1
|
Rawling MJ, Ray-Sinha A, Bestwick M, Carter MK, Chahal SK, Chalmers AJ, Elsey DJ, Giddings A, Gold J, Henderson SH, MacGregor C, Malcolm A, Ortega F, Phelan A, Savory ED, Schwartz AC, Stevenson NG, Turner EL, Vass M, Wright JA, Watson C. Discovery of a Potent, Selective, and Brain-Penetrant Checkpoint Kinase 1 Inhibitor, BEN-28010, for the Treatment of Glioblastoma. J Med Chem 2025. [PMID: 40273287 DOI: 10.1021/acs.jmedchem.5c00279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Glioblastoma (GBM) patients face a dire prognosis and alternative therapeutic options are desperately needed. Inhibition of checkpoint kinase 1 (CHK1) represents a potential therapeutic strategy for GBM through regulation of the DNA damage response (DDR) pathway, but no suitable brain-penetrant CHK1 inhibitors have been reported to date. In this study, we disclose the discovery and optimization of clinical candidate 38 (BEN-28010) as a freely brain-penetrant, potent, and selective CHK1 inhibitor, derived from virtual screening hit 1. In vivo pharmacological studies demonstrated efficacy of orally administered 38 in several GBM CDX and PDX models as a monotherapy and in combination with ionizing radiation, including improved overall survival in an intracranially implanted GBM PDX mouse model. Additionally, 38 utilizes an underrepresented aminoimidazole kinase hinge-binding motif that may have broader utility within kinase inhibitor drug discovery.
Collapse
Affiliation(s)
- Michael J Rawling
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | - Michael Bestwick
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | - Sandeep K Chahal
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, U.K
| | | | | | | | - Jennie Gold
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Scott H Henderson
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | | | - Andrew Malcolm
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Fernando Ortega
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Anne Phelan
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Edward D Savory
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Alejandro C Schwartz
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Neil G Stevenson
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Emma L Turner
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Márton Vass
- BenevolentAI, 4-8 Maple Street, London W1T 5HD, U.K
| | - Jamie A Wright
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| | - Christine Watson
- BenevolentAI, Minerva Building, Babraham Research Campus, Cambridge CB22 3AT, U.K
| |
Collapse
|
2
|
Li W, Zhu K, Liu Y, Liu M, Chen Q. Recent advances in PKC inhibitor development: Structural design strategies and therapeutic applications. Eur J Med Chem 2025; 287:117290. [PMID: 39904144 DOI: 10.1016/j.ejmech.2025.117290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Protein kinase C (PKC) isozymes play critical roles in diverse cellular processes and are implicated in numerous diseases, including cancer, diabetes, and autoimmune disorders. Despite extensive research efforts spanning four decades, only one PKC inhibitor has received clinical approval, highlighting the challenges in developing selective and efficacious PKC-targeting therapeutics. Here we review recent advances in the development of small-molecule PKC inhibitors, focusing on structural design strategies, pharmacological activities, and structure-activity relationships. We analyze emerging approaches including fragment-based drug design, allosteric targeting, and natural product derivatization that have yielded promising new scaffold classes. Special attention is given to innovations in achieving isozyme selectivity, particularly for PKCα and PKCβ, which have proven crucial for therapeutic applications. We discuss how integration of computational methods, structural biology insights, and rational design principles has advanced our understanding of PKC inhibition mechanisms. This comprehensive analysis reveals key challenges in PKC drug development, including the need for enhanced selectivity and reduced off-target effects, while highlighting promising directions for future therapeutic development. Our findings provide a framework for designing next-generation PKC inhibitors with improved clinical potential.
Collapse
Affiliation(s)
- Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuyin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Meixi Liu
- Department of Endocrinology, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, 618000, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
3
|
Zaater MA, El Kerdawy AM, Mahmoud WR, Abou-Seri SM. Going beyond ATP binding site as a novel inhibitor design strategy for tau protein kinases in the treatment of Alzheimer's disease: A review. Int J Biol Macromol 2025; 307:142141. [PMID: 40090653 DOI: 10.1016/j.ijbiomac.2025.142141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 03/01/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
Alzheimer's disease (AD) is among the top mortality causing diseases worldwide. The presence of extracellular β-amyloidosis, as well as intraneuronal neurofibrillary aggregates of the abnormally hyperphosphorylated tau protein are two major characteristics of AD. Targeting protein kinases that are involved in the disease pathways has been a common approach in the fight against AD. Unfortunately, most kinase inhibitors currently available target the adenosine triphosphate (ATP)- binding site, which has proven unsuccessful due to issues with selectivity and resistance. As a result, a pressing need to find other alternative sites beyond the ATP- binding site has profoundly evolved. In this review, we will showcase some case studies of inhibitors of tau protein kinases acting beyond ATP binding site which have shown promising results in alleviating AD.
Collapse
Affiliation(s)
- Marwa A Zaater
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| | - Ahmed M El Kerdawy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt; School of Health and Care Sciences, College of Health and Science, University of Lincoln, Joseph Banks Laboratories, Green Lane, Lincoln, United Kingdom.
| | - Walaa R Mahmoud
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El Aini Street, Cairo 11562, Egypt
| |
Collapse
|
4
|
Navarro AM, Alonso M, Martínez-Pérez E, Lazar T, Gibson TJ, Iserte JA, Tompa P, Marino-Buslje C. Unveiling the Complexity of cis-Regulation Mechanisms in Kinases: A Comprehensive Analysis. Proteins 2025; 93:575-587. [PMID: 39366918 DOI: 10.1002/prot.26751] [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: 04/26/2024] [Revised: 08/29/2024] [Accepted: 09/12/2024] [Indexed: 10/06/2024]
Abstract
Protein cis-regulatory elements (CREs) are regions that modulate the activity of a protein through intramolecular interactions. Kinases, pivotal enzymes in numerous biological processes, often undergo regulatory control via inhibitory interactions in cis. This study delves into the mechanisms of cis regulation in kinases mediated by CREs, employing a combined structural and sequence analysis. To accomplish this, we curated an extensive dataset of kinases featuring annotated CREs, organized into homolog families through multiple sequence alignments. Key molecular attributes, including disorder and secondary structure content, active and ATP-binding sites, post-translational modifications, and disease-associated mutations, were systematically mapped onto all sequences. Additionally, we explored the potential for conformational changes between active and inactive states. Finally, we explored the presence of these kinases within membraneless organelles and elucidated their functional roles therein. CREs display a continuum of structures, ranging from short disordered stretches to fully folded domains. The adaptability demonstrated by CREs in achieving the common goal of kinase inhibition spans from direct autoinhibitory interaction with the active site within the kinase domain, to CREs binding to an alternative site, inducing allosteric regulation revealing distinct types of inhibitory mechanisms, which we exemplify by archetypical representative systems. While this study provides a systematic approach to comprehend kinase CREs, further experimental investigations are imperative to unravel the complexity within distinct kinase families. The insights gleaned from this research lay the foundation for future studies aiming to decipher the molecular basis of kinase dysregulation, and explore potential therapeutic interventions.
