ, Lippy J, Zhang L. An innovative kinome platform to accelerate small-molecule inhibitor discovery and optimization from hits to leads. Drug Discov Today 2021; 26:1115-1125. [PMID: 33497831 DOI: 10.1016/j.drudis.2021.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 07/23/2020] [Accepted: 01/18/2021] [Indexed: 01/09/2023]
Abstract
Kinases, accounting for 20% of the human genome, have been the focus of pharmaceutical drug discovery efforts for over three decades. Despite concerns surrounding the tractability of kinases as drug targets, it is evident that kinase drug discovery offers great potential, underscored by the US Food and Drug Administration (FDA) approval of 48 small-molecule kinase inhibitors. Despite these successes, it is challenging to identify novel kinome selective inhibitors with good pharmacokinetic/pharmacodynamic (PK/PD) properties, and resistance to kinase inhibitor treatment frequently arises. A new era of kinase drug discovery predicates the need for diverse and powerful tools to discover the next generation of kinase inhibitors. Here, we outline key tenets of the Bristol Meyers Squibb (BMS) kinase platform, to enable efficient generation of highly optimized kinase inhibitors.
Collapse
Affiliation(s)
- Charu Chaudhry
- Lead Discovery and Optimization, Bristol Myers Squibb, NJ, USA.
| | - Andrew Tebben
- Molecular Structure and Design, Molecular Discovery Technologies, Bristol Myers Squibb, NJ, USA
| | - John S Tokarski
- Molecular Structure and Design, Molecular Discovery Technologies, Bristol Myers Squibb, NJ, USA
| | | | - William J Pitts
- Immunosciences Discovery Chemistry, Bristol Myers Squibb, NJ, USA
| | - Jonathan Lippy
- Lead Discovery and Optimization, Bristol Myers Squibb, NJ, USA
| | - Litao Zhang
- Lead Discovery and Optimization, Bristol Myers Squibb, NJ, USA
| |
Collapse
|
9
|
Rufer AC. Drug discovery for enzymes. Drug Discov Today 2021; 26:875-886. [PMID: 33454380 DOI: 10.1016/j.drudis.2021.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 12/21/2020] [Accepted: 01/07/2021] [Indexed: 02/06/2023]
Abstract
Enzymes are essential, physiological catalysts involved in all processes of life, including metabolism, cellular signaling and motility, as well as cell growth and division. They are attractive drug targets because of the presence of defined substrate-binding pockets, which can be exploited as binding sites for pharmaceutical enzyme inhibitors. Understanding the reaction mechanisms of enzymes and the molecular mode of action of enzyme inhibitors is indispensable for the discovery and development of potent, efficacious, and safe novel drugs. The combination of classical concepts of enzymology with new experimental and data analysis methods opens new routes for drug discovery.
Collapse
Affiliation(s)
- Arne Christian Rufer
- Pharma Research and Early Development, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 065/208A, 4070 Basel, Switzerland.
| |
Collapse
|
10
|
Kruk M, Widstrom N, Jena S, Wolter NL, Blankenhorn JF, Abdalla I, Yang TY, Parker LL. Assays for tyrosine phosphorylation in human cells. Methods Enzymol 2019; 626:375-406. [PMID: 31606083 DOI: 10.1016/bs.mie.2019.06.026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Tyrosine kinases are important for many cellular processes and disruption of their regulation is a factor in diseases like cancer, therefore they are a major target of anticancer drugs. There are many ways to measure tyrosine kinase activity in cells by monitoring endogenous substrate phosphorylation, or by using peptide substrates and incubating them with cell lysates containing active kinases. However, most of these strategies rely on antibodies and/or are limited in how accurately they model the intracellular environment. In cases in which activity needs to be measured in cells, but endogenous substrates are not known and/or suitable phosphospecific antibodies are not available, cell-deliverable peptide substrates can be an alternative and can provide information on activation and inhibition of kinases in intact, live cells. In this chapter, we review this methodology and provide a protocol for measuring Abl kinase activity in human cells using enzyme-linked immunosorbent assay (ELISA) with a generic antiphosphotyrosine antibody for detection.
Collapse
Affiliation(s)
- Monica Kruk
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Naomi Widstrom
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Sampreeti Jena
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Nicole L Wolter
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - John F Blankenhorn
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Ibrahim Abdalla
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Tzu-Yi Yang
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States
| | - Laurie L Parker
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, United States.
