1
|
Lamore SD, Ahlberg E, Boyer S, Lamb ML, Hortigon-Vinagre MP, Rodriguez V, Smith GL, Sagemark J, Carlsson L, Bates SM, Choy AL, Stålring J, Scott CW, Peters MF. Deconvoluting Kinase Inhibitor Induced Cardiotoxicity. Toxicol Sci 2018; 158:213-226. [PMID: 28453775 PMCID: PMC5837613 DOI: 10.1093/toxsci/kfx082] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Many drugs designed to inhibit kinases have their clinical utility limited by cardiotoxicity-related label warnings or prescribing restrictions. While this liability is widely recognized, designing safer kinase inhibitors (KI) requires knowledge of the causative kinase(s). Efforts to unravel the kinases have encountered pharmacology with nearly prohibitive complexity. At therapeutically relevant concentrations, KIs show promiscuity distributed across the kinome. Here, to overcome this complexity, 65 KIs with known kinome-scale polypharmacology profiles were assessed for effects on cardiomyocyte (CM) beating. Changes in human iPSC-CM beat rate and amplitude were measured using label-free cellular impedance. Correlations between beat effects and kinase inhibition profiles were mined by computation analysis (Matthews Correlation Coefficient) to identify associated kinases. Thirty kinases met criteria of having (1) pharmacological inhibition correlated with CM beat changes, (2) expression in both human-induced pluripotent stem cell-derived cardiomyocytes and adult heart tissue, and (3) effects on CM beating following single gene knockdown. A subset of these 30 kinases were selected for mechanistic follow up. Examples of kinases regulating processes spanning the excitation–contraction cascade were identified, including calcium flux (RPS6KA3, IKBKE) and action potential duration (MAP4K2). Finally, a simple model was created to predict functional cardiotoxicity whereby inactivity at three sentinel kinases (RPS6KB1, FAK, STK35) showed exceptional accuracy in vitro and translated to clinical KI safety data. For drug discovery, identifying causative kinases and introducing a predictive model should transform the ability to design safer KI medicines. For cardiovascular biology, discovering kinases previously unrecognized as influencing cardiovascular biology should stimulate investigation of underappreciated signaling pathways.
Collapse
Affiliation(s)
- Sarah D Lamore
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451
| | - Ernst Ahlberg
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, 43153 Mölndal, Sweden
| | - Scott Boyer
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, 43153 Mölndal, Sweden
| | - Michelle L Lamb
- IMED Oncology, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451
| | | | - Victor Rodriguez
- Clyde Bioscience Limited BioCity Scotland, Lanarkshire ML1 5UH, United Kingdom
| | - Godfrey L Smith
- Clyde Bioscience Limited BioCity Scotland, Lanarkshire ML1 5UH, United Kingdom
| | - Johanna Sagemark
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, 43153 Mölndal, Sweden
| | - Lars Carlsson
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, 43153 Mölndal, Sweden
| | - Stephanie M Bates
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, Cambridge Science Park, Cambridge, United Kingdom
| | - Allison L Choy
- Research & Development Information, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451
| | - Jonna Stålring
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, 43153 Mölndal, Sweden
| | - Clay W Scott
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451
| | - Matthew F Peters
- Department of Drug Safety and Metabolism, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451
| |
Collapse
|
2
|
McArthur K, D'Cruz AA, Segal D, Lackovic K, Wilks AF, O'Donnell JA, Nowell CJ, Gerlic M, Huang DCS, Burns CJ, Croker BA. Defining a therapeutic window for kinase inhibitors in leukemia to avoid neutropenia. Oncotarget 2017; 8:57948-57963. [PMID: 28938529 PMCID: PMC5601625 DOI: 10.18632/oncotarget.19678] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 06/09/2017] [Indexed: 11/25/2022] Open
Abstract
Neutropenia represents one of the major dose-limiting toxicities of many current cancer therapies. To circumvent the off-target effects of cytotoxic chemotherapeutics, kinase inhibitors are increasingly being used as an adjunct therapy to target leukemia. In this study, we conducted a screen of leukemic cell lines in parallel with primary neutrophils to identify kinase inhibitors with the capacity to induce apoptosis of myeloid and lymphoid cell lines whilst sparing primary mouse and human neutrophils. We have utilized a high-throughput live cell imaging platform to demonstrate that cytotoxic drugs have limited effects on neutrophil viability but are toxic to hematopoietic progenitor cells, with the exception of the topoisomerase I inhibitor SN-38. The parallel screening of kinase inhibitors revealed that mouse and human neutrophil viability is dependent on cyclin-dependent kinase (CDK) activity but surprisingly only partially dependent on PI3 kinase and JAK/STAT signaling, revealing dominant pathways contributing to neutrophil viability. Mcl-1 haploinsufficiency sensitized neutrophils to CDK inhibition, demonstrating that Mcl-1 is a direct target for CDK inhibitors. This study reveals a therapeutic window for the kinase inhibitors BEZ235, BMS-3, AZD7762, and (R)-BI-2536 to induce apoptosis of leukemia cell lines whilst maintaining immunocompetence and hemostasis.
