1
|
Veth TS, Kannegieter NM, de Graaf EL, Ruijtenbeek R, Joore J, Ressa A, Altelaar M. Innovative strategies for measuring kinase activity to accelerate the next wave of novel kinase inhibitors. Drug Discov Today 2024; 29:103907. [PMID: 38301799 DOI: 10.1016/j.drudis.2024.103907] [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: 06/23/2023] [Revised: 01/18/2024] [Accepted: 01/25/2024] [Indexed: 02/03/2024]
Abstract
The development of protein kinase inhibitors (PKIs) has gained significance owing to their therapeutic potential for diseases like cancer. In addition, there has been a rise in refining kinase activity assays, each possessing unique biological and analytical characteristics crucial for PKI development. However, the PKI development pipeline experiences high attrition rates and approved PKIs exhibit unexploited potential because of variable patient responses. Enhancing PKI development efficiency involves addressing challenges related to understanding the PKI mechanism of action and employing biomarkers for precision medicine. Selecting appropriate kinase activity assays for these challenges can overcome these attrition rate issues. This review delves into the current obstacles in kinase inhibitor development and elucidates kinase activity assays that can provide solutions.
Collapse
Affiliation(s)
- Tim S Veth
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands; Netherlands Proteomics Center, Padualaan 8, Utrecht 3584 CH, The Netherlands
| | | | - Erik L de Graaf
- Pepscope, Nieuwe Kanaal 7, 6709 PA Wageningen, The Netherlands
| | | | - Jos Joore
- Pepscope, Nieuwe Kanaal 7, 6709 PA Wageningen, The Netherlands
| | - Anna Ressa
- Pepscope, Nieuwe Kanaal 7, 6709 PA Wageningen, The Netherlands
| | - Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, University of Utrecht, Padualaan 8, Utrecht 3584 CH, The Netherlands; Netherlands Proteomics Center, Padualaan 8, Utrecht 3584 CH, The Netherlands.
| |
Collapse
|
2
|
Nowak RP, Jones LH. Target Validation Using PROTACs: Applying the Four Pillars Framework. SLAS DISCOVERY 2020; 26:474-483. [PMID: 33334221 DOI: 10.1177/2472555220979584] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Proteolysis targeting chimeras (PROTACs) are heterobifunctional compounds that recruit the E3 ubiquitin ligase machinery to proteins of interest, resulting in their ubiquitination and subsequent proteasomal degradation. Targeted protein degradation has generated considerable interest in drug discovery because inhibition of one particular function of a protein often does not deliver the therapeutic efficacy that results from whole-protein depletion. However, the physicochemistry and intrinsically complex pharmacology of PROTACs present challenges, particularly for the development of orally bioavailable drugs. Here we describe the application of a translational pharmacology framework (called the four pillars) to expedite PROTAC development by informing pharmacokinetic-pharmacodynamic (PKPD) understanding and helping elucidate structure-activity relationships. Experimental methods are reviewed that help illuminate exposure of the drug or probe at the site of action (pillar 1) and engagement of its target(s) (pillar 2) that drive functional pharmacological effects (pillar 3) resulting in modulation of a relevant phenotype (pillar 4). We hope the guidance will be useful to those developing targeted protein degraders and help establish PROTAC molecules as robust target validation chemical probes.
Collapse
Affiliation(s)
- Radosław P Nowak
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lyn H Jones
- Center for Protein Degradation, Dana-Farber Cancer Institute, Boston, MA, USA
| |
Collapse
|
3
|
Kelly K, West AB. Pharmacodynamic Biomarkers for Emerging LRRK2 Therapeutics. Front Neurosci 2020; 14:807. [PMID: 32903744 PMCID: PMC7438883 DOI: 10.3389/fnins.2020.00807] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 07/10/2020] [Indexed: 12/22/2022] Open
Abstract
Genetic studies have identified variants in the LRRK2 gene as important components of Parkinson's disease (PD) pathobiology. Biochemical and emergent biomarker studies have coalesced around LRRK2 hyperactivation in disease. Therapeutics that diminish LRRK2 activity, either with small molecule kinase inhibitors or anti-sense oligonucleotides, have recently advanced to the clinic. Historically, there have been few successes in the development of therapies that might slow or halt the progression of neurodegenerative diseases. Over the past few decades of biomedical research, retrospective analyses suggest the broad integration of informative biomarkers early in development tends to distinguish successful pipelines from those that fail early. Herein, we discuss the biomarker regulatory process, emerging LRRK2 biomarker candidates, assays, underlying biomarker biology, and clinical integration.