| |
Collapse
|
11
|
Young K, Painter RE, Raghoobar SL, Hairston NN, Racine F, Wisniewski D, Balibar CJ, Villafania A, Zhang R, Sahm DF, Blizzard T, Murgolo N, Hammond ML, Motyl MR. In vitro studies evaluating the activity of imipenem in combination with relebactam against Pseudomonas aeruginosa. BMC Microbiol 2019; 19:150. [PMID: 31272373 PMCID: PMC6610938 DOI: 10.1186/s12866-019-1522-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND The prevalence of antibiotic resistance is increasing, and multidrug-resistant Pseudomonas aeruginosa has been identified as a serious threat to human health. The production of β-lactamase is a key mechanism contributing to imipenem resistance in P. aeruginosa. Relebactam is a novel β-lactamase inhibitor, active against class A and C β-lactamases, that has been shown to restore imipenem susceptibility. In a series of studies, we assessed the interaction of relebactam with key mechanisms involved in carbapenem resistance in P. aeruginosa and to what extent relebactam might overcome imipenem non-susceptibility. RESULTS Relebactam demonstrated no intrinsic antibacterial activity against P. aeruginosa, had no inoculum effect, and was not subject to efflux. Enzymology studies showed relebactam is a potent (overall inhibition constant: 27 nM), practically irreversible inhibitor of P. aeruginosa AmpC. Among P. aeruginosa clinical isolates from the SMART global surveillance program (2009, n = 993; 2011, n = 1702; 2015, n = 5953; 2016, n = 6165), imipenem susceptibility rates were 68.4% in 2009, 67.4% in 2011, 70.4% in 2015, and 67.3% in 2016. With the addition of 4 μg/mL relebactam, imipenem susceptibility rates increased to 87.6, 86.0, 91.7, and 89.8%, respectively. When all imipenem-non-susceptible isolates were pooled, the addition of 4 μg/mL relebactam reduced the mode imipenem minimum inhibitory concentration (MIC) 8-fold (from 16 μg/mL to 2 μg/mL) among all imipenem-non-susceptible isolates. Of 3747 imipenem-non-susceptible isolates that underwent molecular profiling, 1200 (32%) remained non-susceptible to the combination imipenem/relebactam (IMI/REL); 42% of these encoded class B metallo-β-lactamases, 11% encoded a class A GES enzyme, and no class D enzymes were detected. No relationship was observed between alleles of the chromosomally-encoded P. aeruginosa AmpC and IMI/REL MIC. CONCLUSIONS IMI/REL exhibited potential in the treatment of carbapenem-resistant P. aeruginosa infections, with the exception of isolates encoding class B, some GES alleles, and class D carbapenemases.
Collapse
Affiliation(s)
- Katherine Young
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Ronald E. Painter
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Susan L. Raghoobar
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | | | - Fred Racine
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Douglas Wisniewski
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Carl J. Balibar
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Artjohn Villafania
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Rumin Zhang
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | | | - Timothy Blizzard
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Nicholas Murgolo
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Milton L. Hammond
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| | - Mary R. Motyl
- Merck & Co., Inc., 2015 Galloping Hill Road MN-410, Kenilworth, NJ 07033 USA
| |
Collapse
|
12
|
Barros LF, Bolaños JP, Bonvento G, Bouzier-Sore AK, Brown A, Hirrlinger J, Kasparov S, Kirchhoff F, Murphy AN, Pellerin L, Robinson MB, Weber B. Current technical approaches to brain energy metabolism. Glia 2018; 66:1138-1159. [PMID: 29110344 PMCID: PMC5903992 DOI: 10.1002/glia.23248] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Revised: 09/14/2017] [Accepted: 10/04/2017] [Indexed: 12/19/2022]
Abstract
Neuroscience is a technology-driven discipline and brain energy metabolism is no exception. Once satisfied with mapping metabolic pathways at organ level, we are now looking to learn what it is exactly that metabolic enzymes and transporters do and when, where do they reside, how are they regulated, and how do they relate to the specific functions of neurons, glial cells, and their subcellular domains and organelles, in different areas of the brain. Moreover, we aim to quantify the fluxes of metabolites within and between cells. Energy metabolism is not just a necessity for proper cell function and viability but plays specific roles in higher brain functions such as memory processing and behavior, whose mechanisms need to be understood at all hierarchical levels, from isolated proteins to whole subjects, in both health and disease. To this aim, the field takes advantage of diverse disciplines including anatomy, histology, physiology, biochemistry, bioenergetics, cellular biology, molecular biology, developmental biology, neurology, and mathematical modeling. This article presents a well-referenced synopsis of the technical side of brain energy metabolism research. Detail and jargon are avoided whenever possible and emphasis is given to comparative strengths, limitations, and weaknesses, information that is often not available in regular articles.
Collapse
Affiliation(s)
- L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, 5110466, Chile
| | - Juan P Bolaños
- Instituto de Biologia Funcional y Genomica-CSIC, Universidad de Salamanca, CIBERFES, Salamanca, 37007, Spain
| | - Gilles Bonvento
- Commissariat à l'Energie Atomique et aux Energies Alternatives (CEA), Département de la Recherche Fondamentale (DRF), Institut de Biologie François Jacob, Molecular Imaging Research Center (MIRCen), CNRS UMR 9199, Université Paris-Sud, Université Paris-Saclay, Fontenay-aux-Roses, France
| | - Anne-Karine Bouzier-Sore
- Centre de Résonance Magnétique des Systèmes Biologiques UMR 5536, CNRS-Université Bordeaux 146 rue Léo-Saignat, Bordeaux, France
| | - Angus Brown
- School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Johannes Hirrlinger
- Carl Ludwig Institute of Physiology, University of Leipzig, Liebigstr. 27, D-04103, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute for Experimental Medicine, Hermann-Rein-Str. 3, Göttingen, D-37075, Germany
| | - Sergey Kasparov
- School of Physiology, Pharmacology and Neuroscience, University of Bristol, University Walk, BS8 1TD, United Kingdom
- Baltic Federal University, Kalinigrad, Russian Federation
| | - Frank Kirchhoff
- Molecular Physiology, Center for Integrative Physiology and Molecular Medicine, University of Saarland, Building 48, Homburg, 66421, Germany
| | - Anne N Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, CA, 92093
| | - Luc Pellerin
- Département de Physiologie, 7 rue du Bugnon, Lausanne, CH1005, Switzerland
| | - Michael B Robinson
- Department of Pediatrics, and Department of Systems Pharmacology and Translational Therapeutics, Children's Hospital of Philadelphia Research Institute, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|