1
|
Glocker UM, Braun F, Eberl HC, Bantscheff M. A Probe-Based Target Engagement Assay for Kinases in Live Cells. Mol Cell Proteomics 2025; 24:100963. [PMID: 40187494 PMCID: PMC12076712 DOI: 10.1016/j.mcpro.2025.100963] [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: 11/14/2024] [Accepted: 02/27/2025] [Indexed: 04/07/2025] Open
Abstract
The efficacy and safety of kinase inhibitor drugs are largely influenced by their selectivity. Available profiling technologies are primarily based on overexpressed or endogenously expressed kinases in cell extracts. We compared kinase capture with the cell penetrant covalent probe XO44 to three derivatives and found that replacing the alkyne handle with a trans-cyclooctene group allowed the development of a more robust kinase capture and enrichment protocol. An intracellular chemoproteomics target profiling and engagement assay was devised by optimizing probe concentration and incubation time and using an isobaric mass tag-based strategy for relative quantification. Comparing intracellular kinase profiles of the marketed drug dasatinib and the tool compound dinaciclib with the lysate-based kinobeads assay revealed excellent agreement in rank-order of binding. Dinaciclib showed a systematic shift to higher IC50s, suggesting that intracellular cosubstrate concentrations, cell penetration of the compound, as well as kinase localization and complexes in live cells influence target profiles. Further, we show that sepiapterin reductase SPR and multidrug resistance protein 1 ABCC1 are off-targets of kinase inhibitor scaffolds with potential implications on efficacy and safety.
Collapse
Affiliation(s)
| | - Florian Braun
- Chemical Synthesis Core Facility, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | | |
Collapse
|
2
|
Eckert S, Berner N, Kramer K, Schneider A, Müller J, Lechner S, Brajkovic S, Sakhteman A, Graetz C, Fackler J, Dudek M, Pfaffl MW, Knolle P, Wilhelm S, Kuster B. Decrypting the molecular basis of cellular drug phenotypes by dose-resolved expression proteomics. Nat Biotechnol 2025; 43:406-415. [PMID: 38714896 PMCID: PMC11919725 DOI: 10.1038/s41587-024-02218-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 03/25/2024] [Indexed: 03/20/2025]
Abstract
Proteomics is making important contributions to drug discovery, from target deconvolution to mechanism of action (MoA) elucidation and the identification of biomarkers of drug response. Here we introduce decryptE, a proteome-wide approach that measures the full dose-response characteristics of drug-induced protein expression changes that informs cellular drug MoA. Assaying 144 clinical drugs and research compounds against 8,000 proteins resulted in more than 1 million dose-response curves that can be interactively explored online in ProteomicsDB and a custom-built Shiny App. Analysis of the collective data provided molecular explanations for known phenotypic drug effects and uncovered new aspects of the MoA of human medicines. We found that histone deacetylase inhibitors potently and strongly down-regulated the T cell receptor complex resulting in impaired human T cell activation in vitro and ex vivo. This offers a rational explanation for the efficacy of histone deacetylase inhibitors in certain lymphomas and autoimmune diseases and explains their poor performance in treating solid tumors.
Collapse
Affiliation(s)
- Stephan Eckert
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Nicola Berner
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Center Technical University of Munich, Munich, Germany
- German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Karl Kramer
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Annika Schneider
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Julian Müller
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Severin Lechner
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Sarah Brajkovic
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Amirhossein Sakhteman
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Christian Graetz
- Chair of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Jonas Fackler
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Michael Dudek
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Michael W Pfaffl
- Chair of Animal Physiology and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Percy Knolle
- Institute of Molecular Immunology and Experimental Oncology, School of Medicine and Health, Technical University of Munich, Munich, Germany
| | - Stephanie Wilhelm
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany.
- German Cancer Consortium (DKTK), partner site Munich, a partnership between DKFZ and University Center Technical University of Munich, Munich, Germany.
| |
Collapse
|
3
|
Shields JD, Baker D, Balazs AYS, Bommakanti G, Casella R, Cao S, Cook S, Escobar RA, Fawell S, Gibbons FD, Giblin KA, Goldberg FW, Gosselin E, Grebe T, Hariparsad N, Hatoum-Mokdad H, Howells R, Hughes SJ, Jackson A, Karapa Reddy I, Kettle JG, Lamont GM, Lamont S, Li M, Lill SON, Mele DA, Metrano AJ, Mfuh AM, Morrill LA, Peng B, Pflug A, Proia TA, Rezaei H, Richards R, Richter M, Robbins KJ, San Martin M, Schimpl M, Schuller AG, Sha L, Shen M, Sheppeck JE, Singh M, Stokes S, Song K, Sun Y, Tang H, Wagner DJ, Wang J, Wang Y, Wilson DM, Wu A, Wu C, Wu D, Wu Y, Xu K, Yang Y, Yao T, Ye M, Zhang AX, Zhang H, Zhai X, Zhou Y, Ziegler RE, Grimster NP. Discovery and Optimization of Pyrazine Carboxamide AZ3246, a Selective HPK1 Inhibitor. J Med Chem 2025; 68:4582-4595. [PMID: 39928839 DOI: 10.1021/acs.jmedchem.4c02631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
Hematopoietic progenitor kinase 1 (HPK1) is a negative regulator of the T cell receptor signaling pathway and is therefore a target of interest for immunooncology. Nonselective HPK1 inhibitors may affect other kinase components of T cell activation, blunting the beneficial impact of enhanced T cell activity that results from HPK1 inhibition itself. Here, we report the discovery of pyrazine carboxamide HPK1 inhibitors and their optimization through structure-based drug design to afford a highly selective HPK1 inhibitor, compound 24 (AZ3246). This compound induces IL-2 secretion in T cells with an EC50 of 90 nM without inhibiting antagonistic kinases, exhibits pharmacokinetic properties consistent with oral dosing, and demonstrates antitumor activity in the EMT6 syngeneic mouse model.
Collapse
Affiliation(s)
- Jason D Shields
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - David Baker
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Amber Y S Balazs
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Gayathri Bommakanti
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Robert Casella
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Shenggen Cao
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Steve Cook
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Randolph A Escobar
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Stephen Fawell
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Francis D Gibbons
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Kathryn A Giblin
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | | | - Eric Gosselin
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Tyler Grebe
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Niresh Hariparsad
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Holia Hatoum-Mokdad
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Rachel Howells
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Samantha J Hughes
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Anne Jackson
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Iswarya Karapa Reddy
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Jason G Kettle
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Gillian M Lamont
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Scott Lamont
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Min Li
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Sten O Nilsson Lill
- Pharmaceutical Sciences, R&D, AstraZeneca, Pepparedsleden 1, SE-431 83 Mölndal, Sweden
| | - Deanna A Mele
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Anthony J Metrano
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Adelphe M Mfuh
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Lucas A Morrill
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Bo Peng
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Alexander Pflug
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Theresa A Proia
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Hadi Rezaei
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Ryan Richards
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Magdalena Richter
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Kevin J Robbins
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Maryann San Martin
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Marianne Schimpl
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Alwin G Schuller
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Li Sha
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Minhui Shen
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - James E Sheppeck
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Meha Singh
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Stephen Stokes
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Kun Song
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yuanyuan Sun
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Haoran Tang
- Discovery Sciences, R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - David J Wagner
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Jianyan Wang
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yanjun Wang
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - David M Wilson
- Early Oncology R&D, AstraZeneca, 1 Francis Crick Avenue, Cambridge CB2 0AA, U.K
| | - Allan Wu
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Chengyan Wu
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Dedong Wu
- Advanced Drug Delivery, Pharmaceutical Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Ye Wu
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Kevin Xu
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yue Yang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Tieguang Yao
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Minwei Ye
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Andrew X Zhang
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Hui Zhang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Xiang Zhai
- Discovery Sciences, R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Yanxiao Zhou
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, P. R. China
| | - Robert E Ziegler
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| | - Neil P Grimster
- Early Oncology R&D, AstraZeneca, 35 Gatehouse Drive, Waltham, Massachusetts 02451, United States
| |
Collapse
|
4
|
Szymczyk P, Majewska M, Nowak J. Proteins and DNA Sequences Interacting with Tanshinones and Tanshinone Derivatives. Int J Mol Sci 2025; 26:848. [PMID: 39859562 PMCID: PMC11765770 DOI: 10.3390/ijms26020848] [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: 12/20/2024] [Revised: 01/14/2025] [Accepted: 01/19/2025] [Indexed: 01/27/2025] Open
Abstract
Tanshinones, biologically active diterpene compounds derived from Salvia miltiorrhiza, interact with specific proteins and DNA sequences, influencing signaling pathways in animals and humans. This study highlights tanshinone-protein interactions observed at concentrations achievable in vivo, ensuring greater physiological relevance compared to in vitro studies that often employ supraphysiological ligand levels. Experimental data suggest that while tanshinones interact with multiple proteomic targets, only a few enzymes are significantly affected at biologically relevant concentrations. This apparent paradox may be resolved by tanshinones' ability to bind DNA and influence enzymes involved in gene expression or mRNA stability, such as RNA polymerase II and human antigen R protein. These interactions trigger secondary, widespread changes in gene expression, leading to complex proteomic alterations. Although the current understanding of tanshinone-protein interactions remains incomplete, this study provides a foundation for deciphering the molecular mechanisms underlying the therapeutic effects of S. miltiorrhiza diterpenes. Additionally, numerous tanshinone derivatives have been developed to enhance pharmacokinetic properties and biological activity. However, their safety profiles remain poorly characterized, limiting comprehensive insights into their medicinal potential. Further investigation is essential to fully elucidate the therapeutic and toxicological properties of both native and modified tanshinones.
Collapse
Affiliation(s)
- Piotr Szymczyk
- Department of Biology and Pharmaceutical Botany, Medical University of Lodz, Muszyńskiego 1, 90-151 Lodz, Poland
| | - Małgorzata Majewska
- Department of Oncobiology and Epigenetics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland;
| | - Jadwiga Nowak
- Department of Pharmacology and Therapeutics, School of Biomedical Sciences, College of Health Sciences, Makerere University, Kampala P.O. Box 7062, Uganda;
| |
Collapse
|
5
|
Liu Y, Sundah NR, Ho NRY, Shen WX, Xu Y, Natalia A, Yu Z, Seet JE, Chan CW, Loh TP, Lim BY, Shao H. Bidirectional linkage of DNA barcodes for the multiplexed mapping of higher-order protein interactions in cells. Nat Biomed Eng 2024; 8:909-923. [PMID: 38898172 DOI: 10.1038/s41551-024-01225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 05/05/2024] [Indexed: 06/21/2024]
Abstract
Capturing the full complexity of the diverse hierarchical interactions in the protein interactome is challenging. Here we report a DNA-barcoding method for the multiplexed mapping of pairwise and higher-order protein interactions and their dynamics within cells. The method leverages antibodies conjugated with barcoded DNA strands that can bidirectionally hybridize and covalently link to linearize closely spaced interactions within individual 3D protein complexes, encoding and decoding the protein constituents and the interactions among them. By mapping protein interactions in cancer cells and normal cells, we found that tumour cells exhibit a larger diversity and abundance of protein complexes with higher-order interactions. In biopsies of human breast-cancer tissue, the method accurately identified the cancer subtype and revealed that higher-order protein interactions are associated with cancer aggressiveness.
Collapse
Affiliation(s)
- Yu Liu
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Noah R Sundah
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Nicholas R Y Ho
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
| | - Wan Xiang Shen
- Department of Pharmacy and Pharmaceutical Sciences, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Yun Xu
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Auginia Natalia
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Zhonglang Yu
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore
| | - Ju Ee Seet
- Department of Pathology, National University Hospital, Singapore, Singapore
| | - Ching Wan Chan
- Department of Surgery, National University Hospital, Singapore, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Tze Ping Loh
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore
| | - Brian Y Lim
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore.
- Department of Computer Science, School of Computing, National University of Singapore, Singapore, Singapore.
| | - Huilin Shao
- Institute for Health Innovation and Technology, National University of Singapore, Singapore, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore.