Collapse
Affiliation(s)
- Kaela Kelly
- Duke Center for Neurodegeneration Research, Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC, United States
| | - Andrew B West
- Duke Center for Neurodegeneration Research, Departments of Pharmacology and Cancer Biology, Neurology, and Neurobiology, Duke University, Durham, NC, United States
| |
Collapse
|
4
|
Peters SA, Petersson C, Blaukat A, Halle JP, Dolgos H. Prediction of active human dose: learnings from 20 years of Merck KGaA experience, illustrated by case studies. Drug Discov Today 2020; 25:909-919. [PMID: 31981792 DOI: 10.1016/j.drudis.2020.01.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Revised: 12/24/2019] [Accepted: 01/15/2020] [Indexed: 12/12/2022]
Abstract
High-quality dose predictions based on a good understanding of target engagement is one of the main translational goals in drug development. Here, we systematically evaluate active human dose predictions for 15 Merck KGaA/EMD Serono assets spanning several modalities and therapeutic areas. Using case studies, we illustrate the value of adhering to the translational best practices of having an exposure-response relationship in an appropriate animal model; having validated, translatable pharmacodynamic (PD) biomarkers measurable in Phase I populations in the right tissue; having a deeper understanding of biology; and capturing uncertainties in predictions. Given the gap in publications on the subject, we believe that the learnings from this unique diverse data set, which are generic to the industry, will trigger actions to improve future predictions.
Collapse
Affiliation(s)
- Sheila Annie Peters
- Translational Quantitative Pharmacology, Translational Medicine, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany.
| | - Carl Petersson
- Drug Metabolism and Disposition, Discovery Technology, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Andree Blaukat
- Translational Innovation Platform Oncology, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Joern-Peter Halle
- Translational Innovation Platform Immuno Oncology, Biopharma, Global R&D, Merck Healthcare, Frankfurter Str. 250, 64293 Darmstadt, Germany
| | - Hugues Dolgos
- Biopharmacy Center of Excellence, Servier RD, Suresnes, 92150, France
| |
Collapse
|
5
|
Targeting PDK1 for Chemosensitization of Cancer Cells. Cancers (Basel) 2017; 9:cancers9100140. [PMID: 29064423 PMCID: PMC5664079 DOI: 10.3390/cancers9100140] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 01/01/2023] Open
Abstract
Despite the rapid development in the field of oncology, cancer remains the second cause of mortality worldwide, with the number of new cases expected to more than double in the coming years. Chemotherapy is widely used to decelerate or stop tumour development in combination with surgery or radiation therapy when appropriate, and in many cases this improves the symptomatology of the disease. Unfortunately though, chemotherapy is not applicable to all patients and even when it is, there are many cases where a successful initial treatment period is followed by chemotherapeutic drug resistance. This is caused by a number of reasons, ranging from the genetic background of the patient (innate resistance) to the formation of tumour-initiating cells (acquired resistance). In this review, we discuss the potential role of PDK1 in the development of chemoresistance in different types of malignancy, and the design and application of potent inhibitors which can promote chemosensitization.
Collapse
|
6
|
Ciccimaro E, Zhu Y, Ostanin D, Suchard S, MacGuire J, Xiao Q, Dongre A, Chimalakonda A, Olah T, Shipkova P. Antibody Drug-Target Engagement Measurement in Tissue Using Quantitative Affinity Extraction Liquid Chromatography–Mass Spectrometry: Method Development and Qualification. Anal Chem 2017; 89:5115-5123. [DOI: 10.1021/acs.analchem.7b00688] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Eugene Ciccimaro
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Yongxin Zhu
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Dmitry Ostanin
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Suzanne Suchard
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Jamus MacGuire
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Qing Xiao
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Ashok Dongre
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | | | - Timothy Olah
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| | - Petia Shipkova
- Bristol-Myers Squibb, Princeton, New Jersey 08543, United States
| |
Collapse
|
7
|
Abstract
Originally thought to be nondruggable, kinases represent attractive drug targets for pharmaceutical companies and academia. To date, there are over 40 kinase inhibitors approved by the US FDA, with 32 of these being small molecules, in addition to the three mammalian target of rapamycin inhibitor macrolides (sirolimus, temsirolimus and everolimus). Despite the rapid development of kinase inhibitors for cancer, presently none of these agents are approved for CNS indications. This mini perspective highlights selected kinase targets for CNS disorders, of which brain-permeable small-molecule inhibitors are reported, with demonstrated preclinical proof-of-concept efficacy. This is followed by a brief discussion on the key challenges of blood–brain barrier penetration and selectivity profiles in developing kinase inhibitors for CNS disorders.
