1
|
Wei X, Ning G, Ma H, Yin Y, Ma J, Han L, Chen D, Shi Z. Assessment of in vitro assays and quantitative determination of selectivity and modality of inhibitors targeting the cell cycle regulating, oncogenic cyclin-dependent kinases. Arch Biochem Biophys 2025; 767:110349. [PMID: 39978616 DOI: 10.1016/j.abb.2025.110349] [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: 11/11/2024] [Revised: 01/23/2025] [Accepted: 02/17/2025] [Indexed: 02/22/2025]
Abstract
At the heart of cancer pathology lies the dysregulated cell cycle, which is often driven by aberrant activities of the cell cycle regulating, cyclin-dependent kinases (CDKs). Efforts to harness the therapeutic potential of modulating CDK activities have led to the development of inhibitors with tailored CDK selectivity. However, uniformity in the methods used to evaluate CDK inhibitor selectivity has been lacking and consequently, direct comparison and interpretation of selectivity profiles determined under different assay conditions is difficult. Determination of the inhibition modalities crucial to profiling selectivity of a CDK inhibitor requires thorough kinetic analysis carried out under comparable assay conditions. In this study, we employed a streamlined series of in vitro assays for profiling CDK inhibitors wherein intrinsic inhibition constants and cellular binding parameters were measured by using strategically designed enzymatic inhibition and complementary biophysical assays. Our findings demonstrate the effectiveness of this strategy in determining and quantitatively analyzing the selectivity and inhibition modality of a set of representative CDK inhibitors towards the major oncogenic, cell cycle CDKs. In addition, the assay results provide insights into the inhibitor-target interactions that extend beyond potency and selectivity.
Collapse
Affiliation(s)
- Xiaolu Wei
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, The Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Guidan Ning
- School of Health, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Huitong Ma
- Department of Biology, Division of Discovery Research, Ionova Biotherapeutics Co., Inc, Foshan, 528000, PR China
| | - Yujiao Yin
- Department of Biology, Division of Discovery Research, Ionova Biotherapeutics Co., Inc, Foshan, 528000, PR China
| | - Jianchun Ma
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Liang Han
- School of Health, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| | - Danqi Chen
- Department of Biology, Division of Discovery Research, Ionova Biotherapeutics Co., Inc, Foshan, 528000, PR China.
| | - Zhongfeng Shi
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Pharmacy, The Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China.
| |
Collapse
|
2
|
Mader LK, Borean JE, Keillor JW. A practical guide for the assay-dependent characterisation of irreversible inhibitors. RSC Med Chem 2024; 16:d4md00707g. [PMID: 39526224 PMCID: PMC11544421 DOI: 10.1039/d4md00707g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Irreversible targeted covalent inhibitors, in the past regarded as inappropriately reactive and toxic, have seen a recent resurgence in clinical interest. This paradigm shift is attributed to the exploitation of the two-step mechanism, in which a high affinity and selectivity (i.e., low K I) scaffold binds the target and only then does a pendant low intrinsic reactivity warhead react with the target (moderate k inact). This highlights the importance of evaluating inhibitors by deriving both their K I and k inact values. The development of methods to evaluate these inhibitors by accounting for their time-dependent nature has been crucial to the discovery of promising clinical candidates. Herein, we report all the practical kinetic methods available to date to derive k inact and K I values. These methods include direct observation of covalent modification, continuous assay (Kitz & Wilson) evaluation, and discontinuous incubation and pre-incubation time-dependent IC50 assays. We also provide practical guidelines and examples for performing these assays, comparison of their utility, and perspectives for their extended applications. This review aims to provide clarity about the use of these methods for reporting complete inhibitor kinetic profiles, guiding irreversible drug development towards increased target affinity and selectivity, while modulating in vivo stability and on-target reactivity.
Collapse
Affiliation(s)
- Lavleen K Mader
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| | - Jessica E Borean
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| | - Jeffrey W Keillor
- Department of Chemistry and Biomolecular Sciences, University of Ottawa Ottawa Ontario K1N 6N5 Canada
| |
Collapse
|
3
|
Fayed HS, Bakleh MZ, Ashraf JV, Howarth A, Ebner D, Al Haj Zen A. Selective ROCK Inhibitor Enhances Blood Flow Recovery after Hindlimb Ischemia. Int J Mol Sci 2023; 24:14410. [PMID: 37833857 PMCID: PMC10572734 DOI: 10.3390/ijms241914410] [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: 08/15/2023] [Revised: 09/15/2023] [Accepted: 09/18/2023] [Indexed: 10/15/2023] Open
Abstract
The impairment in microvascular network formation could delay the restoration of blood flow after acute limb ischemia. A high-content screen of a GSK-published kinase inhibitor library identified a set of ROCK inhibitor hits enhancing endothelial network formation. Subsequent kinase activity profiling against a panel of 224 protein kinases showed that two indazole-based ROCK inhibitor hits exhibited high selectivity for ROCK1 and ROCK2 isoforms compared to other ROCK inhibitors. One of the chemical entities, GSK429286, was selected for follow-up studies. We found that GSK429286 was ten times more potent in enhancing endothelial tube formation than Fasudil, a classic ROCK inhibitor. ROCK1 inhibition by RNAi phenocopied the angiogenic phenotype of the GSK429286 compound. Using an organotypic angiogenesis co-culture assay, we showed that GSK429286 formed a dense vascular network with thicker endothelial tubes. Next, mice received either vehicle or GSK429286 (10 mg/kg i.p.) for seven days after hindlimb ischemia induction. As assessed by laser speckle contrast imaging, GSK429286 potentiated blood flow recovery after ischemia induction. At the histological level, we found that GSK429286 significantly increased the size of new microvessels in the regenerating areas of ischemic muscles compared with vehicle-treated ones. Our findings reveal that selective ROCK inhibitors have in vitro pro-angiogenic properties and therapeutic potential to restore blood flow in limb ischemia.