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore, Singapore.
| |
Collapse
|
6
|
Chamrád I, Simerský R, Lenobel R, Novák O. Exploring affinity chromatography in proteomics: A comprehensive review. Anal Chim Acta 2024; 1306:342513. [PMID: 38692783 DOI: 10.1016/j.aca.2024.342513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 05/03/2024]
Abstract
Over the past decades, the proteomics field has undergone rapid growth. Progress in mass spectrometry and bioinformatics, together with separation methods, has brought many innovative approaches to the study of the molecular biology of the cell. The potential of affinity chromatography was recognized immediately after its first application in proteomics, and since that time, it has become one of the cornerstones of many proteomic protocols. Indeed, this chromatographic technique exploiting the specific binding between two molecules has been employed for numerous purposes, from selective removal of interfering (over)abundant proteins or enrichment of scarce biomarkers in complex biological samples to mapping the post-translational modifications and protein interactions with other proteins, nucleic acids or biologically active small molecules. This review presents a comprehensive survey of this versatile analytical tool in current proteomics. To navigate the reader, the haphazard space of affinity separations is classified according to the experiment's aims and the separated molecule's nature. Different types of available ligands and experimental strategies are discussed in further detail for each of the mentioned procedures.
Collapse
Affiliation(s)
- Ivo Chamrád
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 241/27, CZ-77900, Olomouc, Holice, Czech Republic.
| | - Radim Simerský
- Department of Chemical Biology, Faculty of Science, Palacký University, Šlechtitelů 241/27, CZ-77900, Olomouc, Holice, Czech Republic
| | - René Lenobel
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 241/27, CZ-77900, Olomouc, Holice, Czech Republic
| | - Ondřej Novák
- Laboratory of Growth Regulators, Faculty of Science, Palacký University and Institute of Experimental Botany of the Czech Academy of Sciences, Šlechtitelů 241/27, CZ-77900, Olomouc, Holice, Czech Republic
| |
Collapse
|
7
|
Liao Y, Remsing Rix LL, Li X, Fang B, Izumi V, Welsh EA, Monastyrskyi A, Haura EB, Koomen JM, Doebele RC, Rix U. Differential network analysis of ROS1 inhibitors reveals lorlatinib polypharmacology through co-targeting PYK2. Cell Chem Biol 2024; 31:284-297.e10. [PMID: 37848034 PMCID: PMC10922442 DOI: 10.1016/j.chembiol.2023.09.011] [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: 01/30/2023] [Revised: 08/02/2023] [Accepted: 09/19/2023] [Indexed: 10/19/2023]
Abstract
Multiple tyrosine kinase inhibitors (TKIs) are often developed for the same indication. However, their relative overall efficacy is frequently incompletely understood and they may harbor unrecognized targets that cooperate with the intended target. We compared several ROS1 TKIs for inhibition of ROS1-fusion-positive lung cancer cell viability, ROS1 autophosphorylation and kinase activity, which indicated disproportionately higher cellular potency of one TKI, lorlatinib. Quantitative chemical and phosphoproteomics across four ROS1 TKIs and differential network analysis revealed that lorlatinib uniquely impacted focal adhesion signaling. Functional validation using pharmacological probes, RNA interference, and CRISPR-Cas9 knockout uncovered a polypharmacology mechanism of lorlatinib by dual targeting ROS1 and PYK2, which form a multiprotein complex with SRC. Rational multi-targeting of this complex by combining lorlatinib with SRC inhibitors exhibited pronounced synergy. Taken together, we show that systems pharmacology-based differential network analysis can dissect mixed canonical/non-canonical polypharmacology mechanisms across multiple TKIs enabling the design of rational drug combinations.
Collapse
Affiliation(s)
- Yi Liao
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Xueli Li
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Bin Fang
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Victoria Izumi
- Proteomics and Metabolomics Core, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Eric A Welsh
- Biostatistics and Bioinformatics Shared Resource, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Andrii Monastyrskyi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA
| | - Eric B Haura
- Department of Thoracic Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - John M Koomen
- Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA; Department of Molecular Oncology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA
| | - Robert C Doebele
- Division of Medical Oncology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL 33612, USA; Department of Oncologic Sciences, University of South Florida, Tampa, FL 33620, USA.
| |
Collapse
|
8
|
Stephenson EH, Higgins JMG. Pharmacological approaches to understanding protein kinase signaling networks. Front Pharmacol 2023; 14:1310135. [PMID: 38164473 PMCID: PMC10757940 DOI: 10.3389/fphar.2023.1310135] [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: 10/09/2023] [Accepted: 11/27/2023] [Indexed: 01/03/2024] Open
Abstract
Protein kinases play vital roles in controlling cell behavior, and an array of kinase inhibitors are used successfully for treatment of disease. Typical drug development pipelines involve biological studies to validate a protein kinase target, followed by the identification of small molecules that effectively inhibit this target in cells, animal models, and patients. However, it is clear that protein kinases operate within complex signaling networks. These networks increase the resilience of signaling pathways, which can render cells relatively insensitive to inhibition of a single kinase, and provide the potential for pathway rewiring, which can result in resistance to therapy. It is therefore vital to understand the properties of kinase signaling networks in health and disease so that we can design effective multi-targeted drugs or combinations of drugs. Here, we outline how pharmacological and chemo-genetic approaches can contribute to such knowledge, despite the known low selectivity of many kinase inhibitors. We discuss how detailed profiling of target engagement by kinase inhibitors can underpin these studies; how chemical probes can be used to uncover kinase-substrate relationships, and how these tools can be used to gain insight into the configuration and function of kinase signaling networks.
Collapse
Affiliation(s)
| | - Jonathan M. G. Higgins
- Faculty of Medical Sciences, Biosciences Institute, Newcastle University, Newcastle uponTyne, United Kingdom
| |
Collapse
|
9
|
Noberini R, Bonaldi T. Proteomics contributions to epigenetic drug discovery. Proteomics 2023; 23:e2200435. [PMID: 37727062 DOI: 10.1002/pmic.202200435] [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: 06/09/2023] [Revised: 08/30/2023] [Accepted: 08/31/2023] [Indexed: 09/21/2023]
Abstract
The combined activity of epigenetic features, which include histone post-translational modifications, DNA methylation, and nucleosome positioning, regulates gene expression independently from changes in the DNA sequence, defining how the shared genetic information of an organism is used to generate different cell phenotypes. Alterations in epigenetic processes have been linked with a multitude of diseases, including cancer, fueling interest in the discovery of drugs targeting the proteins responsible for writing, erasing, or reading histone and DNA modifications. Mass spectrometry (MS)-based proteomics has emerged as a versatile tool that can assist drug discovery pipelines from target validation, through target deconvolution, to monitoring drug efficacy in vivo. Here, we provide an overview of the contributions of MS-based proteomics to epigenetic drug discovery, describing the main approaches that can be used to support different drug discovery pipelines and highlighting how they contributed to the development and characterization of epigenetic drugs.
Collapse
Affiliation(s)
- Roberta Noberini
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Tiziana Bonaldi
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hematology-Oncology, University of Milan, Milan, Italy
| |
Collapse
|
10
|
Griffin ME, Thompson JW, Xiao Y, Sweredoski MJ, Aksenfeld RB, Jensen EH, Koldobskaya Y, Schacht AL, Kim TD, Choudhry P, Lomenick B, Garbis SD, Moradian A, Hsieh-Wilson LC. Functional glycoproteomics by integrated network assembly and partitioning. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.13.541482. [PMID: 37398272 PMCID: PMC10312638 DOI: 10.1101/2023.06.13.541482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
The post-translational modification (PTM) of proteins by O-linked β-N-acetyl-D-glucosamine (O-GlcNAcylation) is widespread across the proteome during the lifespan of all multicellular organisms. However, nearly all functional studies have focused on individual protein modifications, overlooking the multitude of simultaneous O-GlcNAcylation events that work together to coordinate cellular activities. Here, we describe Networking of Interactors and SubstratEs (NISE), a novel, systems-level approach to rapidly and comprehensively monitor O-GlcNAcylation across the proteome. Our method integrates affinity purification-mass spectrometry (AP-MS) and site-specific chemoproteomic technologies with network generation and unsupervised partitioning to connect potential upstream regulators with downstream targets of O-GlcNAcylation. The resulting network provides a data-rich framework that reveals both conserved activities of O-GlcNAcylation such as epigenetic regulation as well as tissue-specific functions like synaptic morphology. Beyond O-GlcNAc, this holistic and unbiased systems-level approach provides a broadly applicable framework to study PTMs and discover their diverse roles in specific cell types and biological states.
Collapse
Affiliation(s)
- Matthew E. Griffin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Co-first author
| | - John W. Thompson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Co-first author
| | - Yao Xiao
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Co-first author
| | - Michael J. Sweredoski
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Rita B. Aksenfeld
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Elizabeth H. Jensen
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Yelena Koldobskaya
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Andrew L. Schacht
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Terry D. Kim
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Priya Choudhry
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Brett Lomenick
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Spiros D. Garbis
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Annie Moradian
- Proteome Exploration Laboratory, Beckman Institute, California Institute of Technology, Pasadena, CA 91125, USA
| | - Linda C. Hsieh-Wilson
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Lead contact
| |
Collapse
|
11
|
Zambo B, Morlet B, Negroni L, Trave G, Gogl G. Native holdup (nHU) to measure binding affinities from cell extracts. SCIENCE ADVANCES 2022; 8:eade3828. [PMID: 36542723 PMCID: PMC9770967 DOI: 10.1126/sciadv.ade3828] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Characterizing macromolecular interactions is essential for understanding cellular processes, yet most methods currently used to detect protein interactions from cells are qualitative. Here, we introduce the native holdup (nHU) approach to estimate equilibrium binding constants of protein interactions directly from cell extracts. Compared to other pull-down-based assays, nHU requires less sample preparation and can be coupled to any analytical methods as readouts, such as Western blotting or mass spectrometry. We use nHU to explore interactions of SNX27, a cargo adaptor of the retromer complex and find good agreement between in vitro affinities and those measured directly from cell extracts using nHU. We discuss the strengths and limitations of nHU and provide simple protocols that can be implemented in most laboratories.
Collapse
Affiliation(s)
- Boglarka Zambo
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, Illkirch F-67404, France
| | - Bastien Morlet
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, Illkirch F-67404, France
| | - Luc Negroni
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, Illkirch F-67404, France
| | - Gilles Trave
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, Illkirch F-67404, France
- Corresponding author. (G.T.); (G.G.)
| | - Gergo Gogl
- Équipe Labellisée Ligue 2015, Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U1258/CNRS UMR 7104/Université de Strasbourg, 1 rue Laurent Fries, BP 10142, Illkirch F-67404, France
- Corresponding author. (G.T.); (G.G.)
| |
Collapse
|
12
|
Melder FTI, Lindemann P, Welle A, Trouillet V, Heißler S, Nazaré M, Selbach M. Compound Interaction Screen on a Photoactivatable Cellulose Membrane (CISCM) Identifies Drug Targets. ChemMedChem 2022; 17:e202200346. [PMID: 35867055 PMCID: PMC9826412 DOI: 10.1002/cmdc.202200346] [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: 07/01/2022] [Indexed: 01/11/2023]
Abstract
Identifying the protein targets of drugs is an important but tedious process. Existing proteomic approaches enable unbiased target identification but lack the throughput needed to screen larger compound libraries. Here, we present a compound interaction screen on a photoactivatable cellulose membrane (CISCM) that enables target identification of several drugs in parallel. To this end, we use diazirine-based undirected photoaffinity labeling (PAL) to immobilize compounds on cellulose membranes. Functionalized membranes are then incubated with protein extract and specific targets are identified via quantitative affinity purification and mass spectrometry. CISCM reliably identifies known targets of natural products in less than three hours of analysis time per compound. In summary, we show that combining undirected photoimmobilization of compounds on cellulose with quantitative interaction proteomics provides an efficient means to identify the targets of natural products.