Collapse
Affiliation(s)
- Alvaro M Navarro
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Macarena Alonso
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | | | - Tamas Lazar
- VIB-VUB Center for Structural Biology, Flanders Institute for Biotechnology (VIB), Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
| | - Toby J Gibson
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Javier A Iserte
- Structural Bioinformatics Unit, Fundación Instituto Leloir, Buenos Aires, Argentina
| | - Peter Tompa
- VIB-VUB Center for Structural Biology, Flanders Institute for Biotechnology (VIB), Brussels, Belgium
- Structural Biology Brussels, Department of Bioengineering, Vrije Universiteit Brussel, Brussels, Belgium
- Research Centre for Natural Sciences, Hungarian Research Network, Institute of Enzymology, Budapest, Hungary
| | | |
Collapse
|
5
|
Stefanovski D, Manevski D, Ribnikar D, Šeruga B. New Serious Safety Warnings for Targeted Anticancer Agents After Their Initial FDA Approval. Cancers (Basel) 2025; 17:584. [PMID: 40002178 PMCID: PMC11853166 DOI: 10.3390/cancers17040584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 01/28/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Background: New safety concerns about targeted anticancer agents (TAAs) often emerge in the first few years after their initial regulatory approval. Our aim was to determine whether new serious and potentially fatal adverse drug reactions (ADRs) continue to emerge in the updated drug labels of TAAs several years after their initial regulatory approval and whether their emergence can be predicted. Methods: The updated drug labels of TAAs approved by the U.S. Food and Drug Administration before July 2013 were analyzed. Serious and potentially fatal ADRs were identified in the Warnings & Precautions (WPs) and Boxed Warnings (BWs) sections of the updated drug labels. Generalized linear mixed models were used to examine the associations between the number of adverse drug reactions and time, drug type (small molecules vs. monoclonal antibodies), and the availability of companion diagnostics for biomarkers. Results: Among 37 eligible TAAs, 25 (68%) were small molecules and 11 (30%) had available companion diagnostics for the biomarkers. Time was a significant predictor of new WPs (p ˂ 0.001) and BWs (p = 0.008). The updated drug labels of the small molecules received significantly more new WPs (p = 0.042) as compared to monoclonal antibodies. The availability of the companion diagnostics for the biomarkers did not have an impact on the emergence of new ADRs. Conclusions: New serious ADRs of TAAs continue to emerge in updated drug labels several years after their initial regulatory approval. Oncologists, regulators, and payers should be aware of the changing risk-benefit ratios of approved TAAs.
Collapse
Affiliation(s)
- Dimitar Stefanovski
- Division of Medical Oncology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia; (D.S.); (D.R.)
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia
| | - Damjan Manevski
- Institute for Biostatistics and Medical Informatics, Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia;
| | - Domen Ribnikar
- Division of Medical Oncology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia; (D.S.); (D.R.)
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia
| | - Boštjan Šeruga
- Division of Medical Oncology, Institute of Oncology Ljubljana, Zaloška Cesta 2, 1000 Ljubljana, Slovenia; (D.S.); (D.R.)
- Faculty of Medicine, University of Ljubljana, Vrazov Trg 2, 1000 Ljubljana, Slovenia
| |
Collapse
|
6
|
de Brevern AG. Special Issue: "Molecular Dynamics Simulations and Structural Analysis of Protein Domains". Int J Mol Sci 2024; 25:10793. [PMID: 39409122 PMCID: PMC11477144 DOI: 10.3390/ijms251910793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 09/30/2024] [Accepted: 10/07/2024] [Indexed: 10/20/2024] Open
Abstract
The 3D protein structure is the basis for all their biological functions [...].
Collapse
Affiliation(s)
- Alexandre G. de Brevern
- DSIMB Bioinformatics Team, BIGR, INSERM, Université Paris Cité, F-75015 Paris, France; ; Tel.: +33-1-4449-3000
- DSIMB Bioinformatics Team, BIGR, INSERM, Université de la Réunion, F-97715 Saint Denis, France
| |
Collapse
|
7
|
Sun Y, Zhou R, Hu J, Feng S, Hu Q. Reversible control of kinase signaling through chemical-induced dephosphorylation. Commun Biol 2024; 7:1073. [PMID: 39217250 PMCID: PMC11366001 DOI: 10.1038/s42003-024-06771-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The coordination between kinases and phosphatases is crucial for regulating the phosphorylation levels of essential signaling molecules. Methods enabling precise control of kinase activities are valuable for understanding the kinase functions and for developing targeted therapies. Here, we use the abscisic acid (ABA)-induced proximity system to reversibly control kinase signaling by recruiting phosphatases. Using this method, we found that the oncogenic tyrosine kinase BCR::ABL1 can be inhibited by recruiting various cytoplasmic phosphatases. We also discovered that the oncogenic serine/threonine kinase BRAF(V600E), which has been reported to bypass phosphorylation regulation, can be positively regulated by protein phosphatase 1 (PP1) and negatively regulated by PP5. Additionally, we observed that the dual-specificity kinase MEK1 can be inhibited by recruiting PP5. This suggests that bifunctional molecules capable of recruiting PP5 to MEK or RAF kinases could be promising anticancer drug candidates. Thus, the ABA-induced dephosphorylation method enables rapid screening of phosphatases to precisely control kinase signaling.
Collapse
Affiliation(s)
- Ying Sun
- Zhejiang University, Hangzhou, Zhejiang, China
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Rihong Zhou
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jin Hu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shan Feng
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Qi Hu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
- Westlake AI Therapeutics Lab, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
8
|
Ren L, Moreno D, Baer BR, Barbour P, Bettendorf T, Bouhana K, Brown K, Brown SA, Fell JB, Hartley DP, Hicken EJ, Laird ER, Lee P, McCown J, Otten JN, Prigaro B, Wallace R, Kahn D. Identification of the Clinical Candidate PF-07284890 ( ARRY-461), a Highly Potent and Brain Penetrant BRAF Inhibitor for the Treatment of Cancer. J Med Chem 2024; 67:13019-13032. [PMID: 39077892 DOI: 10.1021/acs.jmedchem.4c00998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/31/2024]
Abstract
Mutant BRAFV600E is one of the most common oncogenic drivers in metastatic melanoma. While first generation BRAFV600E inhibitors are capable of controlling tumors systemically, they are unable to adequately treat tumors that have metastasized to the brain due to insufficient penetration across the blood-brain barrier (BBB). Through a combination of structure-based drug design (SBDD) and the optimization of physiochemical properties to enhance BBB penetration, we herein report the discovery of the brain-penetrant BRAFV600E inhibitor PF-07284890 (ARRY-461). In mice studies, ARRY-461 proved to be highly brain-penetrant and was able to drive regressions of A375 BRAFV600E tumors implanted both subcutaneously and intracranially. Based on compelling preclinical safety and efficacy studies, ARRY-461 was progressed into a Phase 1 A/B clinical trial (NCT04543188).