Collapse
Affiliation(s)
- Hend Salah Fayed
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | - Mouayad Zuheir Bakleh
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | | | - Alison Howarth
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Daniel Ebner
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford OX3 7FZ, UK
| | - Ayman Al Haj Zen
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
- BHF Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9DU, UK
| |
Collapse
|
4
|
Screening assays for tyrosine kinase inhibitors:A review. J Pharm Biomed Anal 2022; 223:115166. [DOI: 10.1016/j.jpba.2022.115166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 11/13/2022] [Accepted: 11/14/2022] [Indexed: 11/16/2022]
|
5
|
Wang B, Wu H, Hu C, Wang H, Liu J, Wang W, Liu Q. An overview of kinase downregulators and recent advances in discovery approaches. Signal Transduct Target Ther 2021; 6:423. [PMID: 34924565 PMCID: PMC8685278 DOI: 10.1038/s41392-021-00826-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 10/28/2021] [Accepted: 11/05/2021] [Indexed: 12/17/2022] Open
Abstract
Since the clinical approval of imatinib, the discovery of protein kinase downregulators entered a prosperous age. However, challenges still exist in the discovery of kinase downregulator drugs, such as the high failure rate during development, side effects, and drug-resistance problems. With the progress made through multidisciplinary efforts, an increasing number of new approaches have been applied to solve the above problems during the discovery process of kinase downregulators. In terms of in vitro and in vivo drug evaluation, progress was also made in cellular and animal model platforms for better and more clinically relevant drug assessment. Here, we review the advances in drug design strategies, drug property evaluation technologies, and efficacy evaluation models and technologies. Finally, we discuss the challenges and perspectives in the development of kinase downregulator drugs.
Collapse
Affiliation(s)
- Beilei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Hong Wu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Chen Hu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Haizhen Wang
- Hefei PreceDo pharmaceuticals Co., Ltd, Hefei, Anhui, 230088, People's Republic of China
| | - Jing Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Wenchao Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Qingsong Liu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
- Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| |
Collapse
|
6
|
Choe SW, Kim B, Kim M. Progress of Microfluidic Continuous Separation Techniques for Micro-/Nanoscale Bioparticles. BIOSENSORS 2021; 11:464. [PMID: 34821680 PMCID: PMC8615634 DOI: 10.3390/bios11110464] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 11/07/2021] [Accepted: 11/12/2021] [Indexed: 05/03/2023]
Abstract
Separation of micro- and nano-sized biological particles, such as cells, proteins, and nucleotides, is at the heart of most biochemical sensing/analysis, including in vitro biosensing, diagnostics, drug development, proteomics, and genomics. However, most of the conventional particle separation techniques are based on membrane filtration techniques, whose efficiency is limited by membrane characteristics, such as pore size, porosity, surface charge density, or biocompatibility, which results in a reduction in the separation efficiency of bioparticles of various sizes and types. In addition, since other conventional separation methods, such as centrifugation, chromatography, and precipitation, are difficult to perform in a continuous manner, requiring multiple preparation steps with a relatively large minimum sample volume is necessary for stable bioprocessing. Recently, microfluidic engineering enables more efficient separation in a continuous flow with rapid processing of small volumes of rare biological samples, such as DNA, proteins, viruses, exosomes, and even cells. In this paper, we present a comprehensive review of the recent advances in microfluidic separation of micro-/nano-sized bioparticles by summarizing the physical principles behind the separation system and practical examples of biomedical applications.
Collapse
Affiliation(s)
- Se-woon Choe
- Department of Medical IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea;
- Department of IT Convergence Engineering, Kumoh National Institute of Technology, Gumi 39253, Korea
| | - Bumjoo Kim
- Department of Mechanical Engineering and Automotive Engineering, Kongju National University, Cheonan 1223-24, Korea;
- Department of Future Convergence Engineering, Kongju National University, Cheonan 1223-24, Korea
| | - Minseok Kim
- Department of Mechanical System Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
- Department of Aeronautics, Mechanical and Electronic Convergence Engineering, Kumoh National Institute of Technology, Gumi 39177, Korea
| |
Collapse
|
7
|
Freeman-Cook K, Hoffman RL, Miller N, Almaden J, Chionis J, Zhang Q, Eisele K, Liu C, Zhang C, Huser N, Nguyen L, Costa-Jones C, Niessen S, Carelli J, Lapek J, Weinrich SL, Wei P, McMillan E, Wilson E, Wang TS, McTigue M, Ferre RA, He YA, Ninkovic S, Behenna D, Tran KT, Sutton S, Nagata A, Ornelas MA, Kephart SE, Zehnder LR, Murray B, Xu M, Solowiej JE, Visswanathan R, Boras B, Looper D, Lee N, Bienkowska JR, Zhu Z, Kan Z, Ding Y, Mu XJ, Oderup C, Salek-Ardakani S, White MA, VanArsdale T, Dann SG. Expanding control of the tumor cell cycle with a CDK2/4/6 inhibitor. Cancer Cell 2021; 39:1404-1421.e11. [PMID: 34520734 DOI: 10.1016/j.ccell.2021.08.009] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 06/03/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
The CDK4/6 inhibitor, palbociclib (PAL), significantly improves progression-free survival in HR+/HER2- breast cancer when combined with anti-hormonals. We sought to discover PAL resistance mechanisms in preclinical models and through analysis of clinical transcriptome specimens, which coalesced on induction of MYC oncogene and Cyclin E/CDK2 activity. We propose that targeting the G1 kinases CDK2, CDK4, and CDK6 with a small-molecule overcomes resistance to CDK4/6 inhibition. We describe the pharmacodynamics and efficacy of PF-06873600 (PF3600), a pyridopyrimidine with potent inhibition of CDK2/4/6 activity and efficacy in multiple in vivo tumor models. Together with the clinical analysis, MYC activity predicts (PF3600) efficacy across multiple cell lineages. Finally, we find that CDK2/4/6 inhibition does not compromise tumor-specific immune checkpoint blockade responses in syngeneic models. We anticipate that (PF3600), currently in phase 1 clinical trials, offers a therapeutic option to cancer patients in whom CDK4/6 inhibition is insufficient to alter disease progression.
Collapse
Affiliation(s)
- Kevin Freeman-Cook
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Robert L Hoffman
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Nichol Miller
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Jonathan Almaden
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - John Chionis
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Qin Zhang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Koleen Eisele
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Chaoting Liu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Cathy Zhang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Nanni Huser
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Lisa Nguyen
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Cinthia Costa-Jones
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Sherry Niessen
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Jordan Carelli
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - John Lapek
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Scott L Weinrich
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ping Wei
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Elizabeth McMillan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Elizabeth Wilson
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Tim S Wang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Michele McTigue
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Rose Ann Ferre
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - You-Ai He
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Sacha Ninkovic
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Douglas Behenna
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Khanh T Tran
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Scott Sutton
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Asako Nagata
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Martha A Ornelas
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Susan E Kephart
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Luke R Zehnder
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Brion Murray
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Meirong Xu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - James E Solowiej
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ravi Visswanathan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Britton Boras
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - David Looper
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Nathan Lee
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Jadwiga R Bienkowska
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Zhou Zhu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Zhengyan Kan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Ying Ding
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Xinmeng Jasmine Mu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Cecilia Oderup
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Shahram Salek-Ardakani
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Michael A White
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA
| | - Todd VanArsdale
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA.