Collapse
Affiliation(s)
- F. Teresa I. Melder
- Proteome Dynamics LabMax Delbruck Center for Molecular Medicine in the Helmholtz AssociationRobert-Roessle-Str. 1013125BerlinGermany
| | - Peter Lindemann
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)13125BerlinGermany
| | - Alexander Welle
- Institute of Functional Interfaces and Karlsruhe Nano Micro Facility (KNMFi)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz-Platz 176344Eggenstein-LeopoldshafenGermany
| | - Vanessa Trouillet
- Institute for Applied Materials (IAM-ESS) and Karlsruhe Nano Micro Facility (KNMFi)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz-Platz 176344Eggenstein-LeopoldshafenGermany
| | - Stefan Heißler
- Institute of Functional Interfaces and Karlsruhe Nano Micro Facility (KNMFi)Karlsruhe Institute of Technology (KIT)Hermann-von-Helmholtz-Platz 176344Eggenstein-LeopoldshafenGermany
| | - Marc Nazaré
- Medicinal ChemistryLeibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)13125BerlinGermany
| | - Matthias Selbach
- Proteome Dynamics LabMax Delbruck Center for Molecular Medicine in the Helmholtz AssociationRobert-Roessle-Str. 1013125BerlinGermany
| |
Collapse
|
13
|
Zhuo FF, Guo Q, Zheng YZ, Liu TT, Yang Z, Xu QH, Jiang Y, Liu D, Zeng KW, Tu PF. Photoaffinity Labeling-Based Chemoproteomic Strategy Reveals RBBP4 as a Cellular Target of Protopanaxadiol against Colorectal Cancer Cells. Chembiochem 2022; 23:e202200038. [PMID: 35442561 DOI: 10.1002/cbic.202200038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/19/2022] [Indexed: 12/09/2022]
Abstract
Protopanaxadiol (PPD), a main ginseng metabolite, exerts powerful anticancer effects against multiple types of cancer; however, its cellular targets remain elusive. Here, we synthesized a cell-permeable PPD probe via introducing a bifunctional alkyne-containing diazirine photo-crosslinker and performed a photoaffinity labeling-based chemoproteomic study. We identified retinoblastoma binding protein 4 (RBBP4), a chromatin remodeling factor, as an essential cellular target of PPD in HCT116 colorectal cancer cells. PPD significantly decreased RBBP4-dependent trimethylation at lysine 27 of histone H3 (H3K27me3), a crucial epigenetic marker that correlates with histologic signs of colorectal cancer aggressiveness, and PPD inhibition of proliferation and migration of HCT116 cells was antagonized by RBBP4 RNA silencing. Collectively, our study highlights a previously undisclosed anti-colorectal cancer cellular target of the ginseng metabolite and advances the fundamental understanding of RBBP4 functions via a chemical biology strategy.
Collapse
Affiliation(s)
- Fang-Fang Zhuo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Guo
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yong-Zhe Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Ting-Ting Liu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhuo Yang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qi-He Xu
- Renal Science and Integrative Chinese Medicine Laboratory, Department of Inflammation Biology, School of Immunology and Microbial Sciences, King's College London, WC2R 2LS, London, UK
| | - Yong Jiang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Dan Liu
- Proteomics Laboratory, Medical and Healthy Analytical Center, Peking University Health Science Center, Beijing, 100191, China
| | - Ke-Wu Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Peng-Fei Tu
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| |
Collapse
|
14
|
The emerging role of mass spectrometry-based proteomics in drug discovery. Nat Rev Drug Discov 2022; 21:637-654. [PMID: 35351998 DOI: 10.1038/s41573-022-00409-3] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Proteins are the main targets of most drugs; however, system-wide methods to monitor protein activity and function are still underused in drug discovery. Novel biochemical approaches, in combination with recent developments in mass spectrometry-based proteomics instrumentation and data analysis pipelines, have now enabled the dissection of disease phenotypes and their modulation by bioactive molecules at unprecedented resolution and dimensionality. In this Review, we describe proteomics and chemoproteomics approaches for target identification and validation, as well as for identification of safety hazards. We discuss innovative strategies in early-stage drug discovery in which proteomics approaches generate unique insights, such as targeted protein degradation and the use of reactive fragments, and provide guidance for experimental strategies crucial for success.
Collapse
|
15
|
Brehmer D, Beke L, Wu T, Millar HJ, Moy C, Sun W, Mannens G, Pande V, Boeckx A, van Heerde E, Nys T, Gustin EM, Verbist B, Zhou L, Fan Y, Bhargava V, Safabakhsh P, Vinken P, Verhulst T, Gilbert A, Rai S, Graubert TA, Pastore F, Fiore D, Gu J, Johnson A, Philippar U, Morschhäuser B, Walker D, De Lange D, Keersmaekers V, Viellevoye M, Diels G, Schepens W, Thuring JW, Meerpoel L, Packman K, Lorenzi MV, Laquerre S. Discovery and Pharmacological Characterization of JNJ-64619178, a Novel Small-Molecule Inhibitor of PRMT5 with Potent Antitumor Activity. Mol Cancer Ther 2021; 20:2317-2328. [PMID: 34583982 PMCID: PMC9398174 DOI: 10.1158/1535-7163.mct-21-0367] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 07/15/2021] [Accepted: 09/15/2021] [Indexed: 01/07/2023]
Abstract
The protein arginine methyltransferase 5 (PRMT5) methylates a variety of proteins involved in splicing, multiple signal transduction pathways, epigenetic control of gene expression, and mechanisms leading to protein expression required for cellular proliferation. Dysregulation of PRMT5 is associated with clinical features of several cancers, including lymphomas, lung cancer, and breast cancer. Here, we describe the characterization of JNJ-64619178, a novel, selective, and potent PRMT5 inhibitor, currently in clinical trials for patients with advanced solid tumors, non-Hodgkin's lymphoma, and lower-risk myelodysplastic syndrome. JNJ-64619178 demonstrated a prolonged inhibition of PRMT5 and potent antiproliferative activity in subsets of cancer cell lines derived from various histologies, including lung, breast, pancreatic, and hematological malignancies. In primary acute myelogenous leukemia samples, the presence of splicing factor mutations correlated with a higher ex vivo sensitivity to JNJ-64619178. Furthermore, the potent and unique mechanism of inhibition of JNJ-64619178, combined with highly optimized pharmacological properties, led to efficient tumor growth inhibition and regression in several xenograft models in vivo, with once-daily or intermittent oral-dosing schedules. An increase in splicing burden was observed upon JNJ-64619178 treatment. Overall, these observations support the continued clinical evaluation of JNJ-64619178 in patients with aberrant PRMT5 activity-driven tumors.
Collapse
Affiliation(s)
- Dirk Brehmer
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Lijs Beke
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Tongfei Wu
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | | | - Christopher Moy
- Janssen Research and Development, Spring House, Pennsylvania
| | - Weimei Sun
- Janssen Research and Development, Spring House, Pennsylvania
| | - Geert Mannens
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Vineet Pande
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - An Boeckx
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | | | - Thomas Nys
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | | | - Bie Verbist
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Longen Zhou
- Janssen Research and Development, Shanghai, China
| | - Yue Fan
- Janssen Research and Development, Shanghai, China
| | - Vipul Bhargava
- Janssen Research and Development, Spring House, Pennsylvania
| | | | - Petra Vinken
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Tinne Verhulst
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Angelique Gilbert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Sumit Rai
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | - Timothy A Graubert
- Massachusetts General Hospital Cancer Center, Harvard Medical School, Charlestown, Massachusetts
| | | | - Danilo Fiore
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Junchen Gu
- Janssen Research and Development, Spring House, Pennsylvania
| | - Amy Johnson
- Janssen Research and Development, Spring House, Pennsylvania
| | | | | | - David Walker
- Janssen Research and Development, Spring House, Pennsylvania
| | | | | | | | - Gaston Diels
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | - Wim Schepens
- Janssen Research and Development, Beerse, Antwerp, Belgium
| | | | | | - Kathryn Packman
- Janssen Research and Development, Spring House, Pennsylvania
| | | | - Sylvie Laquerre
- Janssen Research and Development, Spring House, Pennsylvania.
| |
Collapse
|
16
|
Werner T, Steidel M, Eberl HC, Bantscheff M. Affinity Enrichment Chemoproteomics for Target Deconvolution and Selectivity Profiling. Methods Mol Biol 2021; 2228:237-252. [PMID: 33950495 DOI: 10.1007/978-1-0716-1024-4_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2023]
Abstract
In order to understand the full mechanism of action of candidate drug molecules, it is critical to thoroughly characterize their interactions with endogenously expressed pharmacological targets and potentially undesired off-targets. Here we describe a chemoproteomics approach that is based on functionalized analogs of the compound of interest to affinity enrich target proteins from cell or tissue extracts. Experiments are designed as competition binding assays where free parental compound is spiked at a range of concentrations into the extracts to compete specific binders off the immobilized compound matrix. Quantification of matrix-bound proteins enables generation of dose-response curves and half-binding concentrations. In addition, the influence of the affinity matrix on the equilibrium is determined in rebinding experiments. TMT10 isobaric mass tags enable analyzing repeat binding and dose-dependent competition samples in a single mass spectrometry analysis run, thus enabling the efficient identification of targets, apparent dissociation constants, and selectivity of small molecules in a single experiment. The workflow is exemplified with the kinase inhibitor sunitinib.
Collapse
Affiliation(s)
- Thilo Werner
- Cellzome GmbH, GlaxoSmithKline, Heidelberg, Germany
| | | | | | | |
Collapse
|
17
|
Rao S, Yao Y, Bauer DE. Editing GWAS: experimental approaches to dissect and exploit disease-associated genetic variation. Genome Med 2021; 13:41. [PMID: 33691767 PMCID: PMC7948363 DOI: 10.1186/s13073-021-00857-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 02/12/2021] [Indexed: 12/17/2022] Open
Abstract
Genome-wide association studies (GWAS) have uncovered thousands of genetic variants that influence risk for human diseases and traits. Yet understanding the mechanisms by which these genetic variants, mainly noncoding, have an impact on associated diseases and traits remains a significant hurdle. In this review, we discuss emerging experimental approaches that are being applied for functional studies of causal variants and translational advances from GWAS findings to disease prevention and treatment. We highlight the use of genome editing technologies in GWAS functional studies to modify genomic sequences, with proof-of-principle examples. We discuss the challenges in interrogating causal variants, points for consideration in experimental design and interpretation of GWAS locus mechanisms, and the potential for novel therapeutic opportunities. With the accumulation of knowledge of functional genetics, therapeutic genome editing based on GWAS discoveries will become increasingly feasible.
Collapse
Affiliation(s)
- Shuquan Rao
- Division of Hematology/Oncology, Boston Children's Hospital; Department of Pediatric Oncology, Dana-Farber Cancer Institute; Harvard Stem Cell Institute; Broad Institute; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Yao Yao
- Division of Hematology/Oncology, Boston Children's Hospital; Department of Pediatric Oncology, Dana-Farber Cancer Institute; Harvard Stem Cell Institute; Broad Institute; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Daniel E Bauer
- Division of Hematology/Oncology, Boston Children's Hospital; Department of Pediatric Oncology, Dana-Farber Cancer Institute; Harvard Stem Cell Institute; Broad Institute; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
18
|
Fan X, Masamsetti VP, Sun JQ, Engholm-Keller K, Osteil P, Studdert J, Graham ME, Fossat N, Tam PP. TWIST1 and chromatin regulatory proteins interact to guide neural crest cell differentiation. eLife 2021; 10:62873. [PMID: 33554859 PMCID: PMC7968925 DOI: 10.7554/elife.62873] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Accepted: 02/05/2021] [Indexed: 12/11/2022] Open
Abstract
Protein interaction is critical molecular regulatory activity underlining cellular functions and precise cell fate choices. Using TWIST1 BioID-proximity-labeling and network propagation analyses, we discovered and characterized a TWIST-chromatin regulatory module (TWIST1-CRM) in the neural crest cells (NCC). Combinatorial perturbation of core members of TWIST1-CRM: TWIST1, CHD7, CHD8, and WHSC1 in cell models and mouse embryos revealed that loss of the function of the regulatory module resulted in abnormal differentiation of NCCs and compromised craniofacial tissue patterning. Following NCC delamination, low level of TWIST1-CRM activity is instrumental to stabilize the early NCC signatures and migratory potential by repressing the neural stem cell programs. High level of TWIST1 module activity at later phases commits the cells to the ectomesenchyme. Our study further revealed the functional interdependency of TWIST1 and potential neurocristopathy factors in NCC development. Shaping the head and face during development relies on a complex ballet of molecular signals that orchestrates the movement and specialization of various groups of cells. In animals with a backbone for example, neural crest cells (NCCs for short) can march long distances from the developing spine to become some of the tissues that form the skull and cartilage but also the pigment cells and nervous system. NCCs mature into specific cell types thanks to a complex array of factors which trigger a precise sequence of binary fate decisions at the right time and place. Amongst these factors, the protein TWIST1 can set up a cascade of genetic events that control how NCCs will ultimately form tissues in the head. To do so, the TWIST1 protein interacts with many other molecular actors, many of which are still unknown. To find some of these partners, Fan et al. studied TWIST1 in the NCCs of mice and cells grown in the lab. The experiments showed that TWIST1 interacted with CHD7, CHD8 and WHSC1, three proteins that help to switch genes on and off, and which contribute to NCCs moving across the head during development. Further work by Fan et al. then revealed that together, these molecular actors are critical for NCCs to form cells that will form facial bones and cartilage, as opposed to becoming neurons. This result helps to show that there is a trade-off between NCCs forming the face or being part of the nervous system. One in three babies born with a birth defect shows anomalies of the head and face: understanding the exact mechanisms by which NCCs contribute to these structures may help to better predict risks for parents, or to develop new approaches for treatment.