Collapse
Affiliation(s)
- Li Ren
- Enliven Therapeutics, Boulder, Colorado 80301, United States
| | - David Moreno
- Enliven Therapeutics, Boulder, Colorado 80301, United States
| | - Brian R Baer
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | | | | | - Karyn Bouhana
- Cogent Biosciences, Boulder, Colorado 80301, United States
| | - Karin Brown
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | - Suzy A Brown
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | - Jay B Fell
- Cogent Biosciences, Boulder, Colorado 80301, United States
| | | | - Erik J Hicken
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | - Ellen R Laird
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | - Patrice Lee
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | - Joseph McCown
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| | | | | | - Ross Wallace
- Loxo Oncology, Louisville, Colorado 80027, United States
| | - Dean Kahn
- Pfizer Boulder R&D, Boulder, Colorado 80301, United States
| |
Collapse
|
9
|
Nozal V, Fernández-Gómez P, García-Rubia A, Martínez-González L, Cuevas EP, Carro E, Palomo V, Martínez A. Designing multitarget ligands for neurodegenerative diseases with improved permeability trough PLGA nanoencapsulation. Biomed Pharmacother 2024; 175:116626. [PMID: 38663103 DOI: 10.1016/j.biopha.2024.116626] [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: 01/05/2024] [Revised: 04/03/2024] [Accepted: 04/17/2024] [Indexed: 06/03/2024] Open
Abstract
Multitarget ligands (MTLs) have emerged as an interesting alternative for addressing complex multifactorial pathologies such as neurodegenerative diseases. However, a common challenge associated with these compounds is often their high molecular weight and low solubility, which becomes a hurdle when trying to permeate over the blood-brain barrier (BBB). In this study, we have designed two new MTLs that modulate three pharmacological targets simultaneously (tau, beta-amyloid and TAR DNA-binding protein 43). To enhance their brain penetration, we have formulated organic polymeric nanoparticles using poly(lactic-co-glycolic acid). The characterization of the formulations, evaluation of their permeability through an in vitro BBB model, and assessment of their activity on disease-representative cellular models, such as Alzheimer's disease and amyotrophic lateral sclerosis, have been conducted. The results demonstrate the potential of the new MTLs and their nanoparticle encapsulation for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Vanesa Nozal
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain
| | - Paula Fernández-Gómez
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid 28049, Spain
| | - Alfonso García-Rubia
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain
| | - Loreto Martínez-González
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Eva P Cuevas
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain
| | - Eva Carro
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain; Neurobiology of Alzheimer's Disease Unit, Functional Unit for Research into Chronic Diseases, Instituto de Salud Carlos III, Madrid 28029, Spain
| | - Valle Palomo
- Instituto Madrileño de Estudios Avanzados en Nanociencia (IMDEA Nanociencia), Madrid 28049, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain; Unidad de Nanobiotecnología Asociada al Centro Nacional de Biotecnología (CSIC), Madrid 28049, Spain.
| | - Ana Martínez
- Centro de Investigaciones Biológicas Margarita Salas-CSIC, Ramiro de Maeztu 9, Madrid 28040, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid 28031, Spain.
| |
Collapse
|
10
|
Thomas J, Wilson S. Molecular and Therapeutic Targets for Amyloid-beta Plaques in Alzheimer's Disease: A Review Study. Basic Clin Neurosci 2024; 15:1-26. [PMID: 39291090 PMCID: PMC11403107 DOI: 10.32598/bcn.2021.3522.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/06/2021] [Accepted: 09/06/2021] [Indexed: 09/19/2024] Open
Abstract
Alzheimer's disease (AD) is characterized by progressive loss of cognition and a gradual decrease in memory. Although AD is considered the most persistent form of dementia and a global concern, no complete cure or agents that can completely halt the progression of AD have been found. In the past years, significant progress has been made in understanding the cellular and molecular changes associated with AD, and numerous drug targets have been identified for the development of drugs for this disease. Amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFT) are the major attributes of AD. Symptomatic relief is the only possible treatment available at present and a disease-modifying drug is of utmost importance. The development of drugs that can inhibit different targets responsible for the formation of plaques is a potential area in AD research. This review is not a complete list of all possible targets for AD but serves to highlight the targets related to Aβ pathology and pathways concerned with the formation of Aβ fragments. This shall serve as a prospect in the identification of Aβ plaque inhibitors and pave the strategies for newer drug treatments. Nevertheless, substantial research is done in this area but to bridle, the clinical difficulty remains a concern.
Collapse
Affiliation(s)
- Jaya Thomas
- Department of Pharmacology, School of Pharmacy University of Amrita Vishwavidyapeetham, Guntur, India
| | - Samson Wilson
- University of Amrita Vishwavidyapeetham, Coimbatore, India
| |
Collapse
|
11
|
Silnitsky S, Rubin SJS, Zerihun M, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part I: Protein Kinase C Activation and Its Role in Cancer and Cardiovascular Diseases. Int J Mol Sci 2023; 24:17600. [PMID: 38139428 PMCID: PMC10743896 DOI: 10.3390/ijms242417600] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/10/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Protein kinases are one of the most significant drug targets in the human proteome, historically harnessed for the treatment of cancer, cardiovascular disease, and a growing number of other conditions, including autoimmune and inflammatory processes. Since the approval of the first kinase inhibitors in the late 1990s and early 2000s, the field has grown exponentially, comprising 98 approved therapeutics to date, 37 of which were approved between 2016 and 2021. While many of these small-molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP binding pocket have been massively successful for oncological indications, their poor selectively for protein kinase isozymes have limited them due to toxicities in their application to other disease spaces. Thus, recent attention has turned to the use of alternative allosteric binding mechanisms and improved drug platforms such as modified peptides to design protein kinase modulators with enhanced selectivity and other pharmacological properties. Herein we review the role of different protein kinase C (PKC) isoforms in cancer and cardiovascular disease, with particular attention to PKC-family inhibitors. We discuss translational examples and carefully consider the advantages and limitations of each compound (Part I). We also discuss the recent advances in the field of protein kinase modulators, leverage molecular docking to model inhibitor-kinase interactions, and propose mechanisms of action that will aid in the design of next-generation protein kinase modulators (Part II).
Collapse
Affiliation(s)
- Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, Safed 1311502, Israel; (S.S.); (M.Z.)
| |
Collapse
|
12
|
Zerihun M, Rubin SJS, Silnitsky S, Qvit N. An Update on Protein Kinases as Therapeutic Targets-Part II: Peptides as Allosteric Protein Kinase C Modulators Targeting Protein-Protein Interactions. Int J Mol Sci 2023; 24:17504. [PMID: 38139336 PMCID: PMC10743673 DOI: 10.3390/ijms242417504] [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: 11/01/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023] Open
Abstract
Human protein kinases are highly-sought-after drug targets, historically harnessed for treating cancer, cardiovascular disease, and an increasing number of autoimmune and inflammatory conditions. Most current treatments involve small molecule protein kinase inhibitors that interact orthosterically with the protein kinase ATP-binding pocket. As a result, these compounds are often poorly selective and highly toxic. Part I of this series reviews the role of PKC isoforms in various human diseases, featuring cancer and cardiovascular disease, as well as translational examples of PKC modulation applied to human health and disease. In the present Part II, we discuss alternative allosteric binding mechanisms for targeting PKC, as well as novel drug platforms, such as modified peptides. A major goal is to design protein kinase modulators with enhanced selectivity and improved pharmacological properties. To this end, we use molecular docking analysis to predict the mechanisms of action for inhibitor-kinase interactions that can facilitate the development of next-generation PKC modulators.