| | - Stephen G Dann
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, CA 92121, USA.
| |
Collapse
|
8
|
Freeman-Cook KD, Hoffman RL, Behenna DC, Boras B, Carelli J, Diehl W, Ferre RA, He YA, Hui A, Huang B, Huser N, Jones R, Kephart SE, Lapek J, McTigue M, Miller N, Murray BW, Nagata A, Nguyen L, Niessen S, Ninkovic S, O'Doherty I, Ornelas MA, Solowiej J, Sutton SC, Tran K, Tseng E, Visswanathan R, Xu M, Zehnder L, Zhang Q, Zhang C, Dann S. Discovery of PF-06873600, a CDK2/4/6 Inhibitor for the Treatment of Cancer. J Med Chem 2021; 64:9056-9077. [PMID: 34110834 DOI: 10.1021/acs.jmedchem.1c00159] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Control of the cell cycle through selective pharmacological inhibition of CDK4/6 has proven beneficial in the treatment of breast cancer. Extending this level of control to additional cell cycle CDK isoforms represents an opportunity to expand to additional tumor types and potentially provide benefits to patients that develop tumors resistant to selective CDK4/6 inhibitors. However, broad-spectrum CDK inhibitors have a long history of failure due to safety concerns. In this approach, we describe the use of structure-based drug design and Free-Wilson analysis to optimize a series of CDK2/4/6 inhibitors. Further, we detail the use of molecular dynamics simulations to provide insights into the basis for selectivity against CDK9. Based on overall potency, selectivity, and ADME profile, PF-06873600 (22) was identified as a candidate for the treatment of cancer and advanced to phase 1 clinical trials.
Collapse
Affiliation(s)
- Kevin D Freeman-Cook
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Robert L Hoffman
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Douglas C Behenna
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Britton Boras
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Jordan Carelli
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Wade Diehl
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Rose Ann Ferre
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - You-Ai He
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Andrea Hui
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Buwen Huang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Nanni Huser
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Rhys Jones
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Susan E Kephart
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - John Lapek
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Michele McTigue
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Nichol Miller
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Brion W Murray
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Asako Nagata
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Lisa Nguyen
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Sherry Niessen
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Sacha Ninkovic
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Inish O'Doherty
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Martha A Ornelas
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - James Solowiej
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Scott C Sutton
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Khanh Tran
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Elaine Tseng
- Pfizer Global Research and Development, Eastern Point Road, Groton, Connecticut 06340, United States
| | - Ravi Visswanathan
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Meirong Xu
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Luke Zehnder
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Qin Zhang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Cathy Zhang
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| | - Stephen Dann
- Pfizer Global Research and Development La Jolla, 10770 Science Center Drive, San Diego, California 92121, United States
| |
Collapse
|
9
|
Jiang J, Jiang B, He Z, Ficarro SB, Che J, Marto JA, Gao Y, Zhang T, Gray NS. Discovery of Covalent MKK4/7 Dual Inhibitor. Cell Chem Biol 2020; 27:1553-1560.e8. [PMID: 32916088 DOI: 10.1016/j.chembiol.2020.08.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/04/2020] [Accepted: 08/20/2020] [Indexed: 12/22/2022]
Abstract
MKK4/7 are kinases that phosphorylate JNKs and regulate the MAPK signaling pathway. Their overexpression has been associated with tumorigenesis and aggressiveness in cancers such as breast, prostate, non-small cell lung, and pediatric leukemia, making them a potential target for inhibitor development. Here, we report the discovery, development, and validation of a dual MKK4/7 inhibitor, BSJ-04-122, that covalently targets a conserved cysteine located before the DFG motif and displays excellent kinome selectivity. BSJ-04-122 exhibits potent cellular target engagement and induces robust target-specific downstream effects. The combination of the dual MKK4/7 inhibitor with a selective, covalent JNK inhibitor demonstrated an enhanced antiproliferative activity against triple-negative breast cancer cells. Taken together, the results show that BSJ-04-122 represents a pharmacological probe for MKK4/7 and credential covalent targeting as a way to explore the therapeutic potential of these kinases.
Collapse
Affiliation(s)
- Jie Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Zhixiang He
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Scott B Ficarro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA
| | - Jianwei Che
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Jarrod A Marto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Blais Proteomics Center, Dana-Farber Cancer Institute, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02215, USA; Department of Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Yang Gao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA
| | - Tinghu Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA.
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
10
|
Ferguson FM, Liu Y, Harshbarger W, Huang L, Wang J, Deng X, Capuzzi SJ, Muratov EN, Tropsha A, Muthuswamy S, Westover KD, Gray NS. Synthesis and Structure-Activity Relationships of DCLK1 Kinase Inhibitors Based on a 5,11-Dihydro-6 H-benzo[ e]pyrimido[5,4- b][1,4]diazepin-6-one Scaffold. J Med Chem 2020; 63:7817-7826. [PMID: 32530623 DOI: 10.1021/acs.jmedchem.0c00596] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Doublecortin-like kinase 1 (DCLK1) is a serine/threonine kinase that is overexpressed in gastrointestinal cancers, including esophageal, gastric, colorectal, and pancreatic cancers. DCLK1 is also used as a marker of tuft cells, which regulate type II immunity in the gut. However, the substrates and functions of DCLK1 are understudied. We recently described the first selective DCLK1/2 inhibitor, DCLK1-IN-1, developed to aid the functional characterization of this important kinase. Here we describe the synthesis and structure-activity relationships of 5,11-dihydro-6H-benzo[e]pyrimido[5,4-b][1,4]diazepin-6-one DCLK1 inhibitors, resulting in the identification of DCLK1-IN-1.
