1
|
Ahuja A, Singh S, Murti Y. Chemical Probes Review: Choosing the Right Path Towards Pharmacological Targets in Drug Discovery, Challenges and Future Perspectives. Comb Chem High Throughput Screen 2024; 27:2544-2564. [PMID: 38083882 DOI: 10.2174/0113862073283304231118155730] [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: 10/03/2023] [Revised: 11/04/2023] [Accepted: 11/07/2023] [Indexed: 09/27/2024]
Abstract
Chemical probes are essential for academic research and target validation for disease identification. They facilitate drug discovery, target function investigation, and translation studies. A chemical probe provides starting material that can accelerate therapeutic values and safety measures for identifying any biological target in drug discovery. Essential read outs depend on their versatility in biochemical testing, proving the hypothesis, selectivity, specificity, affinity towards the target site, and valuable in new therapeutic approaches. Disease management will depend upon chemical probes as a primitive tool to ascertain the physicochemical stability for in vivo and in vitro studies useful for clinical trials and industrial application in the future. For cancer research, bacterial infection, and neurodegenerative disorders, chemical probes are integrated circuits which are on pipeline for the drug discovery process Furthermore, pharmacological modulators incorporate activators, crosslinkers, degraders, and inhibitors. Reports accessed depend on their structural, mechanical, biochemical, and pharmacological characterization in drug discovery research. The perspective for designing any chemical probes concludes with the utilization of drug discovery and identification of the potential target. It focuses mainly on evidence-based studies and produces promising results in successfully delivering novel therapeutics to treat cancers and other disorders at the target site. Moreover, natural product pharmacophores like rapamycin, cephalosporin, and α-lactamase are utilized for drug discovery. Chemical probes revolutionize computational-based study design depending on identifying novel targets within the database framework. Chemical probes are the clinical answers for drug development and goforward tools in solving other riddles for scientists and researchers working in this industries.
Collapse
Affiliation(s)
- Ashima Ahuja
- Institute of Pharmaceutical Research, GLA University, Mathura, India, UP, 281406
| | - Sonia Singh
- Institute of Pharmaceutical Research, GLA University, Mathura, India, UP, 281406
| | - Yogesh Murti
- Institute of Pharmaceutical Research, GLA University, Mathura, India, UP, 281406
| |
Collapse
|
2
|
Punzalan C, Wang L, Bajrami B, Yao X. Measurement and utilization of the proteomic reactivity by mass spectrometry. MASS SPECTROMETRY REVIEWS 2024; 43:166-192. [PMID: 36924435 DOI: 10.1002/mas.21837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 02/17/2023] [Accepted: 02/20/2023] [Indexed: 06/18/2023]
Abstract
Chemical proteomics, which involves studying the covalent modifications of proteins by small molecules, has significantly contributed to our understanding of protein function and has become an essential tool in drug discovery. Mass spectrometry (MS) is the primary method for identifying and quantifying protein-small molecule adducts. In this review, we discuss various methods for measuring proteomic reactivity using MS and covalent proteomics probes that engage through reactivity-driven and proximity-driven mechanisms. We highlight the applications of these methods and probes in live-cell measurements, drug target identification and validation, and characterizing protein-small molecule interactions. We conclude the review with current developments and future opportunities in the field, providing our perspectives on analytical considerations for MS-based analysis of the proteomic reactivity landscape.