Collapse
Affiliation(s)
- Xiaochen Fan
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - V Pragathi Masamsetti
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Jane Qj Sun
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Kasper Engholm-Keller
- Synapse Proteomics Group, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Pierre Osteil
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Joshua Studdert
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Mark E Graham
- Synapse Proteomics Group, Children's Medical Research Institute, The University of Sydney, Sydney, Australia
| | - Nicolas Fossat
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| | - Patrick Pl Tam
- Embryology Unit, Children's Medical Research Institute, The University of Sydney, Sydney, Australia.,The University of Sydney, School of Medical Sciences, Faculty of Medicine and Health, Sydney, Australia
| |
Collapse
|
19
|
Bukhtiyarova M, Cook EM, Hancock PJ, Hruza AW, Shaw AW, Adam GC, Barnard RJO, McKenna PM, Holloway MK, Bell IM, Carroll S, Cornella-Taracido I, Cox CD, Kutchukian PS, Powell DA, Strickland C, Trotter BW, Tudor M, Wolkenberg S, Li J, Tellers DM. Discovery of an Anion-Dependent Farnesyltransferase Inhibitor from a Phenotypic Screen. ACS Med Chem Lett 2021; 12:99-106. [PMID: 33488970 DOI: 10.1021/acsmedchemlett.0c00551] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/15/2020] [Indexed: 12/18/2022] Open
Abstract
By employing a phenotypic screen, a set of compounds, exemplified by 1, were identified which potentiate the ability of histone deacetylase inhibitor vorinostat to reverse HIV latency. Proteome enrichment followed by quantitative mass spectrometric analysis employing a modified analogue of 1 as affinity bait identified farnesyl transferase (FTase) as the primary interacting protein in cell lysates. This ligand-FTase binding interaction was confirmed via X-ray crystallography and temperature dependent fluorescence studies, despite 1 lacking structural and binding similarity to known FTase inhibitors. Although multiple lines of evidence established the binding interaction, these ligands exhibited minimal inhibitory activity in a cell-free biochemical FTase inhibition assay. Subsequent modification of the biochemical assay by increasing anion concentration demonstrated FTase inhibitory activity in this novel class. We propose 1 binds together with the anion in the active site to inhibit farnesyl transferase. Implications for phenotypic screening deconvolution and HIV reactivation are discussed.
Collapse
Affiliation(s)
| | - Erica M. Cook
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Paula J. Hancock
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Alan W. Hruza
- MRL, Merck & Co., Inc., Kenilworth, New Jersey, 07033, United States
| | - Anthony W. Shaw
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Gregory C. Adam
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Philip M. McKenna
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | - Ian M. Bell
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Steve Carroll
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | | | | | | | - David A. Powell
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Corey Strickland
- MRL, Merck & Co., Inc., Kenilworth, New Jersey, 07033, United States
| | | | - Matthew Tudor
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Scott Wolkenberg
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jing Li
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - David M. Tellers
- MRL, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| |
Collapse
|
20
|
Watson NA, Cartwright TN, Lawless C, Cámara-Donoso M, Sen O, Sako K, Hirota T, Kimura H, Higgins JMG. Kinase inhibition profiles as a tool to identify kinases for specific phosphorylation sites. Nat Commun 2020; 11:1684. [PMID: 32245944 PMCID: PMC7125195 DOI: 10.1038/s41467-020-15428-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/06/2020] [Indexed: 01/08/2023] Open
Abstract
There are thousands of known cellular phosphorylation sites, but the paucity of ways to identify kinases for particular phosphorylation events remains a major roadblock for understanding kinase signaling. To address this, we here develop a generally applicable method that exploits the large number of kinase inhibitors that have been profiled on near-kinome-wide panels of protein kinases. The inhibition profile for each kinase provides a fingerprint that allows identification of unknown kinases acting on target phosphosites in cell extracts. We validate the method on diverse known kinase-phosphosite pairs, including histone kinases, EGFR autophosphorylation, and Integrin β1 phosphorylation by Src-family kinases. We also use our approach to identify the previously unknown kinases responsible for phosphorylation of INCENP at a site within a commonly phosphorylated motif in mitosis (a non-canonical target of Cyclin B-Cdk1), and of BCL9L at S915 (PKA). We show that the method has clear advantages over in silico and genetic screening.
Collapse
Affiliation(s)
- Nikolaus A Watson
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Tyrell N Cartwright
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Conor Lawless
- Wellcome Centre for Mitochondrial Research, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Marcos Cámara-Donoso
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Onur Sen
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Kosuke Sako
- The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo, 135-8550, Japan
| | - Toru Hirota
- The Cancer Institute, Japanese Foundation for Cancer Research, Koto, Tokyo, 135-8550, Japan
| | - Hiroshi Kimura
- Cell Biology Center, Institute of Innovative Research, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8503, Japan
| | - Jonathan M G Higgins
- Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
21
|
Palve V, Liao Y, Remsing Rix LL, Rix U. Turning liabilities into opportunities: Off-target based drug repurposing in cancer. Semin Cancer Biol 2020; 68:209-229. [PMID: 32044472 DOI: 10.1016/j.semcancer.2020.02.003] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 12/12/2022]
Abstract
Targeted drugs and precision medicine have transformed the landscape of cancer therapy and significantly improved patient outcomes in many cases. However, as therapies are becoming more and more tailored to smaller patient populations and acquired resistance is limiting the duration of clinical responses, there is an ever increasing demand for new drugs, which is not easily met considering steadily rising drug attrition rates and development costs. Considering these challenges drug repurposing is an attractive complementary approach to traditional drug discovery that can satisfy some of these needs. This is facilitated by the fact that most targeted drugs, despite their implicit connotation, are not singularly specific, but rather display a wide spectrum of target selectivity. Importantly, some of the unintended drug "off-targets" are known anticancer targets in their own right. Others are becoming recognized as such in the process of elucidating off-target mechanisms that in fact are responsible for a drug's anticancer activity, thereby revealing potentially new cancer vulnerabilities. Harnessing such beneficial off-target effects can therefore lead to novel and promising precision medicine approaches. Here, we will discuss experimental and computational methods that are employed to specifically develop single target and network-based off-target repurposing strategies, for instance with drug combinations or polypharmacology drugs. By illustrating concrete examples that have led to clinical translation we will furthermore examine the various scientific and non-scientific factors that cumulatively determine the success of these efforts and thus can inform the future development of new and potentially lifesaving off-target based drug repurposing strategies for cancers that constitute important unmet medical needs.
Collapse
Affiliation(s)
- Vinayak Palve
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Yi Liao
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Lily L Remsing Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA
| | - Uwe Rix
- Department of Drug Discovery, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL, 33612, USA.
| |
Collapse
|
22
|
Gräwe C, Makowski MM, Vermeulen M. PAQMAN: Protein-nucleic acid affinity quantification by MAss spectrometry in nuclear extracts. Methods 2019; 184:70-77. [PMID: 31857188 DOI: 10.1016/j.ymeth.2019.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 12/10/2019] [Accepted: 12/11/2019] [Indexed: 12/01/2022] Open
Abstract
In recent years, various mass spectrometry-based approaches have been developed to determine global protein-DNA binding specificities using DNA affinity purifications from crude nuclear extracts. However, these assays are semi-quantitative and do not provide information about interaction affinities. We recently developed a technology that we call Protein-nucleic acid Affinity Quantification by MAss spectrometry in Nuclear extracts or PAQMAN, that can be used to determine apparent affinities between multiple nuclear proteins and a nucleic acid sequence of interest in one experiment. In PAQMAN, a series of affinity purifications with increasing bait concentrations and fixed amounts of crude nuclear extracts are combined with isobaric stable isotope labeling and quantitative mass spectrometry to generate Hill-like Kd curves for dozens of proteins in a single experiment. Here, we apply PAQMAN to determine apparent affinities for a genetic variant, rs36115365-C, which regulates TERT expression and is associated with an increased risk to develop various malignancies. Furthermore, we describe a detailed protocol for this method including important quality checks.
Collapse
Affiliation(s)
- Cathrin Gräwe
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, The Netherlands
| | - Matthew M Makowski
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, The Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 GA Nijmegen, The Netherlands.
| |
Collapse
|
23
|
Reinecke M, Heinzlmeir S, Wilhelm M, Médard G, Klaeger S, Kuster B. Kinobeads: A Chemical Proteomic Approach for Kinase Inhibitor Selectivity Profiling and Target Discovery. ACTA ACUST UNITED AC 2019. [DOI: 10.1002/9783527818242.ch4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
|
24
|
Epigenetic drug target deconvolution by mass spectrometry-based technologies. Nat Struct Mol Biol 2019; 26:854-857. [PMID: 31582842 DOI: 10.1038/s41594-019-0279-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 07/15/2019] [Indexed: 12/12/2022]
Abstract
The identification of the full target spectrum of active molecules, known as target deconvolution, has become an indispensable step during the drug discovery process. It is now achievable thanks to mass spectrometry-based technologies. Here we discuss these approaches in the context of epigenetic drug discovery.
Collapse
|
25
|
Eberl HC, Werner T, Reinhard FB, Lehmann S, Thomson D, Chen P, Zhang C, Rau C, Muelbaier M, Drewes G, Drewry D, Bantscheff M. Chemical proteomics reveals target selectivity of clinical Jak inhibitors in human primary cells. Sci Rep 2019; 9:14159. [PMID: 31578349 PMCID: PMC6775116 DOI: 10.1038/s41598-019-50335-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/05/2019] [Indexed: 12/15/2022] Open
Abstract
Kinobeads are a set of promiscuous kinase inhibitors immobilized on sepharose beads for the comprehensive enrichment of endogenously expressed protein kinases from cell lines and tissues. These beads enable chemoproteomics profiling of kinase inhibitors of interest in dose-dependent competition studies in combination with quantitative mass spectrometry. We present improved bead matrices that capture more than 350 protein kinases and 15 lipid kinases from human cell lysates, respectively. A multiplexing strategy is suggested that enables determination of apparent dissociation constants in a single mass spectrometry experiment. Miniaturization of the procedure enabled determining the target selectivity of the clinical BCR-ABL inhibitor dasatinib in peripheral blood mononuclear cell (PBMC) lysates from individual donors. Profiling of a set of Jak kinase inhibitors revealed kinase off-targets from nearly all kinase families underpinning the need to profile kinase inhibitors against the kinome. Potently bound off-targets of clinical inhibitors suggest polypharmacology, e.g. through MRCK alpha and beta, which bind to decernotinib with nanomolar affinity.
Collapse
Affiliation(s)
- H Christian Eberl
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| | - Thilo Werner
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Friedrich B Reinhard
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Stephanie Lehmann
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Douglas Thomson
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Peiling Chen
- GlaxoSmithKline, Upper Merion, 709 Swedeland Rd #1539, King of Prussia, PA, 19406, United States
| | - Cunyu Zhang
- GlaxoSmithKline, Upper Merion, 709 Swedeland Rd #1539, King of Prussia, PA, 19406, United States
| | - Christina Rau
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Marcel Muelbaier
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - Gerard Drewes
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany
| | - David Drewry
- GlaxoSmithKline, Research Triangle Park, 5 Moore Drive, North Carolina, 27709, United States.,UNC Eshelman School of Pharmacy, Structural Genomics Consortium, University of North Carolina at Chapel Hill, 120 Mason Farm Rd, Chapel Hill, NC, 27599, United States
| | - Marcus Bantscheff
- Cellzome GmbH, A GlaxoSmithKline Company, Meyerhofstraße 1, 69117, Heidelberg, Germany.
| |
Collapse
|
26
|
Oncogenic Mutations Rewire Signaling Pathways by Switching Protein Recruitment to Phosphotyrosine Sites. Cell 2019; 179:543-560.e26. [DOI: 10.1016/j.cell.2019.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/20/2019] [Accepted: 09/05/2019] [Indexed: 12/22/2022]
|
27
|
Rabalski AJ, Bogdan AR, Baranczak A. Evaluation of Chemically-Cleavable Linkers for Quantitative Mapping of Small Molecule-Cysteinome Reactivity. ACS Chem Biol 2019; 14:1940-1950. [PMID: 31430117 DOI: 10.1021/acschembio.9b00424] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Numerous reagents have been developed to enable chemical proteomic analysis of small molecule-protein interactomes. However, the performance of these reagents has not been systematically evaluated and compared. Herein, we report our efforts to conduct a parallel assessment of two widely used chemically cleavable linkers equipped with dialkoxydiphenylsilane (DADPS linker) and azobenzene (AZO linker) moieties. Profiling a cellular cysteinome using the iodoacetamide alkyne probe demonstrated a significant discrepancy between the experimental results obtained through the application of each of the reagents. To better understand the source of observed discrepancy, we evaluated the key sample preparation steps. We also performed a mass tolerant database search strategy using MSFragger software. This resulted in identifying a previously unreported artifactual modification on the residual mass of the azobenzene linker. Furthermore, we conducted a comparative analysis of enrichment modes using both cleavable linkers. This effort determined that enrichment of proteolytic digests yielded a far greater number of identified cysteine residues than the enrichment conducted prior to protein digest. Inspired by recent studies where multiplexed quantitative labeling strategies were applied to cleavable biotin linkers, we combined this further optimized protocol using the DADPS cleavable linker with tandem mass tag (TMT) labeling to profile the FDA-approved covalent EGFR kinase inhibitor dacomitinib against the cysteinome of an epidermoid cancer cell line. Our analysis resulted in the detection and quantification of over 10,000 unique cysteine residues, a nearly 3-fold increase over previous studies that used cleavable biotin linkers for enrichment. Critically, cysteine residues corresponding to proteins directly as well as indirectly modulated by dacomitinib treatment were identified. Overall, our study suggests that the dialkoxydiphenylsilane linker could be broadly applied wherever chemically cleavable linkers are required for chemical proteomic characterization of cellular proteomes.