Collapse
Affiliation(s)
- Fleur M Ferguson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Yan Liu
- Departments of Radiation Oncology and Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Wayne Harshbarger
- Departments of Radiation Oncology and Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Ling Huang
- Cancer Research Institute and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Jinhua Wang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Xianming Deng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Stephen J Capuzzi
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Eugene N Muratov
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Alexander Tropsha
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Senthil Muthuswamy
- Cancer Research Institute and Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts 02215, United States.,Departments of Medicine and Pathology, Harvard Medical School, Boston, Massachusetts 02215, United States
| | - Kenneth D Westover
- Departments of Radiation Oncology and Biochemistry, The University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Nathanael S Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, Massachusetts 02215, United States
| |
Collapse
|
11
|
Caruso G, Musso N, Grasso M, Costantino A, Lazzarino G, Tascedda F, Gulisano M, Lunte SM, Caraci F. Microfluidics as a Novel Tool for Biological and Toxicological Assays in Drug Discovery Processes: Focus on Microchip Electrophoresis. MICROMACHINES 2020; 11:E593. [PMID: 32549277 PMCID: PMC7344675 DOI: 10.3390/mi11060593] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 06/04/2020] [Accepted: 06/10/2020] [Indexed: 02/07/2023]
Abstract
The last decades of biological, toxicological, and pharmacological research have deeply changed the way researchers select the most appropriate 'pre-clinical model'. The absence of relevant animal models for many human diseases, as well as the inaccurate prognosis coming from 'conventional' pre-clinical models, are among the major reasons of the failures observed in clinical trials. This evidence has pushed several research groups to move more often from a classic cellular or animal modeling approach to an alternative and broader vision that includes the involvement of microfluidic-based technologies. The use of microfluidic devices offers several benefits including fast analysis times, high sensitivity and reproducibility, the ability to quantitate multiple chemical species, and the simulation of cellular response mimicking the closest human in vivo milieu. Therefore, they represent a useful way to study drug-organ interactions and related safety and toxicity, and to model organ development and various pathologies 'in a dish'. The present review will address the applicability of microfluidic-based technologies in different systems (2D and 3D). We will focus our attention on applications of microchip electrophoresis (ME) to biological and toxicological studies as well as in drug discovery and development processes. These include high-throughput single-cell gene expression profiling, simultaneous determination of antioxidants and reactive oxygen and nitrogen species, DNA analysis, and sensitive determination of neurotransmitters in biological fluids. We will discuss new data obtained by ME coupled to laser-induced fluorescence (ME-LIF) and electrochemical detection (ME-EC) regarding the production and degradation of nitric oxide, a fundamental signaling molecule regulating virtually every critical cellular function. Finally, the integration of microfluidics with recent innovative technologies-such as organoids, organ-on-chip, and 3D printing-for the design of new in vitro experimental devices will be presented with a specific attention to drug development applications. This 'composite' review highlights the potential impact of 2D and 3D microfluidic systems as a fast, inexpensive, and highly sensitive tool for high-throughput drug screening and preclinical toxicological studies.
Collapse
Affiliation(s)
- Giuseppe Caruso
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
| | - Nicolò Musso
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy; (N.M.); (G.L.)
| | - Margherita Grasso
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| | - Angelita Costantino
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| | - Giuseppe Lazzarino
- Department of Biomedical and Biotechnological Sciences (BIOMETEC), University of Catania, 95125 Catania, Italy; (N.M.); (G.L.)
| | - Fabio Tascedda
- Department of Life Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
- Center for Neuroscience and Neurotechnology, University of Modena and Reggio Emilia, 41125 Modena, Italy
| | - Massimo Gulisano
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
- Molecular Preclinical and Translational Imaging Research Centre-IMPRonTE, University of Catania, 95125 Catania, Italy
- Interuniversity Consortium for Biotechnology, Area di Ricerca, Padriciano, 34149 Trieste, Italy
| | - Susan M. Lunte
- Ralph N. Adams Institute for Bioanalytical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA;
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
- Department of Chemistry, University of Kansas, Lawrence, KS 66047-1620, USA
| | - Filippo Caraci
- Oasi Research Institute—IRCCS, 94018 Troina (EN), Italy; (M.G.); (F.C.)
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; (A.C.); (M.G.)
| |
Collapse
|
12
|
Miniaturized technologies for high-throughput drug screening enzymatic assays and diagnostics – A review. Trends Analyt Chem 2020. [DOI: 10.1016/j.trac.2020.115862] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
13
|
Ouimet CM, D'Amico CI, Kennedy RT. Droplet sample introduction to microchip gel and zone electrophoresis for rapid analysis of protein-protein complexes and enzymatic reactions. Anal Bioanal Chem 2019; 411:6155-6163. [PMID: 31300857 DOI: 10.1007/s00216-019-02006-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 06/19/2019] [Accepted: 06/28/2019] [Indexed: 02/05/2023]
Abstract
Electrophoresis has demonstrated utility as tool for screening of small molecule modulators of protein-protein interactions and enzyme targets. Screening of large chemical libraries requires high-throughput separations. Such fast separation can be accessed by microchip electrophoresis. Here, microchip gel electrophoresis separations of proteins are achieved in 2.6 s with 1200 V/cm and 3-mm separation lengths. However, such fast separations can still suffer from limited overall throughput from sample introduction constraints. Automated introduction of microfluidic droplets has been demonstrated to overcome this limitation. Most devices for coupling microfluidic droplets to microchip electrophoresis are only compatible with free-solution separations. Here, we present a device that is compatible with coupling droplets to gel and free-solution electrophoresis. In this device, automated sample introduction is based on a novel mechanism of carrier phase separation using the difference in density of the carrier phase and the running buffer. This device is demonstrated for microchip gel electrophoresis and free-solution electrophoresis separations of protein-protein interaction and enzyme samples, respectively. Throughputs of about 10 s per sample are achieved and over 1000 separations are demonstrated without reconditioning of the device. Graphical abstract.
Collapse
Affiliation(s)
- Claire M Ouimet
- Department of Chemistry, University of Michigan, 930 N. University Ave, Ann Arbor, MI, 48109-1055, USA
| | - Cara I D'Amico
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5632, USA
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan, 930 N. University Ave, Ann Arbor, MI, 48109-1055, USA. .,Department of Pharmacology, University of Michigan, 1150 W. Medical Center Dr, Ann Arbor, MI, 48109-5632, USA.