Collapse
Affiliation(s)
- Clodette Punzalan
- Department of Chemistry, University of Connecticut, Storrs, Connecticut, USA
| | - Lei Wang
- Department of Chemistry, University of Connecticut, Storrs, Connecticut, USA
- AD Bio US, Takeda, Lexington, Massachusetts, 02421, USA
| | - Bekim Bajrami
- Chemical Biology & Proteomics, Biogen, Cambridge, Massachusetts, USA
| | - Xudong Yao
- Department of Chemistry, University of Connecticut, Storrs, Connecticut, USA
- Institute for Systems Biology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
3
|
Huang H, Jones LH. Covalent drug discovery using sulfur(VI) fluoride exchange warheads. Expert Opin Drug Discov 2023:1-11. [PMID: 37243622 DOI: 10.1080/17460441.2023.2218642] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/23/2023] [Indexed: 05/29/2023]
Abstract
INTRODUCTION Covalent drug discovery has traditionally focused on targeting cysteine, but the amino acid is often absent in protein binding sites. This review makes the case to move beyond cysteine labeling using sulfur (VI) fluoride exchange (SuFEx) chemistry to expand the druggable proteome. AREAS COVERED Recent advances in SuFEx medicinal chemistry and chemical biology are described, which have enabled the development of covalent chemical probes that site-selectively engage amino acid residues (including tyrosine, lysine, histidine, serine, and threonine) in binding pockets. Areas covered include chemoproteomic mapping of the targetable proteome, structure-based design of covalent inhibitors and molecular glues, metabolic stability profiling, and synthetic methodologies that have expedited the delivery of SuFEx modulators. EXPERT OPINION Despite recent innovations in SuFEx medicinal chemistry, focused preclinical research is required to ensure the field moves from early chemical probe discovery to the delivery of transformational covalent drug candidates. The authors believe that covalent drug candidates designed to engage residues beyond cysteine using sulfonyl exchange warheads will likely enter clinical trials in the coming years.
Collapse
Affiliation(s)
- Huang Huang
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Lyn H Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Abstract
Measurement of target engagement in cells is critical to understand the molecular pharmacology of drugs and chemical probes. Many targeted protein degraders engage the E3 ligase CRL4CRBN and induce proximity with target neosubstrates resulting in their polyubiquitination and subsequent proteasomal degradation. Here we describe the development of a sensitive and robust cellular NanoBRET-based assay that measures occupancy of the CRBN ligand binding site. The assay is based on a bioluminescence resonance energy transfer (BRET) between NanoLuc luciferase tagged CRBN and a BODIPY-lenalidomide tracer which can be competed out by CRBN ligands, including PROTACs and molecular glues. The assay is compatible with a 384-well plate setup, does not require transfections and can be performed in a single day with only 3-4h of laboratory time. The protocols can be used to design other NanoLuc fusion engagement assays based on BODIPY tracers.
Collapse
|
5
|
Bozzola T, Nilsson UJ, Ellervik U. Direct sialic acid 4-OAc substitution by nitrogen, sulfur and carbon nucleophiles with retention of stereochemistry. RSC Adv 2022; 12:11992-11995. [PMID: 35481106 PMCID: PMC9016497 DOI: 10.1039/d2ra01576e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 04/13/2022] [Indexed: 11/21/2022] Open
Abstract
A direct one-step nucleophilic substitution of the 4-OAc of acetyl protected Neu5Ac is presented. Previously published methods for direct substitution of the 4-OAc are limited to cyclic secondary amines. Here we present conditions that allow for a much wider range of nitrogen nucleophiles as well as thiols and cyanide, to be used. The present investigation significantly broadens the scope of 4-aminations and allows for the introduction of a wide variety of different nucleophiles.
Collapse
Affiliation(s)
- Tiago Bozzola
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University P.O. Box 124 SE-221 00 Lund Sweden
| | - Ulf J Nilsson
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University P.O. Box 124 SE-221 00 Lund Sweden
| | - Ulf Ellervik
- Centre for Analysis and Synthesis, Department of Chemistry, Lund University P.O. Box 124 SE-221 00 Lund Sweden
| |
Collapse
|
6
|
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: 145] [Impact Index Per Article: 48.3] [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
|
7
|
Antolin AA, Workman P, Al-Lazikani B. Public resources for chemical probes: the journey so far and the road ahead. Future Med Chem 2021; 13:731-747. [PMID: 31778323 DOI: 10.4155/fmc-2019-0231] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
High-quality small molecule chemical probes are extremely valuable for biological research and target validation. However, frequent use of flawed small-molecule inhibitors produces misleading results and diminishes the robustness of biomedical research. Several public resources are available to facilitate assessment and selection of better chemical probes for specific protein targets. Here, we review chemical probe resources, discuss their current strengths and limitations, and make recommendations for further improvements. Expert review resources provide in-depth analysis but currently cover only a limited portion of the liganded proteome. Computational resources encompass more proteins and are regularly updated, but have limitations in data availability and curation. We show how biomedical scientists may use these resources to choose the best available chemical probes for their research.