Collapse
Affiliation(s)
- Kate McArthur
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Akshay A D'Cruz
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - David Segal
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Kurt Lackovic
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Andrew F Wilks
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia
| | - Joanne A O'Donnell
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Cameron J Nowell
- Monash Institute of Pharmaceutical Sciences, Melbourne, VIC, Australia
| | - Motti Gerlic
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Clinical Microbiology and Immunology, Tel Aviv University, Tel Aviv, Israel
| | - David C S Huang
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia
| | - Christopher J Burns
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,School of Chemistry, Bio21, The University of Melbourne, Melbourne, VIC, Australia
| | - Ben A Croker
- Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC, Australia.,Department of Medical Biology, University of Melbourne, Melbourne, VIC, Australia.,Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
3
|
In silico assessment of adverse drug reactions and associated mechanisms. Drug Discov Today 2015; 21:58-71. [PMID: 26272036 DOI: 10.1016/j.drudis.2015.07.018] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/15/2015] [Accepted: 07/31/2015] [Indexed: 12/31/2022]
Abstract
During recent years, various in silico approaches have been developed to estimate chemical and biological drug features, for example chemical fragments, protein targets, pathways, among others, that correlate with adverse drug reactions (ADRs) and explain the associated mechanisms. These features have also been used for the creation of predictive models that enable estimation of ADRs during the early stages of drug development. In this review, we discuss various in silico approaches to predict these features for a certain drug, estimate correlations with ADRs, establish causal relationships between selected features and ADR mechanisms and create corresponding predictive models.
Collapse
|
4
|
Segall MD, Barber C. Addressing toxicity risk when designing and selecting compounds in early drug discovery. Drug Discov Today 2014; 19:688-93. [DOI: 10.1016/j.drudis.2014.01.006] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 12/17/2013] [Accepted: 01/14/2014] [Indexed: 12/15/2022]
|
5
|
Goodfellow VS, Loweth CJ, Ravula SB, Wiemann T, Nguyen T, Xu Y, Todd DE, Sheppard D, Pollack S, Polesskaya O, Marker DF, Dewhurst S, Gelbard HA. Discovery, synthesis, and characterization of an orally bioavailable, brain penetrant inhibitor of mixed lineage kinase 3. J Med Chem 2013; 56:8032-48. [PMID: 24044867 PMCID: PMC4032177 DOI: 10.1021/jm401094t] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Inhibition of mixed lineage kinase 3 (MLK3) is a potential strategy for treatment of Parkinson's disease and HIV-1 associated neurocognitive disorders (HAND), requiring an inhibitor that can achieve significant brain concentration levels. We report here URMC-099 (1) an orally bioavailable (F = 41%), potent (IC50 = 14 nM) MLK3 inhibitor with excellent brain exposure in mouse PK models and minimal interference with key human CYP450 enzymes or hERG channels. The compound inhibits LPS-induced TNFα release in microglial cells, HIV-1 Tat-induced release of cytokines in human monocytes and up-regulation of phospho-JNK in Tat-injected brains of mice. Compound 1 likely functions in HAND preclinical models by inhibiting multiple kinase pathways, including MLK3 and LRRK2 (IC50 = 11 nM). We compare the kinase specificity and BBB penetration of 1 with CEP-1347 (2). Compound 1 is well tolerated, with excellent in vivo activity in HAND models, and is under investigation for further development.
Collapse
Affiliation(s)
| | - Colin J. Loweth
- Califia Bio Inc, 11575 Sorrento Valley Road, San Diego, California
| | | | - Torsten Wiemann
- Califia Bio Inc, 11575 Sorrento Valley Road, San Diego, California
| | - Thong Nguyen
- Califia Bio Inc, 11575 Sorrento Valley Road, San Diego, California
| | | | - Daniel E. Todd
- BioFocus, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, UK
| | - David Sheppard
- BioFocus, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, UK
| | - Scott Pollack
- BioFocus, Chesterford Research Park, Saffron Walden, Essex CB10 1XL, UK
| | - Oksana Polesskaya
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave Rochester, New York
| | - Daniel F. Marker
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave Rochester, New York
| | - Stephen Dewhurst
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave Rochester, New York
| | - Harris A. Gelbard
- University of Rochester Medical Center, School of Medicine and Dentistry, 601 Elmwood Ave Rochester, New York
| |
Collapse
|
6
|
Schulz S, Göring S, Schmidt B, Hopf C. LRRK2 Kinase Inhibitors as New Drugs for Parkinson’s Disease? EMERGING DRUGS AND TARGETS FOR PARKINSON’S DISEASE 2013. [DOI: 10.1039/9781849737357-00266] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
It is a rare event in drug discovery that mutations in a gene associated with the autosomal dominant forms of a disease, for which there is a large unmet medical need, affect a protein that belongs to a major class of drug targets. As a consequence, in recent years leucine‐rich repeat kinase 2 (LRRK2) has emerged as a major target candidate for therapies of Parkinson’s disease, and selective inhibitors of this kinase are being evaluated as possible new drugs for this detrimental disease. In this chapter, we review recent advances in the design of potent and selective LRRK2 inhibitors as well as the availability of models for their pharmacological evaluation. We also touch upon the challenges ahead – for further improvement of small molecule inhibitors and for in vivo pharmacological target validation.