| |
Collapse
|
14
|
Chen J, Kinoshita T, Gururaja T, Sukbuntherng J, James D, Lu D, Whang J, Versele M, Chang BY. The effect of Bruton's tyrosine kinase (BTK) inhibitors on collagen-induced platelet aggregation, BTK, and tyrosine kinase expressed in hepatocellular carcinoma (TEC). Eur J Haematol 2018; 101:604-612. [PMID: 30030853 DOI: 10.1111/ejh.13148] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/03/2018] [Accepted: 07/04/2018] [Indexed: 02/28/2024]
Abstract
OBJECTIVES Bruton's tyrosine kinase (BTK) and tyrosine kinase expressed in hepatocellular carcinoma (TEC) are expressed by human platelets. These kinases participate in platelet activation through the collagen receptor glycoprotein VI and may perform overlapping functions. In clinical studies, BTK inhibitors (ibrutinib, acalabrutinib, tirabrutinib, zanubrutinib) have been associated with increased bleeding risk, which may result from inhibition of BTK alone or of both BTK and TEC, although the role of TEC in bleeding risk remains unclear. METHODS Here, in vitro catalytic and binding activities of ibrutinib and acalabrutinib were determined with four assay systems. Platelet aggregation assays determined inhibitor potency and its relationship to selectivity between BTK and TEC. RESULTS Neither inhibitor was substantially more selective for BTK over TEC. The potencies at which BTK inhibitors suppressed platelet aggregation correlated with the potencies in on-target BTK assays, including those in cells. At clinically relevant plasma concentration, ibrutinib, acalabrutinib, and tirabrutinib inhibited collagen-induced platelet aggregation to a similar extent, despite differing in vitro IC50 s. CONCLUSIONS Our results suggest BTK inhibition is the primary driver for inhibition of platelet aggregation. The subtle differences between these inhibitors suggest only randomized, double-blind, placebo-controlled clinical studies can fully address the bleeding risks of different BTK inhibitors.
Collapse
Affiliation(s)
- Jun Chen
- Pharmacyclics, LLC, an AbbVie Company, Sunnyvale, CA, USA
| | | | | | | | - Danelle James
- Pharmacyclics, LLC, an AbbVie Company, Sunnyvale, CA, USA
| | - Daniel Lu
- Pharmacyclics, LLC, an AbbVie Company, Sunnyvale, CA, USA
| | - Jennifer Whang
- Pharmacyclics, LLC, an AbbVie Company, Sunnyvale, CA, USA
| | | | - Betty Y Chang
- Pharmacyclics, LLC, an AbbVie Company, Sunnyvale, CA, USA
| |
Collapse
|
15
|
Wang Y, Chen Y, Cheng X, Zhang K, Wang H, Liu B, Wang J. Design, synthesis and biological evaluation of pyrimidine derivatives as novel CDK2 inhibitors that induce apoptosis and cell cycle arrest in breast cancer cells. Bioorg Med Chem 2018; 26:3491-3501. [PMID: 29853338 DOI: 10.1016/j.bmc.2018.05.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 04/27/2018] [Accepted: 05/16/2018] [Indexed: 01/28/2023]
Abstract
Cyclin-dependent kinase 2 (CDK2) plays a key role in eukaryotic cell cycle progression which could facilitate the transition from G1 to S phase. The dysregulation of CDK2 is closely related to many cancers. CDK2 is utilized as one of the most studied kinase targets in oncology. In this article, 24 benzamide derivatives were designed, synthesized and investigated for the inhibition activity against CDK2. Our results revealed that the compound 25 is a potent CDK2 inhibitor exhibiting a broad spectrum anti-proliferative activity against several human breast cancer cells. Additionally, compound 25 could block cell cycle at G0 or G1 and induce significant apoptosis in MDA-MB-468 cells. These findings highlight a rationale for further development of CDK2 inhibitors to treat human breast cancer.
Collapse
Affiliation(s)
- Yiting Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Yanmei Chen
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Xiaoling Cheng
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Ke Zhang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China
| | - Hangyu Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China.
| | - Bo Liu
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China.
| | - Jinhui Wang
- Key Laboratory of Xinjiang Phytomedicine Resource and Utilization, Ministry of Education, College of Pharmacy, Shihezi University, Shihezi 832002, China; School of Pharmacy, Xinjiang Medical University, Urumqi 830054, China; College of Pharmacy, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
16
|
Liu R, Liu YC, Meng J, Zhu H, Zhang X. A microfluidics-based mobility shift assay to identify new inhibitors of β-secretase for Alzheimer’s disease. Anal Bioanal Chem 2017; 409:6635-6642. [DOI: 10.1007/s00216-017-0617-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2017] [Revised: 08/24/2017] [Accepted: 08/30/2017] [Indexed: 02/06/2023]
|
17
|
Beeman K, Baumgärtner J, Laubenheimer M, Hergesell K, Hoffmann M, Pehl U, Fischer F, Pieck JC. Integration of an In Situ MALDI-Based High-Throughput Screening Process: A Case Study with Receptor Tyrosine Kinase c-MET. SLAS DISCOVERY 2017; 22:1203-1210. [DOI: 10.1177/2472555217727701] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mass spectrometry (MS) is known for its label-free detection of substrates and products from a variety of enzyme reactions. Recent hardware improvements have increased interest in the use of matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS for high-throughput drug discovery. Despite interest in this technology, several challenges remain and must be overcome before MALDI-MS can be integrated as an automated “in-line reader” for high-throughput drug discovery. Two such hurdles include in situ sample processing and deposition, as well as integration of MALDI-MS for enzymatic screening assays that usually contain high levels of MS-incompatible components. Here we adapt our c-MET kinase assay to optimize for MALDI-MS compatibility and test its feasibility for compound screening. The pros and cons of the Echo (Labcyte) as a transfer system for in situ MALDI-MS sample preparation are discussed. We demonstrate that this method generates robust data in a 1536-grid format. We use the MALDI-MS to directly measure the ratio of c-MET substrate and phosphorylated product to acquire IC50 curves and demonstrate that the pharmacology is unaffected. The resulting IC50 values correlate well between the common label-based capillary electrophoresis and the label-free MALDI-MS detection method. We predict that label-free MALDI-MS-based high-throughput screening will become increasingly important and more widely used for drug discovery.