Collapse
Affiliation(s)
- Albert A Antolin
- The Department of Data Science, The Institute of Cancer Research, London, SM2 5NG, UK
- CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
- CRUK ICR/Imperial Convergence Science Centre, London, SM2 5NG, UK
| | - Paul Workman
- CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
- CRUK ICR/Imperial Convergence Science Centre, London, SM2 5NG, UK
| | - Bissan Al-Lazikani
- The Department of Data Science, The Institute of Cancer Research, London, SM2 5NG, UK
- CRUK Cancer Therapeutics Unit, The Institute of Cancer Research, London, SM2 5NG, UK
- CRUK ICR/Imperial Convergence Science Centre, London, SM2 5NG, UK
| |
Collapse
|
8
|
Jörg M, Madden KS. The right tools for the job: the central role for next generation chemical probes and chemistry-based target deconvolution methods in phenotypic drug discovery. RSC Med Chem 2021; 12:646-665. [PMID: 34124668 PMCID: PMC8152813 DOI: 10.1039/d1md00022e] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Accepted: 03/15/2021] [Indexed: 12/15/2022] Open
Abstract
The reconnection of the scientific community with phenotypic drug discovery has created exciting new possibilities to develop therapies for diseases with highly complex biology. It promises to revolutionise fields such as neurodegenerative disease and regenerative medicine, where the development of new drugs has consistently proved elusive. Arguably, the greatest challenge in readopting the phenotypic drug discovery approach exists in establishing a crucial chain of translatability between phenotype and benefit to patients in the clinic. This remains a key stumbling block for the field which needs to be overcome in order to fully realise the potential of phenotypic drug discovery. Excellent quality chemical probes and chemistry-based target deconvolution techniques will be a crucial part of this process. In this review, we discuss the current capabilities of chemical probes and chemistry-based target deconvolution methods and evaluate the next advances necessary in order to fully support phenotypic screening approaches in drug discovery.
Collapse
Affiliation(s)
- Manuela Jörg
- School of Natural and Environmental Sciences, Newcastle University Bedson Building Newcastle upon Tyne NE1 7RU UK
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
| | - Katrina S Madden
- School of Natural and Environmental Sciences, Newcastle University Bedson Building Newcastle upon Tyne NE1 7RU UK
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash University Parkville Victoria 3052 Australia
| |
Collapse
|
9
|
Ortiz Zacarías NV, Chahal KK, Šimková T, van der Horst C, Zheng Y, Inoue A, Theunissen E, Mallee L, van der Es D, Louvel J, IJzerman AP, Handel TM, Kufareva I, Heitman LH. Design and Characterization of an Intracellular Covalent Ligand for CC Chemokine Receptor 2. J Med Chem 2021; 64:2608-2621. [PMID: 33600174 PMCID: PMC7958898 DOI: 10.1021/acs.jmedchem.0c01137] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Covalently acting inhibitors constitute a large and growing fraction of approved
small-molecule therapeutics as well as useful tools for a variety of in
vitro and in vivo applications. Here, we aimed to develop a
covalent antagonist of CC chemokine receptor 2 (CCR2), a class A GPCR that has been
pursued as a therapeutic target in inflammation and immuno-oncology. Based on a known
intracellularly binding CCR2 antagonist, several covalent derivatives were synthesized
and characterized by radioligand binding and functional assays. These studies revealed
compound 14 as an intracellular covalent ligand for CCR2. In
silico modeling followed by site-directed mutagenesis confirmed that
14 forms a covalent bond with one of three proximal cysteine residues,
which can be engaged interchangeably. To our knowledge, compound 14
represents the first covalent ligand reported for CCR2. Due to its unique properties, it
may represent a promising tool for ongoing and future studies of CCR2 pharmacology.