Collapse
|
8
|
Ferguson MA, Sutton RM, Karlsson M, Sjövall F, Becker LB, Berg RA, Margulies SS, Kilbaugh TJ. Increased platelet mitochondrial respiration after cardiac arrest and resuscitation as a potential peripheral biosignature of cerebral bioenergetic dysfunction. J Bioenerg Biomembr 2016; 48:269-79. [PMID: 27020568 DOI: 10.1007/s10863-016-9657-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 03/15/2016] [Indexed: 02/02/2023]
Abstract
UNLABELLED Cardiac arrest (CA) results in a sepsis-like syndrome with activation of the innate immune system and increased mitochondrial bioenergetics. OBJECTIVE To determine if platelet mitochondrial respiration increases following CA in a porcine pediatric model of asphyxia-associated ventricular fibrillation (VF) CA, and if this readily obtained biomarker is associated with decreased brain mitochondrial respiration. CA protocol: 7 min of asphyxia, followed by VF, protocolized titration of compression depth to systolic blood pressure of 90 mmHg and vasopressor administration to a coronary perfusion pressure greater than 20 mmHg. PRIMARY OUTCOME platelet integrated mitochondrial electron transport system (ETS) function evaluated pre- and post-CA/ROSC four hours after return of spontaneous circulation (ROSC). Secondary outcome: correlation of platelet mitochondrial bioenergetics to cerebral bioenergetic function. Platelet maximal oxidative phosphorylation (OXPHOSCI+CII), P < 0.02, and maximal respiratory capacity (ETSCI+CII), P < 0.04, were both significantly increased compared to pre-arrest values. This was primarily due to a significant increase in succinate-supported respiration through Complex II (OXPHOSCII, P < 0.02 and ETSCII, P < 0.03). Higher respiration was not due to uncoupling, as the LEAKCI + CII respiration (mitochondrial respiration independent of ATP-production) was unchanged after CA/ROSC. Larger increases in platelet mitochondrial respiratory control ratio (RCR) compared to pre-CA RCR were significantly correlated with lower RCRs in the cortex (P < 0.03) and hippocampus (P < 0.04) compared to sham respiration. Platelet mitochondrial respiration is significantly increased four hours after ROSC. Future studies will identify mechanistic relationships between this serum biomarker and altered cerebral bioenergetics function following cardiac arrest.
Collapse
Affiliation(s)
- Michael A Ferguson
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Robert M Sutton
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Michael Karlsson
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84, Lund, Sweden
| | - Fredrik Sjövall
- Mitochondrial Medicine, Department of Clinical Sciences, Lund University, BMC A13, SE-221 84, Lund, Sweden
| | - Lance B Becker
- Department of Emergency Medicine, Perelman School of Medicine at the University of Pennsylvania, The Hospital of the University of Pennsylvania, 3400 Spruce Street, Philadelphia, PA, 19104, USA
| | - Robert A Berg
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA
| | - Susan S Margulies
- School of Engineering and Applied Science, Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, PA, 19104, USA
| | - Todd J Kilbaugh
- Anesthesiology and Critical Care Medicine, The Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, 34th & Civic Center Blvd., Philadelphia, PA, 19104, USA.
| |
Collapse
|
9
|
Klumpp M. Non-stoichiometric inhibition in integrated lead finding - a literature review. Expert Opin Drug Discov 2015; 11:149-62. [PMID: 26653534 DOI: 10.1517/17460441.2016.1128892] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION Non-stoichiometric inhibition summarizes different mechanisms by which low-molecular weight compounds can reproducibly inhibit high-throughput screening (HTS) and other lead finding assays without binding to a structurally defined site on their molecular target. This disqualifies such molecules from optimization by medicinal chemistry, and therefore their rapid elimination from screening hit lists is essential for productive and effective drug discovery. AREAS COVERED This review covers recent literature that either investigates the various mechanisms behind non-stoichiometric inhibition or suggests assays and readouts to identify them. In addition, combination of the various methods to distill promising molecules out of raw primary hit lists step-by-step is considered. Emerging technologies to demonstrate target engagement in cells are also discussed. EXPERT OPINION Over the last few years, awareness of non-stoichiometric inhibitors within screening libraries and HTS hit lists has considerably increased, not only in the pharmaceutical industry but also in the academic drug discovery community. This has resulted in a variety of methods to detect and handle such compounds. These range from in silico approaches to flag suspicious compounds, and counterassays to measure non-stoichiometric inhibition, to biophysical methods that positively demonstrate stoichiometric binding. In addition, novel technologies to verify target engagement within cells are becoming available. While still a time- and resource-consuming nuisance, non-stoichiometric inhibitors therefore do not fundamentally jeopardize the discovery of low molecular weight lead and drug candidates. Rather, they should be viewed as a manageable issue that with appropriate expertise can be overcome through integration of the above-mentioned approaches.