Collapse
Affiliation(s)
- Katrin Beeman
- Global Research & Development, Discovery Technologies, Discovery Pharmacology, Merck KGaA, Darmstadt, Germany
| | - Jens Baumgärtner
- Analytics Healthcare, Biomolecule Analytics, Merck KGaA, Darmstadt, Germany
| | - Manuel Laubenheimer
- Global Research & Development, Discovery Technologies, Discovery Pharmacology, Merck KGaA, Darmstadt, Germany
| | - Karlheinz Hergesell
- Global Research & Development, Discovery Technologies, Discovery Pharmacology, Merck KGaA, Darmstadt, Germany
| | - Martin Hoffmann
- Global Research & Development, Discovery Technologies, Discovery Pharmacology, Merck KGaA, Darmstadt, Germany
| | - Ulrich Pehl
- Global Research & Development, Discovery Technologies, Discovery Pharmacology, Merck KGaA, Darmstadt, Germany
| | - Frank Fischer
- Analytics Healthcare, Biomolecule Analytics, Merck KGaA, Darmstadt, Germany
| | - Jan-Carsten Pieck
- Global Research & Development, Discovery Technologies, Discovery Pharmacology, Merck KGaA, Darmstadt, Germany
| |
Collapse
|
18
|
Lanyon-Hogg T, Patel NV, Ritzefeld M, Boxall KJ, Burke R, Blagg J, Magee AI, Tate EW. Microfluidic Mobility Shift Assay for Real-Time Analysis of Peptide N-Palmitoylation. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2017; 22:418-424. [PMID: 28296537 PMCID: PMC5453399 DOI: 10.1177/2472555216689529] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2016] [Revised: 12/24/2016] [Accepted: 12/27/2016] [Indexed: 02/06/2023]
Abstract
The Hedgehog pathway is a key developmental signaling pathway but is also implicated in many types of cancer. The extracellular signaling protein Sonic hedgehog (Shh) requires dual lipidation for functional signaling, whereby N-terminal palmitoylation is performed by the enzyme Hedgehog acyltransferase (Hhat). Hhat is an attractive target for small-molecule inhibition to arrest Hedgehog signaling, and methods for assaying Hhat activity are central to understanding its function. However, all existing assays to quantify lipidation of peptides suffer limitations, such as safety hazards, high costs, extensive manual handling, restriction to stopped-assay measurements, or indirect assessment of lipidation. To address these limitations, we developed a microfluidic mobility shift assay (MSA) to analyze Shh palmitoylation. MSA allowed separation of fluorescently labeled Shh amine-substrate and palmitoylated Shh amide-product peptides based on differences in charge and hydrodynamic radius, coupled with online fluorescence intensity measurements for quantification. The MSA format was employed to study Hhat-catalyzed reactions, investigate Hhat kinetics, and determine small-molecule inhibitor IC50 values. Both real-time and stopped assays were performed, with the latter achieved via addition of excess unlabeled Shh peptide. The MSA format therefore allows direct and real-time fluorescence-based measurement of acylation and represents a powerful alternative technique in the study of N-lipidation.
Collapse
Affiliation(s)
| | - Neki V. Patel
- Department of Chemistry, Imperial College London, London, UK
| | | | - Katherine J. Boxall
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Rosemary Burke
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Julian Blagg
- Cancer Research UK Cancer Therapeutics Unit, The Institute of Cancer Research, London, UK
| | - Anthony I. Magee
- Molecular Medicine Section, National Heart & Lung Institute, Imperial College London, London, UK
| | - Edward W. Tate
- Department of Chemistry, Imperial College London, London, UK
| |
Collapse
|
19
|
Ouimet CM, D’Amico CI, Kennedy RT. Advances in capillary electrophoresis and the implications for drug discovery. Expert Opin Drug Discov 2017; 12:213-224. [PMID: 27911223 PMCID: PMC5521262 DOI: 10.1080/17460441.2017.1268121] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Many screening platforms are prone to assay interferences that can be avoided by directly measuring the target or enzymatic product. Capillary electrophoresis (CE) and microchip electrophoresis (MCE) have been applied in a variety of formats to drug discovery. CE provides direct detection of the product allowing for the identification of some forms of assay interference. The high efficiency, rapid separations, and low volume requirements make CE amenable to drug discovery. Areas covered: This article describes advances in capillary electrophoresis throughput, sample introduction, and target assays as they pertain to drug discovery and screening. Instrumental advances discussed include integrated droplet microfluidics platforms and multiplexed arrays. Applications of CE to assays of diverse drug discovery targets, including enzymes and affinity interactions are also described. Expert opinion: Current screening with CE does not fully take advantage of the throughputs or low sample volumes possible with CE and is most suitable as a secondary screening method or for screens that are inaccessible with more common platforms. With further development, droplet microfluidics coupled to MCE could take advantage of the low sample requirements by performing assays on the nanoliter scale at high throughput.
Collapse
Affiliation(s)
- Claire M. Ouimet
- Department of Chemistry, University of Michigan, 930 N. University Ave, Ann Arbor, MI, 48109, United States
| | - Cara I. D’Amico
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, United States
| | - Robert T. Kennedy
- Department of Chemistry, University of Michigan, 930 N. University Ave, Ann Arbor, MI, 48109, United States
- Department of Pharmacology, University of Michigan, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, United States
| |
Collapse
|
20
|
Chen P, Lee NV, Hu W, Xu M, Ferre RA, Lam H, Bergqvist S, Solowiej J, Diehl W, He YA, Yu X, Nagata A, VanArsdale T, Murray BW. Spectrum and Degree of CDK Drug Interactions Predicts Clinical Performance. Mol Cancer Ther 2016; 15:2273-2281. [DOI: 10.1158/1535-7163.mct-16-0300] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Accepted: 07/22/2016] [Indexed: 11/16/2022]
|
21
|
Shetty J, Sinville R, Shumilin IA, Minor W, Zhang J, Hawkinson JE, Georg GI, Flickinger CJ, Herr JC. Recombinant production of enzymatically active male contraceptive drug target hTSSK2 - Localization of the TSKS domain phosphorylated by TSSK2. Protein Expr Purif 2016; 121:88-96. [PMID: 26777341 PMCID: PMC4866589 DOI: 10.1016/j.pep.2016.01.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 01/12/2016] [Accepted: 01/12/2016] [Indexed: 11/26/2022]
Abstract
The testis-specific serine/threonine kinase 2 (TSSK2) has been proposed as a candidate male contraceptive target. Development of a selective inhibitor for this kinase first necessitates the production of highly purified, soluble human TSSK2 and its substrate, TSKS, with high yields and retention of biological activity for crystallography and compound screening. Strategies to produce full-length, soluble, biologically active hTSSK2 in baculovirus expression systems were tested and refined. Soluble preparations of TSSK2 were purified by immobilized-metal affinity chromatography (IMAC) followed by gel filtration chromatography. The biological activities of rec.hTSSK2 were verified by in vitro kinase and mobility shift assays using bacterially produced hTSKS (isoform 2), casein, glycogen synthase peptide (GS peptide) and various TSKS peptides as target substrates. Purified recombinant hTSSK2 showed robust kinase activity in the in vitro kinase assay by phosphorylating hTSKS isoform 2 and casein. The ATP Km values were similar for highly and partially purified fractions of hTSSK2 (2.2 and 2.7 μM, respectively). The broad spectrum kinase inhibitor staurosporine was a potent inhibitor of rec.hTSSK2 (IC50 = 20 nM). In vitro phosphorylation experiments carried out with TSKS (isoform 1) fragments revealed particularly strong phosphorylation of a recombinant N-terminal region representing aa 1-150 of TSKS, indicating that the N-terminus of human TSKS is phosphorylated by human TSSK2. Production of full-length enzymatically active recombinant TSSK2 kinase represents the achievement of a key benchmark for future discovery of TSSK inhibitors as male contraceptive agents.