Collapse
Affiliation(s)
- Natalia V Ortiz Zacarías
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Oncode Institute, 2333 CC Leiden, The Netherlands
| | - Kirti K Chahal
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Tereza Šimková
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Cas van der Horst
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Yi Zheng
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Emy Theunissen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Lloyd Mallee
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Daan van der Es
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Julien Louvel
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands
| | - Tracy M Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California 92093, United States
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Einsteinweg 55, 2333 CC, Leiden, The Netherlands.,Oncode Institute, 2333 CC Leiden, The Netherlands
| |
Collapse
|
10
|
Nowak RP, Jones LH. Target Validation Using PROTACs: Applying the Four Pillars Framework. SLAS DISCOVERY 2020; 26:474-483. [PMID: 33334221 DOI: 10.1177/2472555220979584] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional compounds that recruit the E3 ubiquitin ligase machinery to proteins of interest, resulting in their ubiquitination and subsequent proteasomal degradation. Targeted protein degradation has generated considerable interest in drug discovery because inhibition of one particular function of a protein often does not deliver the therapeutic efficacy that results from whole-protein depletion. However, the physicochemistry and intrinsically complex pharmacology of PROTACs present challenges, particularly for the development of orally bioavailable drugs. Here we describe the application of a translational pharmacology framework (called the four pillars) to expedite PROTAC development by informing pharmacokinetic-pharmacodynamic (PKPD) understanding and helping elucidate structure-activity relationships. Experimental methods are reviewed that help illuminate exposure of the drug or probe at the site of action (pillar 1) and engagement of its target(s) (pillar 2) that drive functional pharmacological effects (pillar 3) resulting in modulation of a relevant phenotype (pillar 4). We hope the guidance will be useful to those developing targeted protein degraders and help establish PROTAC molecules as robust target validation chemical probes.
Collapse
Affiliation(s)
- Radosław P Nowak
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lyn H Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
11
|
Jiang X, Yu J, Zhou Z, Kongsted J, Song Y, Pannecouque C, De Clercq E, Kang D, Poongavanam V, Liu X, Zhan P. Molecular design opportunities presented by solvent‐exposed regions of target proteins. Med Res Rev 2019; 39:2194-2238. [PMID: 31002405 DOI: 10.1002/med.21581] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2018] [Revised: 03/09/2019] [Accepted: 03/16/2019] [Indexed: 12/24/2022]
Affiliation(s)
- Xiangyi Jiang
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Ji Yu
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Zhongxia Zhou
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Jacob Kongsted
- Department of Physics, Chemistry and PharmacyUniversity of Southern Denmark Odense Denmark
| | - Yuning Song
- Department of Clinical PharmacyQilu Hospital of Shandong University Jinan China
| | - Christophe Pannecouque
- Rega Institute for Medical ResearchLaboratory of Virology and Chemotherapy Leuven Belgium
| | - Erik De Clercq
- Rega Institute for Medical ResearchLaboratory of Virology and Chemotherapy Leuven Belgium
| | - Dongwei Kang
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | | | - Xinyong Liu
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| | - Peng Zhan
- Department of Medicinal ChemistryKey Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University Jinan Shandong People's Republic of China
| |
Collapse
|
12
|
Jones LH, Xu H, Fadeyi OO. Quantifying drug-target engagement in live cells using sulfonyl fluoride chemical probes. Methods Enzymol 2019; 622:201-220. [PMID: 31155053 DOI: 10.1016/bs.mie.2019.02.010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Phenotypic screening in disease-relevant models identifies small molecule hits with desirable efficacy but often with unknown modes of action. Target identification and validation are integral to successful biomedical research. Technologies are required to validate the biological target (or targets) through which a pharmacological agent is proposed to exert its effects. This work details the rational structure-based design, synthetic preparation and cell-based application of a clickable sulfonyl fluoride chemical probe to directly report on the mechanism of a series of compounds previously discovered in a reporter gene assay. Quantification of drug-target occupancy in living human primary cells enabled a deeper understanding of the molecular pharmacology of the chemotype. The technology described herein should be of broad interest to those involved in chemical biology research and the drug discovery endeavor.
Collapse
Affiliation(s)
- Lyn H Jones
- Jnana Therapeutics, Boston, MA, United States.
| | - Hua Xu
- Pfizer Inc, Medicine Design, Cambridge, MA, United States
| | | |
Collapse
|
13
|
Li J, Wang B, Juba BM, Vazquez M, Kortum SW, Pierce BS, Pacheco M, Roberts L, Strohbach JW, Jones LH, Hett E, Thorarensen A, Telliez JB, Sharei A, Bunnage M, Gilbert JB. Microfluidic-Enabled Intracellular Delivery of Membrane Impermeable Inhibitors to Study Target Engagement in Human Primary Cells. ACS Chem Biol 2017; 12:2970-2974. [PMID: 29088528 DOI: 10.1021/acschembio.7b00683] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Biochemical screening is a major source of lead generation for novel targets. However, during the process of small molecule lead optimization, compounds with excellent biochemical activity may show poor cellular potency, making structure-activity relationships difficult to decipher. This may be due to low membrane permeability of the molecule, resulting in insufficient intracellular drug concentration. The Cell Squeeze platform increases permeability regardless of compound structure by mechanically disrupting the membrane, which can overcome permeability limitations and bridge the gap between biochemical and cellular studies. In this study, we show that poorly permeable Janus kinase (JAK) inhibitors are delivered into primary cells using Cell Squeeze, inhibiting up to 90% of the JAK pathway, while incubation of JAK inhibitors with or without electroporation had no significant effect. We believe this robust intracellular delivery approach could enable more effective lead optimization and deepen our understanding of target engagement by small molecules and functional probes.