Collapse
Affiliation(s)
- Sandra Schulz
- Instrumental Analysis and Bioanalytics Mannheim University of Applied Sciences Mannheim, Germany
- Center for Applied Research in Biomedical Mass Spectrometry ABIMAS Mannheim University of Applied Sciences Mannheim, Germany
| | - Stefan Göring
- Clemens Schöpf‐Institute of Organic Chemistry and Biochemistry Technische Universität Darmstadt Darmstadt, Germany c.hopf@hs‐mannheim.de;
| | - Boris Schmidt
- Clemens Schöpf‐Institute of Organic Chemistry and Biochemistry Technische Universität Darmstadt Darmstadt, Germany c.hopf@hs‐mannheim.de;
| | - Carsten Hopf
- Instrumental Analysis and Bioanalytics Mannheim University of Applied Sciences Mannheim, Germany
- Center for Applied Research in Biomedical Mass Spectrometry ABIMAS Mannheim University of Applied Sciences Mannheim, Germany
| |
Collapse
|
7
|
Lamore SD, Kamendi HW, Scott CW, Dragan YP, Peters MF. Cellular impedance assays for predictive preclinical drug screening of kinase inhibitor cardiovascular toxicity. Toxicol Sci 2013; 135:402-13. [PMID: 23897988 DOI: 10.1093/toxsci/kft167] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular (CV) toxicity is a leading contributor to drug attrition. Implementing earlier testing has successfully reduced human Ether-à-go-go-Related Gene-related arrhythmias. How- ever, analogous assays targeting functional CV effects remain elusive. Demand to address this gap is particularly acute for kinase inhibitors (KIs) that suffer frequent CV toxicity. The drug class also presents some particularly challenging requirements for assessing functional CV toxicity. Specifically, an assay must sense a downstream response that integrates diverse kinase signaling pathways. In addition, sufficient throughput is essential for handling inherent KI nonselectivity. A new opportunity has emerged with cellular impedance technology, which detects spontaneous beating cardiomyocytes. Impedance assays sense morphology changes downstream of cardiomyocyte contraction. To evaluate cardiomyocyte impedance assays for KI screening, we investigated two distinct KI classes where CV toxicity was discovered late and target risks remain unresolved. Microtubule-associated protein/microtubule affinity regulating kinase (MARK) inhibitors decrease blood pressure in dogs, whereas checkpoint kinase (Chk) inhibitors (AZD7762, SCH900776) exhibit dose-limiting CV toxicities in clinical trials. These in vivo effects manifested in vitro as cardiomyocyte beat cessation. MARK effects were deemed mechanism associated because beat inhibition potencies correlated with kinase inhibition, and gene knockdown and microtubule-targeting agents suppressed beating. MARK inhibitor impedance and kinase potencies aligned with rat blood pressure effects. Chk inhibitor effects were judged off-target because Chk and beat inhibition potencies did not correlate and knockdowns did not alter beating. Taken together, the data demonstrate that cardiomyocyte impedance assays can address three unmet needs-detecting KI functional cardiotoxicity in vitro, determining mechanism of action, and supporting safety structure-activity relationships.