Collapse
Affiliation(s)
- Mulate Zerihun
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed 1311502, Israel; (M.Z.); (S.S.)
| | - Samuel J. S. Rubin
- Department of Medicine, School of Medicine, Stanford University, 300 Pasteur Drive, Stanford, CA 94305, USA;
| | - Shmuel Silnitsky
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed 1311502, Israel; (M.Z.); (S.S.)
| | - Nir Qvit
- The Azrieli Faculty of Medicine in the Galilee, Bar-Ilan University, Henrietta Szold St. 8, P.O. Box 1589, Safed 1311502, Israel; (M.Z.); (S.S.)
| |
Collapse
|
13
|
Despotović A, Janjetović K, Zogović N, Tovilović-Kovačević G. Pharmacological Akt and JNK Kinase Inhibitors 10-DEBC and SP600125 Potentiate Anti-Glioblastoma Effect of Menadione and Ascorbic Acid Combination in Human U251 Glioblastoma Cells. Biomedicines 2023; 11:2652. [PMID: 37893026 PMCID: PMC10604608 DOI: 10.3390/biomedicines11102652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 09/21/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Glioblastoma multiforme (GBM) is the most lethal primary brain tumor in adults, characterized by a highly invasive nature and therapy resistance. Combination of menadione and ascorbic acid (AA+MD) exerts strong ROS-mediated anti-GBM activity in vitro. The objective of this study was to improve AA+MD anti-GBM potential by modulating the activity of Akt and c-Jun N-terminal kinase (JNK), molecules with an important role in GBM development. The effects of Akt and JNK modulation on AA+MD toxicity in U251 human glioblastoma cells were assessed by cell viability assays, flow cytometry, RNA interference and plasmid overexpression, and immunoblot analysis. The AA+MD induced severe oxidative stress, an early increase in Akt phosphorylation followed by its strong inhibition, persistent JNK activation, and U251 cell death. Small molecule Akt kinase inhibitor 10-DEBC enhanced, while pharmacological and genetic Akt activation decreased, AA+MD-induced toxicity. The U251 cell death potentiation by 10-DEBC correlated with an increase in the combination-induced autophagic flux and was abolished by genetic autophagy silencing. Additionally, pharmacological JNK inhibitor SP600125 augmented combination toxicity toward U251 cells, an effect linked with increased ROS accumulation. These results indicate that small Akt and JNK kinase inhibitors significantly enhance AA+MD anti-GBM effects by autophagy potentiation and amplifying deleterious ROS levels.
Collapse
Affiliation(s)
- Ana Despotović
- Department of Neurophysiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia; (A.D.); (K.J.)
| | - Kristina Janjetović
- Department of Neurophysiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia; (A.D.); (K.J.)
| | - Nevena Zogović
- Department of Neurophysiology, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia; (A.D.); (K.J.)
| | - Gordana Tovilović-Kovačević
- Department of Biochemistry, Institute for Biological Research “Siniša Stanković”—National Institute of the Republic of Serbia, University of Belgrade, Bulevar despota Stefana 142, 11000 Belgrade, Serbia
| |
Collapse
|
14
|
Moreno R, Recio J, Barber S, Gil C, Martinez A. The emerging role of mixed lineage kinase 3 (MLK3) and its potential as a target for neurodegenerative diseases therapies. Eur J Med Chem 2023; 257:115511. [PMID: 37247505 DOI: 10.1016/j.ejmech.2023.115511] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 05/16/2023] [Accepted: 05/22/2023] [Indexed: 05/31/2023]
Abstract
Selective and brain-permeable protein kinase inhibitors are in preclinical development for treating neurodegenerative diseases. Among them, MLK3 inhibitors, with a potent neuroprotective biological action have emerged as valuable agents for the treatment of pathologies such as Alzheimer's, Parkinson's disease and amyotrophic lateral sclerosis. In fact, one MLK3 inhibitor, CEP-1347, reached clinical trials for Parkinson's disease. Additionally, another compound called prostetin/12k, a potent and rather selective MLK3 inhibitor has started clinical development for ALS based on its motor neuron protection in both in vitro and in vivo models. In this review, we will focus on the role of MLK3 in neuron-related cell death processes, neurodegenerative diseases, and the potential advantages of targeting this kinase through pharmacological modulation for neuroprotective treatment.
Collapse
Affiliation(s)
- Ricardo Moreno
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Javier Recio
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Santiago Barber
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
| | - Carmen Gil
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Ana Martinez
- Centro de Investigaciones Biológicas "Margarita Salas"-CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Av. Monforte de Lemos, 3-5, 28029, Madrid, Spain.
| |
Collapse
|
15
|
Wu Y, Walker JR, Westberg M, Ning L, Monje M, Kirkland TA, Lin MZ, Su Y. Kinase-Modulated Bioluminescent Indicators Enable Noninvasive Imaging of Drug Activity in the Brain. ACS CENTRAL SCIENCE 2023; 9:719-732. [PMID: 37122464 PMCID: PMC10141594 DOI: 10.1021/acscentsci.3c00074] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Indexed: 05/03/2023]
Abstract
Aberrant kinase activity contributes to the pathogenesis of brain cancers, neurodegeneration, and neuropsychiatric diseases, but identifying kinase inhibitors that function in the brain is challenging. Drug levels in blood do not predict efficacy in the brain because the blood-brain barrier prevents entry of most compounds. Rather, assessing kinase inhibition in the brain requires tissue dissection and biochemical analysis, a time-consuming and resource-intensive process. Here, we report kinase-modulated bioluminescent indicators (KiMBIs) for noninvasive longitudinal imaging of drug activity in the brain based on a recently optimized luciferase-luciferin system. We develop an ERK KiMBI to report inhibitors of the Ras-Raf-MEK-ERK pathway, for which no bioluminescent indicators previously existed. ERK KiMBI discriminates between brain-penetrant and nonpenetrant MEK inhibitors, reveals blood-tumor barrier leakiness in xenograft models, and reports MEK inhibitor pharmacodynamics in native brain tissues and intracranial xenografts. Finally, we use ERK KiMBI to screen ERK inhibitors for brain efficacy, identifying temuterkib as a promising brain-active ERK inhibitor, a result not predicted from chemical characteristics alone. Thus, KiMBIs enable the rapid identification and pharmacodynamic characterization of kinase inhibitors suitable for treating brain diseases.