Collapse
Affiliation(s)
- Adam J. Rabalski
- Drug Discovery Science & Technology, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064-6101, United States
| | - Andrew R. Bogdan
- Drug Discovery Science & Technology, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064-6101, United States
| | - Aleksandra Baranczak
- Drug Discovery Science & Technology, AbbVie Inc., 1 North Waukegan Road, North Chicago, Illinois 60064-6101, United States
| |
Collapse
|
28
|
Mayerthaler F, Finley MF, Pfeifer TA, Antolin AA. Meeting Proceedings from ICBS 2018- Toward Translational Impact. ACS Chem Biol 2019; 14:567-578. [PMID: 30860357 DOI: 10.1021/acschembio.9b00169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Affiliation(s)
- Florian Mayerthaler
- Institute of Biochemistry, Department of Chemistry and Pharmacy, University of Münster, Münster, Germany
| | - Michael F. Finley
- Janssen Research & Development, Spring House, Pennsylvania 19477, United States
| | - Tom A. Pfeifer
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, Canada V6T 1Z3
| | - Albert A. Antolin
- The Department of Data Science, The Institute of Cancer Research, London, SM2 5NG, United Kingdom
- The Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, United Kingdom
| |
Collapse
|
29
|
Needham EJ, Parker BL, Burykin T, James DE, Humphrey SJ. Illuminating the dark phosphoproteome. Sci Signal 2019; 12:12/565/eaau8645. [PMID: 30670635 DOI: 10.1126/scisignal.aau8645] [Citation(s) in RCA: 210] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Protein phosphorylation is a major regulator of protein function and biological outcomes. This was first recognized through functional biochemical experiments, and in the past decade, major technological advances in mass spectrometry have enabled the study of protein phosphorylation on a global scale. This rapidly growing field of phosphoproteomics has revealed that more than 100,000 distinct phosphorylation events occur in human cells, which likely affect the function of every protein. Phosphoproteomics has improved the understanding of the function of even the most well-characterized protein kinases by revealing new downstream substrates and biology. However, current biochemical and bioinformatic approaches have only identified kinases for less than 5% of the phosphoproteome, and functional assignments of phosphosites are almost negligible. Notably, our understanding of the relationship between kinases and their substrates follows a power law distribution, with almost 90% of phosphorylation sites currently assigned to the top 20% of kinases. In addition, more than 150 kinases do not have a single known substrate. Despite a small group of kinases dominating biomedical research, the number of substrates assigned to a kinase does not correlate with disease relevance as determined by pathogenic human mutation prevalence and mouse model phenotypes. Improving our understanding of the substrates targeted by all kinases and functionally annotating the phosphoproteome will be broadly beneficial. Advances in phosphoproteomics technologies, combined with functional screening approaches, should make it feasible to illuminate the connectivity and functionality of the entire phosphoproteome, providing enormous opportunities for discovering new biology, therapeutic targets, and possibly diagnostics.
Collapse
Affiliation(s)
- Elise J Needham
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Benjamin L Parker
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia.,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - Timur Burykin
- Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| | - David E James
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia. .,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia.,Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Sean J Humphrey
- School of Life and Environmental Sciences, University of Sydney, Sydney, NSW 2006, Australia. .,Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
30
|
Szabadkai I, Torka R, Garamvölgyi R, Baska F, Gyulavári P, Boros S, Illyés E, Choidas A, Ullrich A, Őrfi L. Discovery of N-[4-(Quinolin-4-yloxy)phenyl]benzenesulfonamides as Novel AXL Kinase Inhibitors. J Med Chem 2018; 61:6277-6292. [PMID: 29928803 DOI: 10.1021/acs.jmedchem.8b00672] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The overexpression of AXL kinase has been described in many types of cancer. Due to its role in proliferation, survival, migration, and resistance, AXL represents a promising target in the treatment of the disease. In this study we present a novel compound family that successfully targets the AXL kinase. Through optimization and detailed SAR studies we developed low nanomolar inhibitors, and after further biological characterization we identified a potent AXL kinase inhibitor with favorable pharmacokinetic profile. The antitumor activity was determined in xenograft models, and the lead compounds reduced the tumor size by 40% with no observed toxicity as well as lung metastasis formation by 66% when compared to vehicle control.
Collapse
Affiliation(s)
| | - Robert Torka
- Institute of Physiological Chemistry , University Halle-Wittenberg , Halle (Saale) 06108 , Germany
| | - Rita Garamvölgyi
- Vichem Chemie Research Ltd. , Budapest 1022 , Hungary
- Department of Pharmaceutical Chemistry , Semmelweis University , Budapest 1092 , Hungary
| | - Ferenc Baska
- Vichem Chemie Research Ltd. , Budapest 1022 , Hungary
| | - Pál Gyulavári
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry , Semmelweis University , Budapest 1094 , Hungary
| | - Sándor Boros
- Vichem Chemie Research Ltd. , Budapest 1022 , Hungary
| | - Eszter Illyés
- Vichem Chemie Research Ltd. , Budapest 1022 , Hungary
| | - Axel Choidas
- Lead Discovery Center GmbH , Dortmund 44227 , Germany
| | - Axel Ullrich
- Department of Molecular Biology , Max Planck Institute of Biochemistry , Martinsried 82152 , Germany
| | - László Őrfi
- Vichem Chemie Research Ltd. , Budapest 1022 , Hungary
- Department of Pharmaceutical Chemistry , Semmelweis University , Budapest 1092 , Hungary
| |
Collapse
|
31
|
Campe R, Hollenbach E, Kämmerer L, Hendriks J, Höffken HW, Kraus H, Lerchl J, Mietzner T, Tresch S, Witschel M, Hutzler J. A new herbicidal site of action: Cinmethylin binds to acyl-ACP thioesterase and inhibits plant fatty acid biosynthesis. PESTICIDE BIOCHEMISTRY AND PHYSIOLOGY 2018; 148:116-125. [PMID: 29891362 DOI: 10.1016/j.pestbp.2018.04.006] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 05/16/2023]
Abstract
The prevalent occurrence of herbicide resistant weeds increases the necessity for new site of action herbicides for effective control as well as to relax selection pressure on the known sites of action. As a consequence, interest increased in the unexploited molecule cinmethylin as a new solution for the control of weedy grasses in cereals. Therefore, the mechanism of action of cinmethylin was reevaluated. We applied the chemoproteomic approach cellular Target Profiling™ from Evotec to identify the cinmethylin target in Lemna paucicostata protein extracts. We found three potential targets belonging to the same protein family of fatty acid thioesterases (FAT) to bind to cinmethylin with high affinity. Binding of cinmethylin to FAT proteins from Lemna and Arabidopsis was confirmed by fluorescence-based thermal shift assay. The plastid localized enzyme FAT plays a crucial role in plant lipid biosynthesis, by mediating the release of fatty acids (FA) from its acyl carrier protein (ACP) which is necessary for FA export to the endoplasmic reticulum. GC-MS analysis of free FA composition in Lemna extracts revealed strong reduction of unsaturated C18 as well as saturated C14, and C16 FAs upon treatment with cinmethylin, indicating that FA release for subsequent lipid biosynthesis is the primary target of cinmethylin. Lipid biosynthesis is a prominent target of different herbicide classes. To assess whether FAT inhibition constitutes a new mechanism of action within this complex pathway, we compared physiological effects of cinmethylin to different ACCase and VLCFA synthesis inhibitors and identified characteristic differences in plant symptomology and free FA composition upon treatment with the three herbicide classes. Also, principal component analysis of total metabolic profiling of treated Lemna plants showed strong differences in overall metabolic changes after cinmethylin, ACCase or VLCFA inhibitor treatments. Our results identified and confirmed FAT as the cinmethylin target and validate FAT inhibition as a new site of action different from other lipid biosynthesis inhibitor classes.
Collapse
Affiliation(s)
- Ruth Campe
- BASF SE, Speyerer Straße 2, D-67117 Limburgerhof, Germany.
| | - Eva Hollenbach
- BASF SE, Speyerer Straße 2, D-67117 Limburgerhof, Germany
| | - Lara Kämmerer
- BASF SE, Speyerer Straße 2, D-67117 Limburgerhof, Germany
| | | | | | - Helmut Kraus
- BASF Corporation, 26 Davis Drive, Research Triangle Park, 27709-3528 NC, United States
| | - Jens Lerchl
- BASF SE, Speyerer Straße 2, D-67117 Limburgerhof, Germany
| | - Thomas Mietzner
- BASF SE, Carl Bosch Straße 38, D-67056 Ludwigshafen, Germany
| | - Stefan Tresch
- BASF SE, Carl Bosch Straße 38, D-67056 Ludwigshafen, Germany
| | | | | |
Collapse
|
32
|
Makowski MM, Gräwe C, Foster BM, Nguyen NV, Bartke T, Vermeulen M. Global profiling of protein-DNA and protein-nucleosome binding affinities using quantitative mass spectrometry. Nat Commun 2018; 9:1653. [PMID: 29695722 PMCID: PMC5916898 DOI: 10.1038/s41467-018-04084-0] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Accepted: 04/03/2018] [Indexed: 01/02/2023] Open
Abstract
Interaction proteomics studies have provided fundamental insights into multimeric biomolecular assemblies and cell-scale molecular networks. Significant recent developments in mass spectrometry-based interaction proteomics have been fueled by rapid advances in label-free, isotopic, and isobaric quantitation workflows. Here, we report a quantitative protein–DNA and protein–nucleosome binding assay that uses affinity purifications from nuclear extracts coupled with isobaric chemical labeling and mass spectrometry to quantify apparent binding affinities proteome-wide. We use this assay with a variety of DNA and nucleosome baits to quantify apparent binding affinities of monomeric and multimeric transcription factors and chromatin remodeling complexes. Quantitative mass spectrometry enables the proteome-wide assessment of biomolecular binding affinities. While previous approaches mainly focused on protein–small molecule interactions, the authors here present a method to probe protein–DNA and protein–nucleosome binding affinities at proteome scale.