Collapse
Affiliation(s)
- Jagathpala Shetty
- Department of Cell Biology, Center for Research in Contraceptive and Reproductive Health, University of Virginia, Charlottesville, VA, USA
| | - Rondedrick Sinville
- Institute for Therapeutics Discovery and Development, Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware Street SE, Minneapolis, Minnesota, USA
| | - Igor A Shumilin
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Wladek Minor
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Jianhai Zhang
- Department of Cell Biology, Center for Research in Contraceptive and Reproductive Health, University of Virginia, Charlottesville, VA, USA; Shanxi Key Laboratory of Ecological Animal Science and Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu, Shanxi, China
| | - Jon E Hawkinson
- Institute for Therapeutics Discovery and Development, Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware Street SE, Minneapolis, Minnesota, USA
| | - Gunda I Georg
- Institute for Therapeutics Discovery and Development, Department of Medicinal Chemistry, College of Pharmacy, University of Minnesota, 717 Delaware Street SE, Minneapolis, Minnesota, USA
| | - Charles J Flickinger
- Department of Cell Biology, Center for Research in Contraceptive and Reproductive Health, University of Virginia, Charlottesville, VA, USA
| | - John C Herr
- Department of Cell Biology, Center for Research in Contraceptive and Reproductive Health, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
22
|
Abstract
Biochemical selectivity profiling is an integral part of early drug development. Typically compounds from optimization phase are regularly tested for off-target activities within or across target families. This article presents workflow and critical aspects of biochemical protein kinase profiling based on microfluidic mobility shift assays.
Collapse
Affiliation(s)
- Peter Drueckes
- CPC Screening Sciences, Novartis Pharma AG, Novartis Institutes for Biomedical Research, 4002, Basel, Switzerland.
| |
Collapse
|
23
|
Guetschow ED, Kumar S, Lombard DB, Kennedy RT. Identification of sirtuin 5 inhibitors by ultrafast microchip electrophoresis using nanoliter volume samples. Anal Bioanal Chem 2015; 408:721-31. [PMID: 26635020 DOI: 10.1007/s00216-015-9206-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Revised: 11/13/2015] [Accepted: 11/19/2015] [Indexed: 01/26/2023]
Abstract
Sirtuin 5 (SIRT5) is a member of the sirtuin family of protein deacylases that catalyzes removal of post-translational modifications, such as succinyl and malonyl moieties, on lysine residues. In light of SIRT5's roles in regulating metabolism, and its reported oncogenic functions, SIRT5 modulators would be valuable tools for basic biological research and perhaps clinically. Several fluorescence assays for sirtuin modulators have been developed; however, the use of fluorogenic substrates has the potential to cause false positive results due to interactions of engineered substrates with enzyme or test compounds. Therefore, development of high-throughput screening (HTS) assays based on other methods is valuable. In this study, we report the development of a SIRT5 assay using microchip electrophoresis (MCE) for identification of SIRT5 modulators. A novel SIRT5 substrate based on succinate dehydrogenase (SDH) was developed to allow rapid and efficient separation of substrate and product peptide. To achieve high throughput, samples were injected onto the microchip using a droplet-based scheme. By coupling this approach to existing HTS sample preparation workflows, 1408 samples were analyzed at 0.5 Hz in 46 min. Using a 250 ms separation time, eight MCE injections could be made from each sample generating >11,000 electropherograms during analysis. Of the 1280 chemicals tested, eight were identified as inhibiting SIRT5 activity by at least 70% and verified by dose-response analysis.
Collapse
Affiliation(s)
- Erik D Guetschow
- Department of Chemistry, University of Michigan, 930 N University Ave, Ann Arbor, MI, 48109, USA
| | - Surinder Kumar
- Department of Pathology, University of Michigan, 1301 Catherine St, Ann Arbor, MI, 48109, USA
| | - David B Lombard
- Department of Pathology, University of Michigan, 1301 Catherine St, Ann Arbor, MI, 48109, USA.,Institute of Gerontology, University of Michigan, 300 N Ingalls St, Ann Arbor, MI, 48109, USA
| | - Robert T Kennedy
- Department of Chemistry, University of Michigan, 930 N University Ave, Ann Arbor, MI, 48109, USA. .,Department of Pharmacology, University of Michigan, 1150 W. Medical Center Dr., Ann Arbor, MI, 48109, USA.
| |
Collapse
|
24
|
Nehmé R, Morin P. Advances in capillary electrophoresis for miniaturizing assays on kinase enzymes for drug discovery. Electrophoresis 2015; 36:2768-2797. [DOI: 10.1002/elps.201500239] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/02/2015] [Accepted: 07/14/2015] [Indexed: 11/10/2022]
Affiliation(s)
- Reine Nehmé
- Institut de Chimie Organique et Analytique (ICOA); Université d'Orléans - CNRS; UMR 7311 Orléans France
| | - Philippe Morin
- Institut de Chimie Organique et Analytique (ICOA); Université d'Orléans - CNRS; UMR 7311 Orléans France
| |
Collapse
|
25
|
Alexander LT, Möbitz H, Drueckes P, Savitsky P, Fedorov O, Elkins JM, Deane CM, Cowan-Jacob SW, Knapp S. Type II Inhibitors Targeting CDK2. ACS Chem Biol 2015; 10:2116-25. [PMID: 26158339 DOI: 10.1021/acschembio.5b00398] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Kinases can switch between active and inactive conformations of the ATP/Mg(2+) binding motif DFG, which has been explored for the development of type I or type II inhibitors. However, factors modulating DFG conformations remain poorly understood. We chose CDK2 as a model system to study the DFG in-out transition on a target that was thought to have an inaccessible DFG-out conformation. We used site-directed mutagenesis of key residues identified in structural comparisons in conjunction with biochemical and biophysical characterization of the generated mutants. As a result, we identified key residues that facilitate the DFG-out movement, facilitating binding of type II inhibitors. However, surprisingly, we also found that wild type CDK2 is able to bind type II inhibitors. Using protein crystallography structural analysis of the CDK2 complex with an aminopyrimidine-phenyl urea inhibitor (K03861) revealed a canonical type II binding mode and the first available type II inhibitor CDK2 cocrystal structure. We found that the identified type II inhibitors compete with binding of activating cyclins. In addition, analysis of the binding kinetics of the identified inhibitors revealed slow off-rates. The study highlights the importance of residues that may be distant to the ATP binding pocket in modulating the energetics of the DFG-out transition and hence inhibitor binding. The presented data also provide the foundation for a new class of slow off-rate cyclin-competitive CDK2 inhibitors targeting the inactive DFG-out state of this important kinase target.