Collapse
Affiliation(s)
- Yan Wu
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| | - Joel R. Walker
- Promega
Biosciences LLC, San Luis Obispo, California 93401, United States
| | - Michael Westberg
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department
of Chemistry, Aarhus University, Aarhus 8000, Denmark
| | - Lin Ning
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| | - Michelle Monje
- Department
of Neurology and Neurological Sciences, Stanford University, Stanford, California 94305, United States
- Howard Hughes
Medical Institute, Stanford University, Stanford, California 94305, United States
| | - Thomas A. Kirkland
- Promega
Biosciences LLC, San Luis Obispo, California 93401, United States
| | - Michael Z. Lin
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
- Department
of Pediatrics, Stanford University, Stanford, California 94305, United States
- Department
of Chemical and Systems Biology, Stanford
University, Stanford, California 94305, United States
| | - Yichi Su
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
- Department
of Neurobiology, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
16
|
Hanson QM, Hoxie N, Shen M, Guo H, Cho IJ, Chakraborty I, Aragon BM, Rai G, Patnaik S, Janiszewski JS, Hall MD. Target Class Profiling of Small-Molecule Methyltransferases. ACS Chem Biol 2023; 18:969-981. [PMID: 36976909 PMCID: PMC10983791 DOI: 10.1021/acschembio.3c00124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Target class profiling (TCP) is a chemical biology approach to investigate understudied biological target classes. TCP is achieved by developing a generalizable assay platform and screening curated compound libraries to interrogate the chemical biological space of members of an enzyme family. In this work, we took a TCP approach to investigate inhibitory activity across a set of small-molecule methyltransferases (SMMTases), a subclass of methyltransferase enzymes, with the goal of creating a launchpad to explore this largely understudied target class. Using the representative enzymes nicotinamide N-methyltransferase (NNMT), phenylethanolamine N-methyltransferase (PNMT), histamine N-methyltransferase (HNMT), glycine N-methyltransferase (GNMT), catechol O-methyltransferase (COMT), and guanidinoacetate N-methyltransferase (GAMT), we optimized high-throughput screening (HTS)-amenable assays to screen 27,574 unique small molecules against all targets. From this data set, we identified a novel inhibitor which selectively inhibits the SMMTase HNMT and demonstrated how this platform approach can be leveraged for a targeted drug discovery campaign using the example of HNMT.
Collapse
Affiliation(s)
- Quinlin M Hanson
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Nate Hoxie
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Min Shen
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Hui Guo
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Ig-Jun Cho
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Ipsita Chakraborty
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Brooklyn M Aragon
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Ganesha Rai
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Samarjit Patnaik
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - John S. Janiszewski
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| | - Matthew D Hall
- National Center for Advancing Translational Science, National Institutes of Health, Rockville, 20850, United States of America
| |
Collapse
|
17
|
faiad naief M, Mishaal Mohammed A, Khalaf YH. Kinetic and thermodynamic study of ALP enzyme in the presence and absence MWCNTs and Pt-NPs nanocomposites. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2023.100844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023] Open
|
18
|
Therapeutic Monitoring of Orally Administered, Small-Molecule Anticancer Medications with Tumor-Specific Cellular Protein Targets in Peripheral Fluid Spaces-A Review. Pharmaceutics 2023; 15:pharmaceutics15010239. [PMID: 36678867 PMCID: PMC9864625 DOI: 10.3390/pharmaceutics15010239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 12/29/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Orally administered, small-molecule anticancer drugs with tumor-specific cellular protein targets (OACD) have revolutionized oncological pharmacotherapy. Nevertheless, the differences in exposure to these drugs in the systemic circulation and extravascular fluid compartments have led to several cases of therapeutic failure, in addition to posing unknown risks of toxicity. The therapeutic drug monitoring (TDM) of OACDs in therapeutically relevant peripheral fluid compartments is therefore essential. In this work, the available knowledge regarding exposure to OACD concentrations in these fluid spaces is summarized. A review of the literature was conducted by searching Embase, PubMed, and Web of Science for clinical research articles and case reports published between 10 May 2001 and 31 August 2022. Results show that, to date, penetration into cerebrospinal fluid has been studied especially intensively, in addition to breast milk, leukocytes, peripheral blood mononuclear cells, peritoneal fluid, pleural fluid, saliva and semen. The typical clinical indications of peripheral fluid TDM of OACDs were (1) primary malignancy, (2) secondary malignancy, (3) mental disorder, and (4) the assessment of toxicity. Liquid chromatography-tandem mass spectrometry was most commonly applied for analysis. The TDM of OACDs in therapeutically relevant peripheral fluid spaces is often indispensable for efficient and safe treatments.
Collapse
|
19
|
Olagoke OC, Segatto ALA, Afolabi BA, Ardisson-Araujo D, Aschner M, Rocha JBT. RPS6 transcriptional modulation in neural tissues of Nauphoeta cinerea during streptozotocin-associated sugar metabolism impairment. Comp Biochem Physiol B Biochem Mol Biol 2023; 263:110785. [PMID: 35931314 DOI: 10.1016/j.cbpb.2022.110785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/29/2022] [Accepted: 07/31/2022] [Indexed: 11/19/2022]
Abstract
The use of insects to model molecular events that characterize degenerative conditions was originally met with scepticism. However, the discovery of insect insulin-like peptides in the 1970's and the demonstration of evolutionary conservation of insulin-related signalling from insects to mammals have highlighted the importance and reduced cost of insect models in biomedical research. Here, we expand on our earlier described modelling of streptozotocin-induced brain glucose metabolic disruption in Nauphoeta cinerea, using RNA-sequencing analysis to study the transcriptional and genetic signatures of degeneration and stress signalling when glucose levels are elevated in the brain of the lobster cockroach. Nymphs were randomly divided into three groups: Control (0.8% NaCl), and two single streptozotocin injection doses (74 nmol and 740 nmol). The transcriptional analyses featured a dysregulation of 226 genes at high dose STZ treatment and 278 genes at the low dose. Our mRNA-sequencing data showed that ribosomal protein genes were the most upregulated genes at both 74 and 740 nmol STZ treatment. We therefore used RT-qPCR and relative transcriptional methods to validate our proposed mechanism of brain glucose toxicity-induced degeneration in Nauphoeta cinerea, which involved the upregulation of ribosomal proteins and rpS6 regulators (mTORC1, protein kinases, casein kinase 1 and Death-associated protein kinase), the upregulation of MAPK cascades (RAS, ERK, P38 and JNK), alongside the downregulation of the PI3K/AKT cascade. Taken together, this study highlights the remarkable opportunity for Nauphoeta cinerea use as an experimental organism in hyperglycaemia, degeneration, and stress signalling.