Collapse
Affiliation(s)
- Matthew M Makowski
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, 6500 HB, The Netherlands.,Oncode Institute, Radboud University, Nijmegen, 6500 HB, The Netherlands
| | - Cathrin Gräwe
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, 6500 HB, The Netherlands.,Oncode Institute, Radboud University, Nijmegen, 6500 HB, The Netherlands
| | - Benjamin M Foster
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764, Neuherberg, Germany.,MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Nhuong V Nguyen
- MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK.,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK
| | - Till Bartke
- Institute of Functional Epigenetics, Helmholtz Zentrum München, 85764, Neuherberg, Germany. .,MRC London Institute of Medical Sciences (LMS), London, W12 0NN, UK. .,Institute of Clinical Sciences (ICS), Faculty of Medicine, Imperial College London, London, W12 0NN, UK.
| | - Michiel Vermeulen
- Department of Molecular Biology, Radboud Institute for Molecular Life Sciences, Radboud University, Nijmegen, 6500 HB, The Netherlands. .,Oncode Institute, Radboud University, Nijmegen, 6500 HB, The Netherlands.
| |
Collapse
|
33
|
Klaeger S, Heinzlmeir S, Wilhelm M, Polzer H, Vick B, Koenig PA, Reinecke M, Ruprecht B, Petzoldt S, Meng C, Zecha J, Reiter K, Qiao H, Helm D, Koch H, Schoof M, Canevari G, Casale E, Depaolini SR, Feuchtinger A, Wu Z, Schmidt T, Rueckert L, Becker W, Huenges J, Garz AK, Gohlke BO, Zolg DP, Kayser G, Vooder T, Preissner R, Hahne H, Tõnisson N, Kramer K, Götze K, Bassermann F, Schlegl J, Ehrlich HC, Aiche S, Walch A, Greif PA, Schneider S, Felder ER, Ruland J, Médard G, Jeremias I, Spiekermann K, Kuster B. The target landscape of clinical kinase drugs. Science 2018; 358:358/6367/eaan4368. [PMID: 29191878 DOI: 10.1126/science.aan4368] [Citation(s) in RCA: 574] [Impact Index Per Article: 82.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Accepted: 09/20/2017] [Indexed: 02/06/2023]
Abstract
Kinase inhibitors are important cancer therapeutics. Polypharmacology is commonly observed, requiring thorough target deconvolution to understand drug mechanism of action. Using chemical proteomics, we analyzed the target spectrum of 243 clinically evaluated kinase drugs. The data revealed previously unknown targets for established drugs, offered a perspective on the "druggable" kinome, highlighted (non)kinase off-targets, and suggested potential therapeutic applications. Integration of phosphoproteomic data refined drug-affected pathways, identified response markers, and strengthened rationale for combination treatments. We exemplify translational value by discovering SIK2 (salt-inducible kinase 2) inhibitors that modulate cytokine production in primary cells, by identifying drugs against the lung cancer survival marker MELK (maternal embryonic leucine zipper kinase), and by repurposing cabozantinib to treat FLT3-ITD-positive acute myeloid leukemia. This resource, available via the ProteomicsDB database, should facilitate basic, clinical, and drug discovery research and aid clinical decision-making.
Collapse
Affiliation(s)
- Susan Klaeger
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephanie Heinzlmeir
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Mathias Wilhelm
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Harald Polzer
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Binje Vick
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, German Center for Environmental Health (HMGU), Munich, Germany
| | | | - Maria Reinecke
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Benjamin Ruprecht
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Svenja Petzoldt
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chen Meng
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Jana Zecha
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Katrin Reiter
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Huichao Qiao
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Dominic Helm
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Heiner Koch
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Melanie Schoof
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | | | - Elena Casale
- Oncology, Nerviano Medical Sciences Srl, Milan, Italy
| | | | - Annette Feuchtinger
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Zhixiang Wu
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Tobias Schmidt
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | | | | | | | - Anne-Kathrin Garz
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine III, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Bjoern-Oliver Gohlke
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Structural Bioinformatics Group, Charité-Universitätsmedizin, Berlin, Germany
| | - Daniel Paul Zolg
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Gian Kayser
- Institute of Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Tonu Vooder
- Center of Thoracic Surgery, Krefeld, Germany.,Estonian Genome Center, University of Tartu, Tartu, Estonia.,Tartu University Hospital, Tartu, Estonia
| | - Robert Preissner
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Structural Bioinformatics Group, Charité-Universitätsmedizin, Berlin, Germany
| | - Hannes Hahne
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Neeme Tõnisson
- Estonian Genome Center, University of Tartu, Tartu, Estonia.,Tartu University Hospital, Tartu, Estonia
| | - Karl Kramer
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Katharina Götze
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine III, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Florian Bassermann
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Medicine III, Klinikum rechts der Isar, TUM, Munich, Germany
| | | | | | | | - Axel Walch
- Research Unit Analytical Pathology, Helmholtz Zentrum München, Neuherberg, Germany
| | - Philipp A Greif
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Sabine Schneider
- Department of Chemistry, TUM, Garching, Germany.,Center For Integrated Protein Science Munich (CIPSM), Munich, Germany
| | | | - Juergen Ruland
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Institut für Klinische Chemie und Pathobiochemie, TUM, Munich, Germany
| | - Guillaume Médard
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany
| | - Irmela Jeremias
- German Cancer Consortium (DKTK), Heidelberg, Germany.,Department of Apoptosis in Hematopoietic Stem Cells, Helmholtz Center Munich, German Center for Environmental Health (HMGU), Munich, Germany.,Department of Pediatrics, Dr von Hauner Children's Hospital, LMU, Munich, Germany
| | - Karsten Spiekermann
- German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Internal Medicine III, University Hospital, Ludwig-Maximilians-Universität (LMU) München, Munich, Germany
| | - Bernhard Kuster
- Chair of Proteomics and Bioanalytics, Technical University of Munich (TUM), Freising, Germany. .,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany.,Center For Integrated Protein Science Munich (CIPSM), Munich, Germany.,Bavarian Biomolecular Mass Spectrometry Center (BayBioMS), TUM, Freising, Germany
| |
Collapse
|
34
|
Roolf C, Dybowski N, Sekora A, Mueller S, Knuebel G, Tebbe A, Murua Escobar H, Godl K, Junghanss C, Schaab C. Phosphoproteome Analysis Reveals Differential Mode of Action of Sorafenib in Wildtype and Mutated FLT3 Acute Myeloid Leukemia (AML) Cells. Mol Cell Proteomics 2017; 16:1365-1376. [PMID: 28450419 PMCID: PMC5500767 DOI: 10.1074/mcp.m117.067462] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/30/2017] [Indexed: 01/07/2023] Open
Abstract
Constitutively activating internal tandem duplication (ITD) alterations of the receptor tyrosine kinase FLT3 (Fms-like tyrosine kinase 3) are common in acute myeloid leukemia (AML) and classifies FLT3 as an attractive therapeutic target. So far, applications of FLT3 small molecule inhibitors have been investigated primarily in FLT3-ITD+ patients. Only recently, a prolonged event-free survival has been observed in AML patients who were treated with the multikinase inhibitor sorafenib in addition to standard therapy. Here, we studied the sorafenib effect on proliferation in a panel of 13 FLT3-ITD− and FLT3-ITD+ AML cell lines. Sorafenib IC50 values ranged from 0.001 to 5.6 μm, whereas FLT3-ITD+ cells (MOLM-13, MV4-11) were found to be more sensitive to sorafenib than FLT3-ITD− cells. However, we identified two FLT3-ITD− cell lines (MONO-MAC-1 and OCI-AML-2) which were also sorafenib sensitive. Phosphoproteome analyses revealed that the affected pathways differed in sorafenib sensitive FLT3-ITD− and FLT3-ITD+ cells. In MV4-11 cells sorafenib suppressed mTOR signaling by direct inhibition of FLT3. In MONO-MAC-1 cells sorafenib inhibited the MEK/ERK pathway. These data suggest that the FLT3 status in AML patients might not be the only factor predicting response to treatment with sorafenib.
Collapse
Affiliation(s)
- Catrin Roolf
- From the ‡Department of Medicine, Clinic III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, University of Rostock, 18057 Rostock, Germany
| | | | - Anett Sekora
- From the ‡Department of Medicine, Clinic III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, University of Rostock, 18057 Rostock, Germany
| | - Stefan Mueller
- From the ‡Department of Medicine, Clinic III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, University of Rostock, 18057 Rostock, Germany
| | - Gudrun Knuebel
- From the ‡Department of Medicine, Clinic III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, University of Rostock, 18057 Rostock, Germany
| | | | - Hugo Murua Escobar
- From the ‡Department of Medicine, Clinic III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, University of Rostock, 18057 Rostock, Germany
| | - Klaus Godl
- §Evotec (München) GmbH, 82152 Martinsried, Germany
| | - Christian Junghanss
- From the ‡Department of Medicine, Clinic III-Hematology/Oncology/Palliative Care, Rostock University Medical Center, University of Rostock, 18057 Rostock, Germany
| | - Christoph Schaab
- §Evotec (München) GmbH, 82152 Martinsried, Germany; .,¶Department of Proteomics and Signal Transduction, Max-Planck Institute for Biochemistry, 82152 Martinsried, Germany
| |
Collapse
|
35
|
Golkowski M, Vidadala RSR, Lombard CK, Suh HW, Maly DJ, Ong SE. Kinobead and Single-Shot LC-MS Profiling Identifies Selective PKD Inhibitors. J Proteome Res 2017; 16:1216-1227. [PMID: 28102076 DOI: 10.1021/acs.jproteome.6b00817] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
ATP-competitive protein kinase inhibitors are important research tools and therapeutic agents. Because there are >500 human kinases that contain highly conserved active sites, the development of selective inhibitors is extremely challenging. Methods to rapidly and efficiently profile kinase inhibitor targets in cell lysates are urgently needed to discover selective compounds and to elucidate the mechanisms of action for polypharmacological inhibitors. Here, we describe a protocol for microgram-scale chemoproteomic profiling of ATP-competitive kinase inhibitors using kinobeads. We employed a gel-free in situ digestion protocol coupled to nanoflow liquid chromatography-mass spectrometry to profile ∼200 kinases in single analytical runs using as little as 5 μL of kinobeads and 300 μg of protein. With our kinobead reagents, we obtained broad coverage of the kinome, monitoring the relative expression levels of 312 kinases in a diverse panel of 11 cancer cell lines. Further, we profiled a set of pyrrolopyrimidine- and pyrazolopyrimidine-based kinase inhibitors in competition-binding experiments with label-free quantification, leading to the discovery of a novel selective and potent inhibitor of protein kinase D (PKD) 1, 2, and 3. Our protocol is useful for rapid and sensitive profiling of kinase expression levels and ATP-competitive kinase inhibitor selectivity in native proteomes.
Collapse
Affiliation(s)
- Martin Golkowski
- Department of Pharmacology, School of Medicine and Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | - Rama Subba Rao Vidadala
- Department of Pharmacology, School of Medicine and Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | - Chloe K Lombard
- Department of Pharmacology, School of Medicine and Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | - Hyong Won Suh
- Department of Pharmacology, School of Medicine and Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | - Dustin J Maly
- Department of Pharmacology, School of Medicine and Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| | - Shao-En Ong
- Department of Pharmacology, School of Medicine and Department of Chemistry, University of Washington , Seattle, Washington 98195, United States
| |
Collapse
|
36
|
Cheeseman M, Chessum NEA, Rye CS, Pasqua AE, Tucker M, Wilding B, Evans LE, Lepri S, Richards M, Sharp SY, Ali S, Rowlands M, O’Fee L, Miah A, Hayes A, Henley AT, Powers M, te Poele R, De Billy E, Pellegrino L, Raynaud F, Burke R, van Montfort RLM, Eccles SA, Workman P, Jones K. Discovery of a Chemical Probe Bisamide (CCT251236): An Orally Bioavailable Efficacious Pirin Ligand from a Heat Shock Transcription Factor 1 (HSF1) Phenotypic Screen. J Med Chem 2017; 60:180-201. [PMID: 28004573 PMCID: PMC6014687 DOI: 10.1021/acs.jmedchem.6b01055] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2016] [Indexed: 12/20/2022]
Abstract
Phenotypic screens, which focus on measuring and quantifying discrete cellular changes rather than affinity for individual recombinant proteins, have recently attracted renewed interest as an efficient strategy for drug discovery. In this article, we describe the discovery of a new chemical probe, bisamide (CCT251236), identified using an unbiased phenotypic screen to detect inhibitors of the HSF1 stress pathway. The chemical probe is orally bioavailable and displays efficacy in a human ovarian carcinoma xenograft model. By developing cell-based SAR and using chemical proteomics, we identified pirin as a high affinity molecular target, which was confirmed by SPR and crystallography.
Collapse
Affiliation(s)
- Matthew
D. Cheeseman
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Nicola E. A. Chessum
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Carl S. Rye
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - A. Elisa Pasqua
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Michael
J. Tucker
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Birgit Wilding
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Lindsay E. Evans
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Susan Lepri
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Meirion Richards
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Swee Y. Sharp
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Salyha Ali
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
- Division
of Structural Biology at The Institute of
Cancer Research, London SW7 3RP, United Kingdom
| | - Martin Rowlands
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Lisa O’Fee
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Asadh Miah
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Angela Hayes
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Alan T. Henley
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Marissa Powers
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Robert te Poele
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Emmanuel De Billy
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Loredana Pellegrino
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Florence Raynaud
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Rosemary Burke
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Rob L. M. van Montfort
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
- Division
of Structural Biology at The Institute of
Cancer Research, London SW7 3RP, United Kingdom
| | - Suzanne A. Eccles
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Paul Workman
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| | - Keith Jones
- Cancer
Research UK Cancer Therapeutics Unit at The Institute of Cancer Research, London SW7 3RP, United Kingdom
| |
Collapse
|
37
|
Heinzlmeir S, Kudlinzki D, Sreeramulu S, Klaeger S, Gande SL, Linhard V, Wilhelm M, Qiao H, Helm D, Ruprecht B, Saxena K, Médard G, Schwalbe H, Kuster B. Chemical Proteomics and Structural Biology Define EPHA2 Inhibition by Clinical Kinase Drugs. ACS Chem Biol 2016; 11:3400-3411. [PMID: 27768280 DOI: 10.1021/acschembio.6b00709] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The receptor tyrosine kinase EPHA2 (Ephrin type-A receptor 2) plays important roles in oncogenesis, metastasis, and treatment resistance, yet therapeutic targeting, drug discovery, or investigation of EPHA2 biology is hampered by the lack of appropriate inhibitors and structural information. Here, we used chemical proteomics to survey 235 clinical kinase inhibitors for their kinase selectivity and identified 24 drugs with submicromolar affinities for EPHA2. NMR-based conformational dynamics together with nine new cocrystal structures delineated drug-EPHA2 interactions in full detail. The combination of selectivity profiling, structure determination, and kinome wide sequence alignment allowed the development of a classification system in which amino acids in the drug binding site of EPHA2 are categorized into key, scaffold, potency, and selectivity residues. This scheme should be generally applicable in kinase drug discovery, and we anticipate that the provided information will greatly facilitate the development of selective EPHA2 inhibitors in particular and the repurposing of clinical kinase inhibitors in general.