Collapse
Affiliation(s)
- Leila T. Alexander
- Structural
Genomics Consortium, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
- Department
of Statistics, University of Oxford, 1 South Parks Road, Oxford, OX1 3TG, United Kingdom
| | - Henrik Möbitz
- Novartis Institutes of Biomedical Research, Basel, Switzerland, Novartis Pharma AG, Postfach, CH-4002 Basel, Switzerland
| | - Peter Drueckes
- Novartis Institutes of Biomedical Research, Basel, Switzerland, Novartis Pharma AG, Postfach, CH-4002 Basel, Switzerland
| | - Pavel Savitsky
- Structural
Genomics Consortium, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
| | - Oleg Fedorov
- Structural
Genomics Consortium, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
- Target
Discovery Institute, University of Oxford, NDM Research Building, Roosevelt
Drive, Oxford, OX3 7FZ, United Kingdom
| | - Jonathan M. Elkins
- Structural
Genomics Consortium, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
- Target
Discovery Institute, University of Oxford, NDM Research Building, Roosevelt
Drive, Oxford, OX3 7FZ, United Kingdom
| | - Charlotte M. Deane
- Department
of Statistics, University of Oxford, 1 South Parks Road, Oxford, OX1 3TG, United Kingdom
| | - Sandra W. Cowan-Jacob
- Novartis Institutes of Biomedical Research, Basel, Switzerland, Novartis Pharma AG, Postfach, CH-4002 Basel, Switzerland
| | - Stefan Knapp
- Structural
Genomics Consortium, University of Oxford, Old Road Campus Research Building,
Roosevelt Drive, Oxford, OX3 7DQ, United Kingdom
- Target
Discovery Institute, University of Oxford, NDM Research Building, Roosevelt
Drive, Oxford, OX3 7FZ, United Kingdom
- Institute
for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str.
9, D-60438 Frankfurt
am Main, Germany
| |
Collapse
|
26
|
Espada A, Molina-Martin M. Capillary electrophoresis and small molecule drug discovery: a perfect match? Drug Discov Today 2012; 17:396-404. [PMID: 22387356 DOI: 10.1016/j.drudis.2012.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2011] [Revised: 12/19/2011] [Accepted: 02/10/2012] [Indexed: 01/27/2023]
Abstract
Capillary electrophoresis (CE) is an analytical technique based on the separation of the analytes within a capillary owing to their different electrophoretic mobilities. It is widely used in pharmaceutical analyses owing to its versatility and high separation power. However, its penetration into the drug discovery scene has been relatively limited until recent years. Several factors have contributed to this low implementation, including the maturity of liquid chromatography, the scarcity of experienced CE practitioners, and certain limitations intrinsic to the technique. Recently, instrumental improvements and the growing demand for analytical information have lead to a continuously expanding range of routine electrophoretic applications throughout pharmaceutical discovery and development. In this article we review CE fundamentals, review well-established CE methodologies in drug discovery of small molecules and discuss trends that, in our opinion, might emerge in the coming years.
Collapse
Affiliation(s)
- Alfonso Espada
- Analytical Technologies Department, Centro de Investigación Lilly SA, Avda de la Industria 30, 28108-Alcobendas, Madrid, Spain
| | | |
Collapse
|
27
|
Okagbare PI, Soper SA. Polymer-based dense fluidic networks for high throughput screening with ultrasensitive fluorescence detection. Electrophoresis 2010; 31:3074-82. [PMID: 20872611 DOI: 10.1002/elps.201000209] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Microfluidics represents a viable platform for performing high throughput screening (HTS) because of its ability to automate fluid handling and generate fluidic networks with high number densities over small footprints appropriate for the simultaneous optical interrogation of many screening assays. While most HTS campaigns depend on fluorescence, readers typically use point detection and serially address the assay results significantly lowering throughput or detection sensitivity due to a low duty cycle. To address this challenge, we present here the fabrication of a high-density microfluidic network packed into the imaging area of a large field-of-view (FoV) ultrasensitive fluorescence detection system. The fluidic channels were 1, 5 or 10 μm (width), 1 μm (depth) with a pitch of 1-10 μm and each fluidic processor was individually addressable. The fluidic chip was produced from a molding tool using hot embossing and thermal fusion bonding to enclose the fluidic channels. A 40× microscope objective (numerical aperture=0.75) created an FoV of 200 μm, providing the ability to interrogate ∼25 channels using the current fluidic configuration. An ultrasensitive fluorescence detection system with a large FoV was used to transduce fluorescence signals simultaneously from each fluidic processor onto the active area of an electron multiplying charge-coupled device. The utility of these multichannel networks for HTS was demonstrated by carrying out the high throughput monitoring of the activity of an enzyme, apurinic Endonuclease 1, used as a model-screening assay.
Collapse
Affiliation(s)
- Paul I Okagbare
- Department of Chemistry, Louisiana State University, Baton Rouge, LA 70803, USA
| | | |
Collapse
|
28
|
Fourtounis J, Falgueyret JP, Sayegh CE. Assessing protein-RNA interactions using microfluidic capillary mobility shift assays. Anal Biochem 2010; 411:161-3. [PMID: 21130065 DOI: 10.1016/j.ab.2010.11.042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2010] [Revised: 11/20/2010] [Accepted: 11/26/2010] [Indexed: 01/15/2023]
Abstract
We describe a novel method of characterizing protein-RNA interactions using a fluorescence-based multiwell capillary electrophoresis platform based on microfluidic technology. As a proof of concept, we studied the binding of human immunodeficiency virus 1 (HIV-1) transactivator of transcription (Tat) to the transactivation-responsive RNA (TAR). We established conditions to quantify the binding of recombinant HIV-1 Tat to TAR RNA and validated the assay by demonstrating the dependence of this interaction on the presence of the UCU bulge in TAR. In addition, we showed that neomycin inhibited Tat-TAR binding in a dose-dependent manner (IC(50)=1.6-3.0μM). This microfluidic-based method is high-throughput screening compatible and may be applicable to targeting other nucleic acid-protein interactions.
Collapse
Affiliation(s)
- Jimmy Fourtounis
- Department of Antiviral Research, Merck Research Laboratories, Montreal, Quebec, Canada H9H 3L1
| | | | | |
Collapse
|