Collapse
Affiliation(s)
- Sarah D Lamore
- Molecular Toxicology,Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Harriet W Kamendi
- Molecular Toxicology and Safety Pharmacology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Clay W Scott
- Molecular Toxicology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Yvonne P Dragan
- Molecular Toxicology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| | - Matthew F Peters
- Molecular Toxicology, Global Safety Assessment, AstraZeneca Pharmaceuticals, Waltham, Massachusetts 02451, USA
| |
Collapse
|
8
|
Pharmacological inhibition of Polo Like Kinase 2 (PLK2) does not cause chromosomal damage or result in the formation of micronuclei. Toxicol Appl Pharmacol 2013; 269:1-7. [DOI: 10.1016/j.taap.2013.02.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 02/19/2013] [Accepted: 02/20/2013] [Indexed: 11/18/2022]
|
9
|
|
10
|
Kramer T, Lo Monte F, Göring S, Okala Amombo GM, Schmidt B. Small molecule kinase inhibitors for LRRK2 and their application to Parkinson's disease models. ACS Chem Neurosci 2012; 3:151-60. [PMID: 22860184 PMCID: PMC3369800 DOI: 10.1021/cn200117j] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2011] [Accepted: 01/18/2012] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder. Several single gene mutations have been linked to this disease. Mutations in the gene encoding leucine-rich repeat kinase 2 (LRRK2) indicate LRRK2 as promising therapeutic target for the treatment of PD. LRRK2 mutations were observed in sporadic as well as familial PD patients and have been investigated intensively. LRRK2 is a large and complex protein, with multiple enzymatic and protein-interaction domains, each of which is effected by mutations. The most common mutation in PD patients is G2019S. Several LRRK2 inhibitors have been reported already, although the crystal structure of LRRK2 has not yet been determined. This review provides a summary of known LRRK2 inhibitors and will discuss recent in vitro and in vivo results of these inhibitors.
Collapse
Affiliation(s)
| | | | - Stefan Göring
- Clemens Schöpf - Institute
of Organic Chemistry
and Biochemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Ghislaine Marlyse Okala Amombo
- Clemens Schöpf - Institute
of Organic Chemistry
and Biochemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| | - Boris Schmidt
- Clemens Schöpf - Institute
of Organic Chemistry
and Biochemistry, Technische Universität Darmstadt, 64287 Darmstadt, Germany
| |
Collapse
|
11
|
Ramsden N, Perrin J, Ren Z, Lee BD, Zinn N, Dawson VL, Tam D, Bova M, Lang M, Drewes G, Bantscheff M, Bard F, Dawson TM, Hopf C. Chemoproteomics-based design of potent LRRK2-selective lead compounds that attenuate Parkinson's disease-related toxicity in human neurons. ACS Chem Biol 2011; 6:1021-8. [PMID: 21812418 DOI: 10.1021/cb2002413] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Leucine-rich repeat kinase-2 (LRRK2) mutations are the most important cause of familial Parkinson's disease, and non-selective inhibitors are protective in rodent disease models. Because of their poor potency and selectivity, the neuroprotective mechanism of these tool compounds has remained elusive so far, and it is still unknown whether selective LRRK2 inhibition can attenuate mutant LRRK2-dependent toxicity in human neurons. Here, we employ a chemoproteomics strategy to identify potent, selective, and metabolically stable LRRK2 inhibitors. We demonstrate that CZC-25146 prevents mutant LRRK2-induced injury of cultured rodent and human neurons with mid-nanomolar potency. These precise chemical probes further validate this emerging therapeutic strategy. They will enable more detailed studies of LRRK2-dependent signaling and pathogenesis and accelerate drug discovery.
Collapse
Affiliation(s)
- Nigel Ramsden
- Cellzome Ltd., Chesterford Research Park, Cambridge CB10 1XL, United Kingdom
| | - Jessica Perrin
- Cellzome AG, Meyerhofstrasse 1, D-69117 Heidelberg, Germany
| | - Zhao Ren
- Elan Corporation PLC, 800 Gateway Boulevard, South San Francisco, California 94080, United States
| | - Byoung Dae Lee
- Institute for Cell Engineering, Neuroregeneration and Stem Cell Programs, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Nico Zinn
- Cellzome AG, Meyerhofstrasse 1, D-69117 Heidelberg, Germany
| | - Valina L. Dawson
- Institute for Cell Engineering, Neuroregeneration and Stem Cell Programs, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Danny Tam
- Elan Corporation PLC, 800 Gateway Boulevard, South San Francisco, California 94080, United States
| | - Michael Bova
- Elan Corporation PLC, 800 Gateway Boulevard, South San Francisco, California 94080, United States
| | - Manja Lang
- Cellzome AG, Meyerhofstrasse 1, D-69117 Heidelberg, Germany
| | - Gerard Drewes
- Cellzome AG, Meyerhofstrasse 1, D-69117 Heidelberg, Germany
| | | | - Frederique Bard
- Elan Corporation PLC, 800 Gateway Boulevard, South San Francisco, California 94080, United States
| | - Ted M. Dawson
- Institute for Cell Engineering, Neuroregeneration and Stem Cell Programs, Johns Hopkins University School of Medicine, 733 N. Broadway, Baltimore, Maryland 21205, United States
| | - Carsten Hopf
- Cellzome AG, Meyerhofstrasse 1, D-69117 Heidelberg, Germany
| |
Collapse
|