Collapse
Affiliation(s)
- Stephanie Heinzlmeir
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Denis Kudlinzki
- Center
for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University, 60438 Frankfurt, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sridhar Sreeramulu
- Center
for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University, 60438 Frankfurt, Germany
| | - Susan Klaeger
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Santosh Lakshmi Gande
- Center
for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University, 60438 Frankfurt, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Verena Linhard
- Center
for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University, 60438 Frankfurt, Germany
| | - Mathias Wilhelm
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Huichao Qiao
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Dominic Helm
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Benjamin Ruprecht
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Krishna Saxena
- Center
for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University, 60438 Frankfurt, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Guillaume Médard
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
| | - Harald Schwalbe
- Center
for Biomolecular Magnetic Resonance, Johann Wolfgang Goethe University, 60438 Frankfurt, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Bernhard Kuster
- Chair
of Proteomics and Bioanalytics, Technical University of Munich, 85354 Freising, Germany
- German Cancer Consortium (DKTK), 69120 Heidelberg, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Integrated Protein Science Munich (CIPSM), 85354 Freising, Germany
- Bavarian
Biomolecular Mass Spectrometry Center (BayBioMS), Technical University of Munich, 85354 Freising, Germany
| |
Collapse
|
38
|
Cansfield AD, Ladduwahetty T, Sunose M, Ellard K, Lynch R, Newton AL, Lewis A, Bennett G, Zinn N, Thomson DW, Rüger AJ, Feutrill JT, Rausch O, Watt AP, Bergamini G. CZ415, a Highly Selective mTOR Inhibitor Showing in Vivo Efficacy in a Collagen Induced Arthritis Model. ACS Med Chem Lett 2016; 7:768-73. [PMID: 27563401 DOI: 10.1021/acsmedchemlett.6b00149] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Accepted: 06/10/2016] [Indexed: 12/20/2022] Open
Abstract
CZ415, a potent ATP-competitive mTOR inhibitor with unprecedented selectivity over any other kinase is described. In addition to a comprehensive characterization of its activities in vitro, in vitro ADME, and in vivo pharmacokinetic data are reported. The suitability of this inhibitor for studying in vivo mTOR biology is demonstrated in a mechanistic mouse model monitoring mTOR proximal downstream phosphorylation signaling. Furthermore, the compound reported here is the first ATP-competitive mTOR inhibitor described to show efficacy in a semitherapeutic collagen induced arthritis (CIA) mouse model.
Collapse
Affiliation(s)
- Andrew D. Cansfield
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Tammy Ladduwahetty
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Mihiro Sunose
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Katie Ellard
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Rosemary Lynch
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Anthea L. Newton
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Ann Lewis
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Gavin Bennett
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Nico Zinn
- Cellzome AG, Meyerhofstraße
1, 69117 Heidelberg, Germany
| | | | - Anne J. Rüger
- Cellzome AG, Meyerhofstraße
1, 69117 Heidelberg, Germany
| | - John T. Feutrill
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Oliver Rausch
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | - Alan P. Watt
- Cellzome Ltd, Chesterford Research
Park, Saffron Waldon, CB10
1XL, United Kingdom
| | | |
Collapse
|
39
|
Dale T, Clarke PA, Esdar C, Waalboer D, Adeniji-Popoola O, Ortiz-Ruiz MJ, Mallinger A, Samant RS, Czodrowski P, Musil D, Schwarz D, Schneider K, Stubbs M, Ewan K, Fraser E, TePoele R, Court W, Box G, Valenti M, de Haven Brandon A, Gowan S, Rohdich F, Raynaud F, Schneider R, Poeschke O, Blaukat A, Workman P, Schiemann K, Eccles SA, Wienke D, Blagg J. A selective chemical probe for exploring the role of CDK8 and CDK19 in human disease. Nat Chem Biol 2015; 11:973-980. [PMID: 26502155 PMCID: PMC4677459 DOI: 10.1038/nchembio.1952] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 10/01/2015] [Indexed: 12/31/2022]
Abstract
There is unmet need for chemical tools to explore the role of the Mediator complex in human pathologies ranging from cancer to cardiovascular disease. Here we determine that CCT251545, a small-molecule inhibitor of the WNT pathway discovered through cell-based screening, is a potent and selective chemical probe for the human Mediator complex-associated protein kinases CDK8 and CDK19 with >100-fold selectivity over 291 other kinases. X-ray crystallography demonstrates a type 1 binding mode involving insertion of the CDK8 C terminus into the ligand binding site. In contrast to type II inhibitors of CDK8 and CDK19, CCT251545 displays potent cell-based activity. We show that CCT251545 and close analogs alter WNT pathway-regulated gene expression and other on-target effects of modulating CDK8 and CDK19, including expression of genes regulated by STAT1. Consistent with this, we find that phosphorylation of STAT1(SER727) is a biomarker of CDK8 kinase activity in vitro and in vivo. Finally, we demonstrate in vivo activity of CCT251545 in WNT-dependent tumors.
Collapse
Affiliation(s)
- Trevor Dale
- School of Bioscience, Cardiff University, Cardiff, UK
| | - Paul A. Clarke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | | | - Dennis Waalboer
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | | | - Maria-Jesus Ortiz-Ruiz
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Aurélie Mallinger
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Rahul S. Samant
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | | | | | | | | | - Mark Stubbs
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Ken Ewan
- School of Bioscience, Cardiff University, Cardiff, UK
| | | | - Robert TePoele
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Will Court
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Gary Box
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Melanie Valenti
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Alexis de Haven Brandon
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Sharon Gowan
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | | | - Florence Raynaud
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | | | | | | | - Paul Workman
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | | | - Suzanne A. Eccles
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| | - Dirk Wienke
- Merck KGaA, Merck Serono, Darmstadt, Germany
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SW7 3RP
| |
Collapse
|
40
|
Humphrey SJ, James DE, Mann M. Protein Phosphorylation: A Major Switch Mechanism for Metabolic Regulation. Trends Endocrinol Metab 2015; 26:676-687. [PMID: 26498855 DOI: 10.1016/j.tem.2015.09.013] [Citation(s) in RCA: 351] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 09/26/2015] [Accepted: 09/28/2015] [Indexed: 12/20/2022]
Abstract
Metabolism research is undergoing a renaissance because many diseases are increasingly recognized as being characterized by perturbations in intracellular metabolic regulation. Metabolic changes can be conferred through changes to the expression of metabolic enzymes, the concentrations of substrates or products that govern reaction kinetics, or post-translational modification (PTM) of the proteins that facilitate these reactions. On the 60th anniversary since its discovery, reversible protein phosphorylation is widely appreciated as an essential PTM regulating metabolism. With the ability to quantitatively measure dynamic changes in protein phosphorylation on a global scale - hereafter referred to as phosphoproteomics - we are now entering a new era in metabolism research, with mass spectrometry (MS)-based proteomics at the helm.
Collapse
Affiliation(s)
- Sean J Humphrey
- Department of Proteomics and Signal Transduction, Max Planck Institute for Biochemistry, Martinsried 82152, Germany
| | - David E James
- Charles Perkins Centre, School of Molecular Bioscience, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia.
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max Planck Institute for Biochemistry, Martinsried 82152, Germany.
| |
Collapse
|
41
|
Bondeson DP, Mares A, Smith IED, Ko E, Campos S, Miah AH, Mulholland KE, Routly N, Buckley DL, Gustafson JL, Zinn N, Grandi P, Shimamura S, Bergamini G, Faelth-Savitski M, Bantscheff M, Cox C, Gordon DA, Willard RR, Flanagan JJ, Casillas LN, Votta BJ, den Besten W, Famm K, Kruidenier L, Carter PS, Harling JD, Churcher I, Crews CM. Catalytic in vivo protein knockdown by small-molecule PROTACs. Nat Chem Biol 2015; 11:611-7. [PMID: 26075522 PMCID: PMC4629852 DOI: 10.1038/nchembio.1858] [Citation(s) in RCA: 918] [Impact Index Per Article: 91.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/03/2015] [Indexed: 01/01/2023]
Abstract
The current predominant therapeutic paradigm is based on maximizing drug-receptor occupancy to achieve clinical benefit. This strategy, however, generally requires excessive drug concentrations to ensure sufficient occupancy, often leading to adverse side effects. Here, we describe major improvements to the proteolysis targeting chimeras (PROTACs) method, a chemical knockdown strategy in which a heterobifunctional molecule recruits a specific protein target to an E3 ubiquitin ligase, resulting in the target's ubiquitination and degradation. These compounds behave catalytically in their ability to induce the ubiquitination of super-stoichiometric quantities of proteins, providing efficacy that is not limited by equilibrium occupancy. We present two PROTACs that are capable of specifically reducing protein levels by >90% at nanomolar concentrations. In addition, mouse studies indicate that they provide broad tissue distribution and knockdown of the targeted protein in tumor xenografts. Together, these data demonstrate a protein knockdown system combining many of the favorable properties of small-molecule agents with the potent protein knockdown of RNAi and CRISPR.
Collapse
Affiliation(s)
- Daniel P Bondeson
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Alina Mares
- GSK Medicines Research Centre, Stevenage, UK
| | | | - Eunhwa Ko
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | | | | | | | | | - Dennis L Buckley
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Jeffrey L Gustafson
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | - Nico Zinn
- Cellzome, a GSK company, Heidelberg, Germany
| | | | | | | | | | | | - Carly Cox
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
| | | | | | | | - Linda N Casillas
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Bartholomew J Votta
- Pattern Recognition Receptor Discovery Performance Unit, GlaxoSmithKline, Collegeville, Pennsylvania, USA
| | - Willem den Besten
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, California, USA
| | | | | | | | | | | | - Craig M Crews
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, Connecticut, USA
- Department of Chemistry, Yale University, New Haven, Connecticut, USA
- Department of Pharmacology, Yale University, New Haven, Connecticut, USA
| |
Collapse
|
42
|
Bamborough P, Chung CW, Furze RC, Grandi P, Michon AM, Sheppard RJ, Barnett H, Diallo H, Dixon DP, Douault C, Jones EJ, Karamshi B, Mitchell DJ, Prinjha RK, Rau C, Watson RJ, Werner T, Demont EH. Structure-Based Optimization of Naphthyridones into Potent ATAD2 Bromodomain Inhibitors. J Med Chem 2015; 58:6151-78. [PMID: 26230603 DOI: 10.1021/acs.jmedchem.5b00773] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
ATAD2 is a bromodomain-containing protein whose overexpression is linked to poor outcomes in a number of different cancer types. To date, no potent and selective inhibitors of the bromodomain have been reported. This article describes the structure-based optimization of a series of naphthyridones from micromolar leads with no selectivity over the BET bromodomains to inhibitors with sub-100 nM ATAD2 potency and 100-fold BET selectivity.
Collapse
Affiliation(s)
| | | | | | - Paola Grandi
- ∥Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | - Anne-Marie Michon
- ∥Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | - Christina Rau
- ∥Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | | - Thilo Werner
- ∥Cellzome GmbH, Molecular Discovery Research, GlaxoSmithKline, Meyerhofstrasse 1, 69117 Heidelberg, Germany
| | | |
Collapse
|
43
|
Meyer K, Selbach M. Quantitative affinity purification mass spectrometry: a versatile technology to study protein-protein interactions. Front Genet 2015; 6:237. [PMID: 26236332 PMCID: PMC4500955 DOI: 10.3389/fgene.2015.00237] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/25/2015] [Indexed: 01/11/2023] Open
Abstract
While the genomic revolution has dramatically accelerated the discovery of disease-associated genes, the functional characterization of the corresponding proteins lags behind. Most proteins fulfill their tasks in complexes with other proteins, and analysis of protein–protein interactions (PPIs) can therefore provide insights into protein function. Several methods can be used to generate large-scale protein interaction networks. However, most of these approaches are not quantitative and therefore cannot reveal how perturbations affect the network. Here, we illustrate how a clever combination of quantitative mass spectrometry with different biochemical methods provides a rich toolkit to study different aspects of PPIs including topology, subunit stoichiometry, and dynamic behavior.