Collapse
Affiliation(s)
- Jing Li
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Bu Wang
- SQZ Biotechnologies, 134 Coolidge Avenue, Watertown, Massachusetts 02472, United States
| | - Brian M. Juba
- Inflammation and Immunology Research Unit, Pfizer Inc, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Michael Vazquez
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Steve W. Kortum
- Medicine Design, Pfizer Inc, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Betsy S. Pierce
- Medicine Design, Pfizer Inc, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Michael Pacheco
- Medicine Design, Pfizer Inc, Eastern
Point Road, Groton, Connecticut 06340, United States
| | - Lee Roberts
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Joseph W. Strohbach
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Lyn H. Jones
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Erik Hett
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Atli Thorarensen
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Jean-Baptiste Telliez
- Inflammation and Immunology Research Unit, Pfizer Inc, 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | - Armon Sharei
- SQZ Biotechnologies, 134 Coolidge Avenue, Watertown, Massachusetts 02472, United States
| | - Mark Bunnage
- Medicine Design, Pfizer, Inc., 1 Portland Street, Cambridge, Massachusetts 02139, United States
| | | |
Collapse
|
14
|
An activity-dependent proximity ligation platform for spatially resolved quantification of active enzymes in single cells. Nat Commun 2017; 8:1775. [PMID: 29176560 PMCID: PMC5701173 DOI: 10.1038/s41467-017-01854-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 10/19/2017] [Indexed: 01/06/2023] Open
Abstract
Integration of chemical probes into proteomic workflows enables the interrogation of protein activity, rather than abundance. Current methods limit the biological contexts that can be addressed due to sample homogenization, signal-averaging, and bias toward abundant proteins. Here we report a platform that integrates family-wide chemical probes with proximity-dependent oligonucleotide amplification and imaging to quantify enzyme activity in native contexts with high spatial resolution. Application of this method, activity-dependent proximity ligation (ADPL), to serine hydrolase and cysteine protease enzymes enables quantification of differential enzyme activity resulting from endogenous changes in localization and expression. In a competitive format, small-molecule target engagement with endogenous proteins in live cells can be quantified. Finally, retention of sample architecture enables interrogation of complex environments such as cellular co-culture and patient samples. ADPL should be amenable to diverse probe and protein families to detect active enzymes at scale and resolution out of reach with current methods. The interrogation of enzyme activity involves the ensemble averaging of many cells, loss of spatial relationships and is often biased to abundant proteins. Here the authors develop activity-dependent proximity ligation to quantify enzyme activity at the cellular and sub-cellular level in relevant biological contexts.
Collapse
|
15
|
Jones LH. Understanding the chemically-reactive proteome. MOLECULAR BIOSYSTEMS 2017; 12:1728-30. [PMID: 26726011 DOI: 10.1039/c5mb00760g] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The reactivity of amino acid residues in proteins is context-dependent and difficult to predict. Chemical biology can be used to understand the chemical modifications of proteins to help elucidate the nature of the reactive proteome. The resulting insights can be applied to pharmacoproteomics, target identification and molecular pathology.
Collapse
Affiliation(s)
- Lyn H Jones
- Worldwide Medicinal Chemistry, Pfizer, 610 Main Street, Cambridge MA, 02139, USA.
| |
Collapse
|
16
|
Prediction of intracellular exposure bridges the gap between target- and cell-based drug discovery. Proc Natl Acad Sci U S A 2017; 114:E6231-E6239. [PMID: 28701380 DOI: 10.1073/pnas.1701848114] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Inadequate target exposure is a major cause of high attrition in drug discovery. Here, we show that a label-free method for quantifying the intracellular bioavailability (Fic) of drug molecules predicts drug access to intracellular targets and hence, pharmacological effect. We determined Fic in multiple cellular assays and cell types representing different targets from a number of therapeutic areas, including cancer, inflammation, and dementia. Both cytosolic targets and targets localized in subcellular compartments were investigated. Fic gives insights on membrane-permeable compounds in terms of cellular potency and intracellular target engagement, compared with biochemical potency measurements alone. Knowledge of the amount of drug that is locally available to bind intracellular targets provides a powerful tool for compound selection in early drug discovery.