Collapse
Affiliation(s)
- Olawande C Olagoke
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences (CCNE), Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil; Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA.
| | - Ana L A Segatto
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences (CCNE), Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil
| | | | - Daniel Ardisson-Araujo
- Department of Cell Biology, Institute of Biological Sciences, University of Brasilia, DF 70910-900, Brazil
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - João B T Rocha
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences (CCNE), Federal University of Santa Maria, 97105-900 Santa Maria, RS, Brazil
| |
Collapse
|
20
|
Jang HY, Oh JM, Kim IW. Drug repurposing using meta-analysis of gene expression in Alzheimer's disease. Front Neurosci 2022; 16:989174. [PMID: 36440278 PMCID: PMC9684643 DOI: 10.3389/fnins.2022.989174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/19/2022] [Indexed: 01/25/2023] Open
Abstract
INTRODUCTION Alzheimer's disease and other forms of dementia are disease that bring an increased global burden. However, the medicine developed to date remains limited. The purpose of this study is to predict drug repositioning candidates using a computational method that integrates gene expression profiles on Alzheimer's disease and compound-induced changes in gene expression levels. METHODS Gene expression data on Alzheimer's disease were obtained from the Gene Expression Omnibus (GEO) and we conducted a meta-analysis of their gene expression levels. The reverse scores of compound-induced gene expressions were computed based on the reversal relationship between disease and drug gene expression profiles. RESULTS Reversal genes and the candidate compounds were identified by the leave-one-out cross-validation procedure. Additionally, the half-maximal inhibitory concentration (IC50) values and the blood-brain barrier (BBB) permeability of candidate compounds were obtained from ChEMBL and PubChem, respectively. CONCLUSION New therapeutic target genes and drug candidates against Alzheimer's disease were identified by means of drug repositioning.
Collapse
Affiliation(s)
- Ha Young Jang
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea
| | - Jung Mi Oh
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea,College of Pharmacy, Seoul National University, Seoul, South Korea
| | - In-Wha Kim
- Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, South Korea,*Correspondence: In-Wha Kim,
| |
Collapse
|
21
|
Baier A, Szyszka R. CK2 and protein kinases of the CK1 superfamily as targets for neurodegenerative disorders. Front Mol Biosci 2022; 9:916063. [PMID: 36275622 PMCID: PMC9582958 DOI: 10.3389/fmolb.2022.916063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/02/2022] [Indexed: 11/13/2022] Open
Abstract
Casein kinases are involved in a variety of signaling pathways, and also in inflammation, cancer, and neurological diseases. Therefore, they are regarded as potential therapeutic targets for drug design. Recent studies have highlighted the importance of the casein kinase 1 superfamily as well as protein kinase CK2 in the development of several neurodegenerative pathologies, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis. CK1 kinases and their closely related tau tubulin kinases as well as CK2 are found to be overexpressed in the mammalian brain. Numerous substrates have been detected which play crucial roles in neuronal and synaptic network functions and activities. The development of new substances for the treatment of these pathologies is in high demand. The impact of these kinases in the progress of neurodegenerative disorders, their bona fide substrates, and numerous natural and synthetic compounds which are able to inhibit CK1, TTBK, and CK2 are discussed in this review.
Collapse
Affiliation(s)
- Andrea Baier
- Institute of Biological Sciences, The John Paul II Catholic University of Lublin, Lublin, Poland
| | - Ryszard Szyszka
- Institute of Biological Sciences, The John Paul II Catholic University of Lublin, Lublin, Poland
| |
Collapse
|
22
|
Sueca-Comes M, Rusu EC, Grabowska AM, Bates DO. Looking Under the Lamppost: The Search for New Cancer Targets in the Human Kinome. Pharmacol Rev 2022; 74:1136-1145. [PMID: 36180110 DOI: 10.1124/pharmrev.121.000410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 02/02/2022] [Accepted: 02/15/2022] [Indexed: 11/22/2022] Open
Abstract
The number of cancer drugs is increasing as new chemical entities are developed to target molecules, often protein kinases, driving cancer progression. In 2009, Fedorov et al. identified that of the protein kinases in the human kinome, most of the focus has been on a small subset. They highlighted that many poorly investigated protein kinases were cancer drivers, but there was no relationship between publications and involvement in cancer development or progression. Since 2009, there has been a doubling in the number of publications, patents, and drugs targeting the kinome. To determine whether this was an expansion in knowledge of well-studied targets-searching in the light under the lamppost-or an explosion of investigations into previously poorly investigated targets, we searched the literature for publications on each kinase, updating Federov et al.'s assessment of the druggable kinome. The proportion of papers focusing on the 50 most-studied kinases had not changed, and the makeup of those 50 had barely changed. The majority of new drugs (80%) were against the same group of 50 kinases identified as targets 10 years ago, and the proportion of studies investigating previously poorly investigated kinases (<1%) was unchanged. With three exceptions [p38 mitogenactivated protein kinase (p38a), AMP-activated protein kinase catalytic α-subunit 1,2, and B-Raf proto-oncogene (BRAF) serine/threonine kinase], >95% of publications addressing kinases still focused on a relatively small proportion (<50%) of the human kinome independently of their involvement as cancer drivers. There is, therefore, still extensive scope for discovery of therapeutics targeting different protein kinases in cancer and still a bias toward well-characterized targets over the innovative searchlight into the unknown. SIGNIFICANCE STATEMENT: This study presents evidence that drug discovery efforts in cancer are still to some extent focused on a narrow group of well-studied kinases 10 years after the identification of multiple novel cancer targets in the human kinome. This suggests that there is still room for researchers in academia, industry, and the not-for-profit sector to develop new and diverse therapies targeting kinases for cancer.
Collapse
Affiliation(s)
- Mireia Sueca-Comes
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| | - Elena Cristina Rusu
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| | - Anna M Grabowska
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| | - David O Bates
- Division of Cancer and Stem Cells, Centre for Cancer Sciences, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom (M.S.-C., A.M.G., D.O.B.); Institute of Integrative Systems Biology (I2Sysbio), University of Valencia and Consejo Superior de Investigaciones Científicas, Valencia, Spain (E.C.R.); and SeqPlexing SL, Valencia, Spain (E.C.R.)
| |
Collapse
|
23
|
Thakur G, Kumar V, Lee KW, Won C. Structural Insights and Development of LRRK2 Inhibitors for Parkinson's Disease in the Last Decade. Genes (Basel) 2022; 13:1426. [PMID: 36011337 PMCID: PMC9408223 DOI: 10.3390/genes13081426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/09/2022] [Accepted: 08/10/2022] [Indexed: 12/01/2022] Open
Abstract
Parkinson's disease (PD) is the second most prevalent neurodegenerative disease, characterized by the specific loss of dopaminergic neurons in the midbrain. The pathophysiology of PD is likely caused by a variety of environmental and hereditary factors. Many single-gene mutations have been linked to this disease, but a significant number of studies indicate that mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) are a potential therapeutic target for both sporadic and familial forms of PD. Consequently, the identification of potential LRRK2 inhibitors has been the focus of drug discovery. Various investigations have been conducted in academic and industrial organizations to investigate the mechanism of LRRK2 in PD and further develop its inhibitors. This review summarizes the role of LRRK2 in PD and its structural details, especially the kinase domain. Furthermore, we reviewed in vitro and in vivo findings of selected inhibitors reported to date against wild-type and mutant versions of the LRRK2 kinase domain as well as the current trends researchers are employing in the development of LRRK2 inhibitors.