Collapse
Affiliation(s)
- Katrina Meyer
- Proteome Dynamics, Max Delbrück Center for Molecular Medicine , Berlin, Germany
| | - Matthias Selbach
- Proteome Dynamics, Max Delbrück Center for Molecular Medicine , Berlin, Germany
| |
Collapse
|
44
|
Ursu A, Waldmann H. Hide and seek: Identification and confirmation of small molecule protein targets. Bioorg Med Chem Lett 2015; 25:3079-86. [PMID: 26115575 DOI: 10.1016/j.bmcl.2015.06.023] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Revised: 06/01/2015] [Accepted: 06/04/2015] [Indexed: 12/14/2022]
Abstract
Target identification and confirmation for small molecules is often the rate limiting step in drug discovery. A robust method to identify proteins addressed by small molecules is affinity chromatography using chemical probes. These usually consist of the compound of interest equipped with a linker molecule and a proper tag. Recently, methods emerged that allow the identification of protein targets without prior functionalization of the small molecule of interest. The digest offers an update on the newest developments in the area of target identification with special focus on confirmation techniques.
Collapse
Affiliation(s)
- Andrei Ursu
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany; Chemical Biology, Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44221 Dortmund, Germany
| | - Herbert Waldmann
- Department of Chemical Biology, Max-Planck-Institute of Molecular Physiology, Otto-Hahn-Strasse 11, 44227 Dortmund, Germany; Chemical Biology, Faculty of Chemistry and Chemical Biology, TU Dortmund, Otto-Hahn-Strasse 6, 44221 Dortmund, Germany.
| |
Collapse
|
45
|
Li LY, Zhang K, Jiang H, Xie YM, Liao LD, Chen B, Du ZP, Zhang PX, Chen H, Huang W, Jia W, Cao HH, Zheng W, Li EM, Xu LY. Quantitative proteomics reveals the downregulation of GRB2 as a prominent node of F806-targeted cell proliferation network. J Proteomics 2015; 117:145-55. [PMID: 25659534 DOI: 10.1016/j.jprot.2015.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2014] [Revised: 12/15/2014] [Accepted: 01/18/2015] [Indexed: 02/05/2023]
Abstract
UNLABELLED High-throughput proteomics has successfully identified thousands of proteins as potential therapeutic targets during investigations into mechanisms of drug action. A novel macrolide analog, denoted F806, is a potential antitumor drug. Here, using the quantitative proteomic approach of stable isotope labeling with amino acids in cell culture (SILAC) coupled to high-resolution mass spectrometry (MS), we characterize the F806-regulating protein profiles and identify the potential target molecules or pathways of F806 in esophageal squamous cell carcinoma (ESCC) cells. From a total of 1931 quantified proteins, 181 proteins were found to be down-regulated (FDR p-value<0.1, H/L ratio<0.738), and 119 proteins were up-regulated (FDR p-value<0.1, H/L ratio>1.156). Among the down-regulated proteins, we uncovered the over- and under-represented protein clusters in biological process and molecular function respectively by Gene Ontology analysis. Furthermore, down-regulated and up-regulated proteins were significantly enriched in 37 pathways and 60 sub-pathways by bioinformatic analysis (FDR p-value<0.1), while a down-regulated molecule growth factor receptor-bound protein 2 (GRB2) was a prominent node in fourteen cell proliferation-related sub-pathways. We concluded that GRB2 downregulation would be a potential target of F806 in ESCC cells. BIOLOGICAL SIGNIFICANCE This study used SILAC-based quantitative proteomics screen to systematically characterize molecular changes induced by a novel macrolide analog F806 in esophageal squamous cell carcinoma (ESCC) cells. Followed by bioinformatic analyses, signal pathway networks generated from the quantified proteins, would facilitate future investigation into the further mechanisms of F806 in ESCC cells. Notably, it provided information that growth factor receptor-bound protein 2 (GRB2) would be a prominent node in the F806-targeted cell proliferation network.
Collapse
Affiliation(s)
- Li-Yan Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Kai Zhang
- Tianjin Key Laboratory of Medical Epigenetics, Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, PR China
| | - Hong Jiang
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, Fujian, PR China
| | - Yang-Min Xie
- Experimental Animal Center, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Lian-Di Liao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Bo Chen
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Ze-Peng Du
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Pi-Xian Zhang
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Hong Chen
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, Fujian, PR China
| | - Wei Huang
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, Fujian, PR China
| | - Wei Jia
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, Fujian, PR China
| | - Hui-Hui Cao
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China
| | - Wei Zheng
- Fujian Provincial Key Laboratory of Screening for Novel Microbial Products, Fujian Institute of Microbiology, Fuzhou, Fujian, PR China.
| | - En-Min Li
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Department of Biochemistry and Molecular Biology, Shantou University Medical College, Shantou, Guangdong, PR China.
| | - Li-Yan Xu
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Shantou University Medical College, Shantou, Guangdong, PR China; Institute of Oncologic Pathology, Shantou University Medical College, Shantou, Guangdong, PR China.
| |
Collapse
|
46
|
Médard G, Pachl F, Ruprecht B, Klaeger S, Heinzlmeir S, Helm D, Qiao H, Ku X, Wilhelm M, Kuehne T, Wu Z, Dittmann A, Hopf C, Kramer K, Kuster B. Optimized chemical proteomics assay for kinase inhibitor profiling. J Proteome Res 2015; 14:1574-86. [PMID: 25660469 DOI: 10.1021/pr5012608] [Citation(s) in RCA: 103] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Solid supported probes have proven to be an efficient tool for chemical proteomics. The kinobeads technology features kinase inhibitors covalently attached to Sepharose for affinity enrichment of kinomes from cell or tissue lysates. This technology, combined with quantitative mass spectrometry, is of particular interest for the profiling of kinase inhibitors. It often leads to the identification of new targets for medicinal chemistry campaigns where it allows a two-in-one binding and selectivity assay. The assay can also uncover resistance mechanisms and molecular sources of toxicity. Here we report on the optimization of the kinobead assay resulting in the combination of five chemical probes and four cell lines to cover half the human kinome in a single assay (∼ 260 kinases). We show the utility and large-scale applicability of the new version of kinobeads by reprofiling the small molecule kinase inhibitors Alvocidib, Crizotinib, Dasatinib, Fasudil, Hydroxyfasudil, Nilotinib, Ibrutinib, Imatinib, and Sunitinib.
Collapse
Affiliation(s)
- Guillaume Médard
- Chair of Proteomics and Bioanalytics, Technische Universität München , Freising, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Wang J, Zhang CJ, Zhang J, He Y, Lee YM, Chen S, Lim TK, Ng S, Shen HM, Lin Q. Mapping sites of aspirin-induced acetylations in live cells by quantitative acid-cleavable activity-based protein profiling (QA-ABPP). Sci Rep 2015; 5:7896. [PMID: 25600173 PMCID: PMC5379034 DOI: 10.1038/srep07896] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Accepted: 12/16/2014] [Indexed: 02/06/2023] Open
Abstract
Target-identification and understanding of mechanism-of-action (MOA) are challenging for development of small-molecule probes and their application in biology and drug discovery. For example, although aspirin has been widely used for more than 100 years, its molecular targets have not been fully characterized. To cope with this challenge, we developed a novel technique called quantitative acid-cleavable activity-based protein profiling (QA-ABPP) with combination of the following two parts: (i) activity-based protein profiling (ABPP) and iTRAQ™ quantitative proteomics for identification of target proteins and (ii) acid-cleavable linker-based ABPP for identification of peptides with specific binding sites. It is known that reaction of aspirin with its target proteins leads to acetylation. We thus applied the above technique using aspirin-based probes in human cancer HCT116 cells. We identified 1110 target proteins and 2775 peptides with exact acetylation sites. By correlating these two sets of data, 523 proteins were identified as targets of aspirin. We used various biological assays to validate the effects of aspirin on inhibition of protein synthesis and induction of autophagy which were elicited from the pathway analysis of Aspirin target profile. This technique is widely applicable for target identification in the field of drug discovery and biology, especially for the covalent drugs.
Collapse
Affiliation(s)
- Jigang Wang
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore, 117543
| | - Chong-Jing Zhang
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Science, Baodao Xincun, Danzhou, Hainan, P.R. China, 571737
| | - Jianbin Zhang
- Department of Physiology, Yong Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Yingke He
- Department of Anaesthesiology, Singapore General Hospital, Outram Road, Singapore, 169608
| | - Yew Mun Lee
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore, 117543
| | - Songbi Chen
- Tropical Crops Genetic Resources Institute, Chinese Academy of Tropical Agricultural Science, Baodao Xincun, Danzhou, Hainan, P.R. China, 571737
| | - Teck Kwang Lim
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore, 117543
| | - Shukie Ng
- Department of Physiology, Yong Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, Block MD9, 2 Medical Drive, National University of Singapore, Singapore 117597
| | - Qingsong Lin
- Department of Biological Sciences, 14 Science Drive 4, National University of Singapore, Singapore, 117543
| |
Collapse
|
48
|
Abstract
Small molecule inhibitors of protein kinases are key tools for signal transduction research and represent a major class of targeted drugs. Recent developments in quantitative proteomics enable an unbiased view on kinase inhibitor selectivity and modes of action in the biological context. While chemical proteomics techniques utilizing quantitative mass spectrometry interrogate both target specificity and affinity in cellular extracts, proteome-wide phosphorylation analyses upon kinase inhibitor treatment identify signal transduction pathway and network regulation in an unbiased manner. Thus, critical information is provided to promote new insights into mechanisms of kinase signaling and their relevance for kinase inhibitor drug discovery.
Collapse
Affiliation(s)
- Henrik Daub
- Evotec (München) GmbH, Am Klopferspitz
19a, 82152 Martinsried, Germany
| |
Collapse
|
49
|
Russell C, Lin AJS, Hains P, Simone MI, Robinson PJ, McCluskey A. An integrated flow and microwave approach to a broad spectrum protein kinase inhibitor. RSC Adv 2015. [DOI: 10.1039/c5ra09426g] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The protein kinase inhibitor CTx-0152960 and the piperazinyl analogue CTx-0294885 were prepared using a hybrid flow and microwave approach.
Collapse
Affiliation(s)
- Cecilia Russell
- Centre for Chemical Biology, Chemistry
- School of Environmental and Life Science
- The University of Newcastle
- Callaghan
- Australia
| | - Andrew J. S. Lin
- Centre for Chemical Biology, Chemistry
- School of Environmental and Life Science
- The University of Newcastle
- Callaghan
- Australia
| | - Peter Hains
- Children's Medical Research Institute
- Australia
| | - Michela I. Simone
- Centre for Chemical Biology, Chemistry
- School of Environmental and Life Science
- The University of Newcastle
- Callaghan
- Australia
| | | | - Adam McCluskey
- Centre for Chemical Biology, Chemistry
- School of Environmental and Life Science
- The University of Newcastle
- Callaghan
- Australia
| |
Collapse
|
50
|
Prieto JH, Fischer E, Koncarevic S, Yates J, Becker K. Large-scale differential proteome analysis in Plasmodium falciparum under drug treatment. Methods Mol Biol 2014; 1201:269-79. [PMID: 25388121 DOI: 10.1007/978-1-4939-1438-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
Here, we establish a methodology for large-scale quantitative proteomics using SIL (stable isotope labeling) to examine protein expression changes in trophozoite stages of the malaria parasite Plasmodium falciparum following drug treatment. For this purpose, exposure to (13)C6 (15)N1-isoleucine was optimized in order to obtain 99% atomic enrichment. Proteome fractionation with anion exchange chromatography was used to reduce sample complexity and increase quantitative coverage of protein expression. Tryptic peptides of subfractions were subjected to SCX/RP separation, measured by LC-MS/MS, and quantified using the software tool Census. In drug-treated parasites, we identified a total number of 1,253 proteins, thus increasing the overall number of proteins so far identified in the trophozoite stage by 30% in the previous literature. A relative quantification was obtained for more than 800 proteins. About 5% of proteins showed a clear up- or downregulation upon drug treatment.
Collapse
|