Collapse
|
17
|
Jones LH, Neubert H. Clinical chemoproteomics-Opportunities and obstacles. Sci Transl Med 2017; 9:9/386/eaaf7951. [PMID: 28424333 DOI: 10.1126/scitranslmed.aaf7951] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 03/06/2017] [Indexed: 12/19/2022]
Abstract
Chemoproteomics is the large-scale study of proteins using chemical methods. Although chemoproteomic techniques are becoming commonplace in preclinical research, few examples have found clinical utility. We explore the prospects for advancing chemoproteomics into the clinical setting to understand drug-target interactions and to identify new therapeutically relevant targets.
Collapse
Affiliation(s)
- Lyn H Jones
- Medicine Design, Pfizer, 610 Main Street, Cambridge, MA 02139, USA.
| | | |
Collapse
|
18
|
Xu H, Roberts LR, Chou S, Pierce B, Narayanan A, Jones LH. Quantitative measurement of intracellular HDAC1/2 drug occupancy using a trans-cyclooctene largazole thiol probe. MEDCHEMCOMM 2017; 8:767-770. [PMID: 30108795 DOI: 10.1039/c6md00633g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 12/23/2016] [Indexed: 11/21/2022]
Abstract
Histone deacetylases (HDACs) regulate diverse cellular processes, and are promising targets for a number of diseases. Here we describe the design and utilization of a largazole-based chemical probe to quantitatively measure the intracellular occupancy of HDAC1 and HDAC2 by dacinostat. Surprisingly, the probe was unable to enrich HDAC3 despite its nanomolar potency in a biochemical assay, further proving the necessity of cell-based target occupancy assays to understand compound potency in physiologically-relevant settings. This occupancy assay has the potential to aid the development of novel HDAC1/2 inhibitors in drug discovery.
Collapse
Affiliation(s)
- Hua Xu
- Pfizer Inc. Medicine Design , 610 Main Street , Cambridge , MA 02135 , USA .
| | - Lee R Roberts
- Pfizer Inc. Medicine Design , 610 Main Street , Cambridge , MA 02135 , USA .
| | - Song Chou
- Pfizer Inc. Rare Diseases Research Unit , 610 Main Street , Cambridge , MA 02135 , USA
| | - Betsy Pierce
- Pfizer Inc. Medicine Design , Eastern Point Road , Groton , CT 06340 , USA
| | - Arjun Narayanan
- Pfizer Inc. Medicine Design , 610 Main Street , Cambridge , MA 02135 , USA .
| | - Lyn H Jones
- Pfizer Inc. Medicine Design , 610 Main Street , Cambridge , MA 02135 , USA .
| |
Collapse
|
19
|
Advances at the intersection of chemical biology and medicinal chemistry. Future Med Chem 2016; 7:2089-90. [PMID: 26571116 DOI: 10.4155/fmc.15.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
20
|
Affiliation(s)
- Lyn H Jones
- a Worldwide Medicinal Chemistry, Pfizer , Cambridge , MA , USA
| |
Collapse
|
21
|
Xu H, Gopalsamy A, Hett EC, Salter S, Aulabaugh A, Kyne RE, Pierce B, Jones LH. Cellular thermal shift and clickable chemical probe assays for the determination of drug-target engagement in live cells. Org Biomol Chem 2016; 14:6179-83. [DOI: 10.1039/c6ob01078d] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Proof of drug-target engagement in physiologically-relevant contexts is a key pillar of successful therapeutic target validation.
Collapse
Affiliation(s)
- Hua Xu
- Worldwide Medicinal Chemistry
- Cambridge
- USA
| | | | | | | | - Ann Aulabaugh
- Structural Biology and Biophysics
- Worldwide Medicinal Chemistry
- Groton
- USA
| | | | | | | |
Collapse
|