Collapse
Affiliation(s)
- Gunjan Thakur
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| | - Vikas Kumar
- Division of Life Sciences, Department of Bio & Medical Big Data (BK4 Program), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea
| | - Keun Woo Lee
- Division of Life Sciences, Department of Bio & Medical Big Data (BK4 Program), Research Institute of Natural Science (RINS), Gyeongsang National University (GNU), 501 Jinju-daero, Jinju 52828, Korea
| | - Chungkil Won
- Institute of Animal Medicine, College of Veterinary Medicine, Gyeongsang National University, Jinju 52828, Korea
| |
Collapse
|
24
|
Inhibition of PLK2 activity affects APP and tau pathology and improves synaptic content in a sex-dependent manner in a 3xTg mouse model of Alzheimer's disease. Neurobiol Dis 2022; 172:105833. [PMID: 35905928 DOI: 10.1016/j.nbd.2022.105833] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Revised: 07/14/2022] [Accepted: 07/22/2022] [Indexed: 11/23/2022] Open
Abstract
Converging lines of evidence suggest that abnormal accumulation of the kinase Polo-like kinase 2 (PLK2) might play a role in the pathogenesis of Alzheimer's disease (AD), possibly through its role in regulating the amyloid β (Aβ) cascade. In the present study, we investigated the effect of inhibiting PLK2 kinase activity in in vitro and in vivo models of AD neuropathology. First, we confirmed that PLK2 overexpression modulated APP and Tau protein levels and phosphorylation in cell culture, in a kinase activity dependent manner. Furthermore, a transient treatment of triple transgenic mouse model of AD (3xTg-AD) with a potent and specific PLK2 pharmacological inhibitor (PLK2i #37) reduced some neuropathological aspects in a sex-dependent manner. In 3xTg-AD males, treatment with PLK2i #37 led to lower Tau burden, higher synaptic protein content, and prevented learning and memory deficits. In contrast, treated females showed an exacerbation of Tau pathology, associated with a reduction in amyloid plaque accumulation. Overall, our findings suggest that PLK2 inhibition alters key components of AD neuropathology in a sex-dependent manner and might display a therapeutic potential for the treatment for AD and related dementia.
Collapse
|
25
|
Eshraghi M, Ahmadi M, Afshar S, Lorzadeh S, Adlimoghaddam A, Rezvani Jalal N, West R, Dastghaib S, Igder S, Torshizi SRN, Mahmoodzadeh A, Mokarram P, Madrakian T, Albensi BC, Łos MJ, Ghavami S, Pecic S. Enhancing autophagy in Alzheimer's disease through drug repositioning. Pharmacol Ther 2022; 237:108171. [PMID: 35304223 DOI: 10.1016/j.pharmthera.2022.108171] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/18/2022] [Accepted: 03/08/2022] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is one of the biggest human health threats due to increases in aging of the global population. Unfortunately, drugs for treating AD have been largely ineffective. Interestingly, downregulation of macroautophagy (autophagy) plays an essential role in AD pathogenesis. Therefore, targeting autophagy has drawn considerable attention as a therapeutic approach for the treatment of AD. However, developing new therapeutics is time-consuming and requires huge investments. One of the strategies currently under consideration for many diseases is "drug repositioning" or "drug repurposing". In this comprehensive review, we have provided an overview of the impact of autophagy on AD pathophysiology, reviewed the therapeutics that upregulate autophagy and are currently used in the treatment of other diseases, including cancers, and evaluated their repurposing as a possible treatment option for AD. In addition, we discussed the potential of applying nano-drug delivery to neurodegenerative diseases, such as AD, to overcome the challenge of crossing the blood brain barrier and specifically target molecules/pathways of interest with minimal side effects.
Collapse
Affiliation(s)
- Mehdi Eshraghi
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Saeid Afshar
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Shahrokh Lorzadeh
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Aida Adlimoghaddam
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada
| | | | - Ryan West
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America
| | - Sanaz Dastghaib
- Endocrinology and Metabolism Research Center, Shiraz University of Medical Sciences, Shiraz Iran
| | - Somayeh Igder
- Department of Clinical Biochemistry, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Amir Mahmoodzadeh
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah 6734667149, Iran
| | - Pooneh Mokarram
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan, Iran; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Benedict C Albensi
- St. Boniface Hospital Albrechtsen Research Centre, Division of Neurodegenerative Disorders, Winnipeg, MB R2H2A6, Canada; Nova Southeastern Univ. College of Pharmacy, Davie, FL, United States of America; University of Manitoba, College of Medicine, Winnipeg, MB R3E 0V9, Canada
| | - Marek J Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Saeid Ghavami
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada; Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada; Faculty of Medicine in Zabrze, University of Technology in Katowice, Academia of Silesia, 41-800 Zabrze, Poland
| | - Stevan Pecic
- Department of Chemistry and Biochemistry, California State University, Fullerton, United States of America.
| |
Collapse
|
26
|
Manley PW, Huth F, Moussaoui S, Schoepfer J. A kinase inhibitor which specifically targets the ABL myristate pocket (STAMP), but unlike asciminib crosses the blood–brain barrier. Bioorg Med Chem Lett 2022; 59:128577. [DOI: 10.1016/j.bmcl.2022.128577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 01/03/2022] [Accepted: 01/15/2022] [Indexed: 11/25/2022]
|
27
|
Brown DG, Wobst HJ. A survey of the clinical pipeline in neuroscience. Bioorg Med Chem Lett 2022; 56:128482. [PMID: 34864194 DOI: 10.1016/j.bmcl.2021.128482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 11/02/2022]
Abstract
Many new first-in-class drugs for neuroscience indications have been introduced in the past decade including new treatments for migraine, amyotrophic lateral sclerosis, depression, and multiple sclerosis. However, significant unmet patient needs remain in areas such as chronic pain, neurodegeneration, psychiatric diseases, and epilepsy. This review summarizes some of the advanced clinical compounds for these indications. Additionally, current opportunities and challenges that remain with respect to genetic validation, biomarkers, and translational models are discussed.
Collapse
Affiliation(s)
- Dean G Brown
- Jnana Therapeutics, 6 Tide St, MA 02210, United States.
| | - Heike J Wobst
- Jnana Therapeutics, 6 Tide St, MA 02210, United States
| |
Collapse
|
28
|
Rana S, Mallareddy JR, Singh S, Boghean L, Natarajan A. Inhibitors, PROTACs and Molecular Glues as Diverse Therapeutic Modalities to Target Cyclin-Dependent Kinase. Cancers (Basel) 2021; 13:5506. [PMID: 34771669 PMCID: PMC8583118 DOI: 10.3390/cancers13215506] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/27/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
The cyclin-dependent kinase (CDK) family of proteins play prominent roles in transcription, mRNA processing, and cell cycle regulation, making them attractive cancer targets. Palbociclib was the first FDA-approved CDK inhibitor that non-selectively targets the ATP binding sites of CDK4 and CDK6. In this review, we will briefly inventory CDK inhibitors that are either part of over 30 active clinical trials or recruiting patients. The lack of selectivity among CDKs and dose-limiting toxicities are major challenges associated with the development of CDK inhibitors. Proteolysis Targeting Chimeras (PROTACs) and Molecular Glues have emerged as alternative therapeutic modalities to target proteins. PROTACs and Molecular glues utilize the cellular protein degradation machinery to destroy the target protein. PROTACs are heterobifunctional molecules that form a ternary complex with the target protein and E3-ligase by making two distinct small molecule-protein interactions. On the other hand, Molecular glues function by converting the target protein into a "neo-substrate" for an E3 ligase. Unlike small molecule inhibitors, preclinical studies with CDK targeted PROTACs have exhibited improved CDK selectivity. Moreover, the efficacy of PROTACs and molecular glues are not tied to the dose of these molecular entities but to the formation of the ternary complex. Here, we provide an overview of PROTACs and molecular glues that modulate CDK function as emerging therapeutic modalities.
Collapse
Affiliation(s)
- Sandeep Rana
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA;
| | - Jayapal Reddy Mallareddy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (J.R.M.); (S.S.); (L.B.)
| | - Sarbjit Singh
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (J.R.M.); (S.S.); (L.B.)
| | - Lidia Boghean
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (J.R.M.); (S.S.); (L.B.)
| | - Amarnath Natarajan
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA; (J.R.M.); (S.S.); (L.B.)
- Pharmaceutical Sciences and University of Nebraska Medical Center, Omaha, NE 68198, USA
- Genetics Cell Biology and Anatomy, University of Nebraska Medical Center, Omaha, NE 68198, USA
- Fred and Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| |
Collapse
|
29
|
Roth A, Gihring A, Göser F, Peifer C, Knippschild U, Bischof J. Assessing the Inhibitory Potential of Kinase Inhibitors In Vitro: Major Pitfalls and Suggestions for Improving Comparability of Data Using CK1 Inhibitors as an Example. Molecules 2021; 26:4898. [PMID: 34443486 PMCID: PMC8401859 DOI: 10.3390/molecules26164898] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/06/2021] [Accepted: 08/09/2021] [Indexed: 11/16/2022] Open
Abstract
Phosphorylation events catalyzed by protein kinases represent one of the most prevalent as well as important regulatory posttranslational modifications, and dysregulation of protein kinases is associated with the pathogenesis of different diseases. Therefore, interest in developing potent small molecule kinase inhibitors has increased enormously within the last two decades. A critical step in the development of new inhibitors is cell-free in vitro testing with the intention to determine comparable parameters like the commonly used IC50 value. However, values described in the literature are often biased as experimental setups used for determination of kinase activity lack comparability due to different readout parameters, insufficient normalization or the sheer number of experimental approaches. Here, we would like to hold a brief for highly sensitive, radioactive-based in vitro kinase assays especially suitable for kinases exhibiting autophosphorylation activity. Therefore, we demonstrate a systematic workflow for complementing and validating results from high-throughput screening as well as increasing the comparability of enzyme-specific inhibitor parameters for radiometric as well as non-radiometric assays. Using members of the CK1 family of serine/threonine-specific protein kinases and established CK1-specific inhibitors as examples, we clearly demonstrate the power of our proposed workflow, which has the potential to support the generation of more comparable data for biological characterization of kinase inhibitors.
Collapse
Affiliation(s)
- Aileen Roth
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (F.G.); (J.B.)
| | - Adrian Gihring
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (F.G.); (J.B.)
| | - Florian Göser
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (F.G.); (J.B.)
| | - Christian Peifer
- Institute of Pharmacy, Christian-Albrechts-University of Kiel, Gutenbergstraße 76, 24118 Kiel, Germany;
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (F.G.); (J.B.)
| | - Joachim Bischof
- Department of General and Visceral Surgery, Surgery Center, Ulm University Medical Center, Albert-Einstein-Allee 23, 89081 Ulm, Germany; (A.R.); (A.G.); (F.G.); (J.B.)
| |
Collapse
|
30
|
Gilboa T, Ogata AF, Walt D. Single-molecule enzymology for diagnostics: profiling alkaline phosphatase activity in clinical samples. Chembiochem 2021; 23:e202100358. [PMID: 34375495 DOI: 10.1002/cbic.202100358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/06/2021] [Indexed: 11/05/2022]
Abstract
Enzymes can be used as biomarkers for a variety of diseases. However, profiling enzyme activity in clinical samples is challenging due to the heterogeneity in enzyme activity, and the low abundance of the target enzyme in biofluids. Single-molecule methods can overcome these challenges by providing information on the distribution of enzyme activities in a sample. Here, we describe the concept of using the single-molecule enzymology (SME) method to analyze enzymatic activity in clinical samples. We present recent work focused on measuring alkaline phosphatase isotypes in serum samples using SME. Future work will involve improving and simplifying this technology, and applying it to other enzymes for diagnostics.
Collapse
Affiliation(s)
- Tal Gilboa
- Brigham and Women's Hospital, pathology, 60 Fenwood Rd, Bbf-8006, 02115-6195, Boston, UNITED STATES
| | - Alana F Ogata
- Brigham and Women's Hospital, pathology, UNITED STATES
| | - David Walt
- Harvard Medical School, -, -, -, -, UNITED STATES
| |
Collapse
|
31
|
Benn CL, Gibson KR, Reynolds DS. Drugging DNA Damage Repair Pathways for Trinucleotide Repeat Expansion Diseases. J Huntingtons Dis 2021; 10:203-220. [PMID: 32925081 PMCID: PMC7990437 DOI: 10.3233/jhd-200421] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
DNA damage repair (DDR) mechanisms have been implicated in a number of neurodegenerative diseases (both genetically determined and sporadic). Consistent with this, recent genome-wide association studies in Huntington’s disease (HD) and other trinucleotide repeat expansion diseases have highlighted genes involved in DDR mechanisms as modifiers for age of onset, rate of progression and somatic instability. At least some clinical genetic modifiers have been shown to have a role in modulating trinucleotide repeat expansion biology and could therefore provide new disease-modifying therapeutic targets. In this review, we focus on key considerations with respect to drug discovery and development using DDR mechanisms as a target for trinucleotide repeat expansion diseases. Six areas are covered with specific reference to DDR and HD: 1) Target identification and validation; 2) Candidate selection including therapeutic modality and delivery; 3) Target drug exposure with particular focus on blood-brain barrier penetration, engagement and expression of pharmacology; 4) Safety; 5) Preclinical models as predictors of therapeutic efficacy; 6) Clinical outcome measures including biomarkers.
Collapse
Affiliation(s)
- Caroline L Benn
- LoQus23 Therapeutics, Riverside, Babraham Research Campus, Cambridge, UK
| | - Karl R Gibson
- Sandexis Medicinal Chemistry Ltd, Innovation House, Discovery Park, Sandwich, Kent, UK
| | - David S Reynolds
- LoQus23 Therapeutics, Riverside, Babraham Research Campus, Cambridge, UK
| |
Collapse
|