Collapse
Affiliation(s)
- Martin Klumpp
- a Novartis Institute of Biomedical Research Basel, Novartis Pharma AG , Basel , Switzerland
| |
Collapse
|
10
|
Vinegoni C, Dubach JM, Thurber GM, Miller MA, Mazitschek R, Weissleder R. Advances in measuring single-cell pharmacology in vivo. Drug Discov Today 2015; 20:1087-92. [PMID: 26024776 DOI: 10.1016/j.drudis.2015.05.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Revised: 03/10/2015] [Accepted: 05/20/2015] [Indexed: 11/26/2022]
Abstract
Measuring key pharmacokinetic and pharmacodynamic parameters in vivo at the single cell level is likely to enhance drug discovery and development. In this review, we summarize recent advances in this field and highlight current and future capabilities.
Collapse
Affiliation(s)
- Claudio Vinegoni
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA.
| | - J Matthew Dubach
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | - Greg M Thurber
- Department of Chemical Engineering, Department of Biomedical Engineering, University of Michigan, 2300 Hayward Avenue, Ann Arbor, MI 48109, USA
| | - Miles A Miller
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | - Ralph Mazitschek
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| | - Ralph Weissleder
- Center for System Biology, Massachusetts General Hospital and Harvard Medical School, Richard B. Simches Research Center, 185 Cambridge Street, Boston, MA 02114, USA
| |
Collapse
|
11
|
Dittmar G, Selbach M. SILAC for biomarker discovery. Proteomics Clin Appl 2015; 9:301-6. [PMID: 25504673 DOI: 10.1002/prca.201400112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/05/2014] [Accepted: 12/10/2014] [Indexed: 12/22/2022]
Abstract
SILAC has been employed in MS-based proteomics for nearly a decade. This method is based on cells in culture metabolically incorporating isotope-coded essential amino acids and allows the quantification of global protein populations to identify characteristic changes. Variations of this technique developed over the years allow the application of SILAC not only to cell culture derived samples but also to tissues and human specimens, making this powerful technique amenable to clinically relevant samples. In this review, we provide an overview of different SILAC-derived methods and their use in the identification and development of biomarkers.
Collapse
Affiliation(s)
- Gunnar Dittmar
- Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | | |
Collapse
|
12
|
Pharmacokinetics, pharmacodynamics, and immunogenicity of belatacept in adult kidney transplant recipients. Clin Drug Investig 2014; 34:117-26. [PMID: 24217983 PMCID: PMC3899455 DOI: 10.1007/s40261-013-0153-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
BACKGROUND AND OBJECTIVES Belatacept is a first-in-class, selective co-stimulation blocker recently approved for the prophylaxis of organ rejection in adult kidney transplant recipients. The objective of this study was to report the pharmacokinetics, pharmacodynamics, and immunogenicity of belatacept. METHODS The pharmacokinetics, pharmacodynamics (CD86 receptor occupancy), and immunogenicity of belatacept were studied in de novo adult kidney transplant recipients in phase II and III clinical studies. RESULTS Following multiple doses of 5 or 10 mg/kg, the geometric mean (percentage coefficient of variation) maximum serum concentration and area under the serum concentration-time curve over one dosing interval of belatacept were 136 (20%) and 238 (27%) μg/mL, and 13,587 (27%) and 21,241 (35%) μg·h/mL, respectively. The median belatacept elimination half-life was 8-9 days. Belatacept exhibited concentration-dependent binding to CD86 receptors. The pre-dose CD86 receptor occupancy by belatacept decreased from 94 to 65% between day 5 and 1 year post-transplant, with corresponding pre-dose trough serum concentrations of belatacept decreasing from ~35 to 4 μg/mL during this period. The cumulative incidence of developing anti-belatacept antibodies was 5.3% up to 3 years post-transplant and had no impact on belatacept exposure. CONCLUSIONS Belatacept in adult kidney transplant demonstrated linear pharmacokinetics with low variability, concentration-dependent pharmacodynamics, and a low incidence of anti-drug antibodies.
Collapse
|
13
|
Pedraza CE, Taylor C, Pereira A, Seng M, Tham CS, Izrael M, Webb M. Induction of oligodendrocyte differentiation and in vitro myelination by inhibition of rho-associated kinase. ASN Neuro 2014; 6:6/4/1759091414538134. [PMID: 25289646 PMCID: PMC4189421 DOI: 10.1177/1759091414538134] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
In inflammatory demyelinating diseases such as multiple sclerosis (MS), myelin
degradation results in loss of axonal function and eventual axonal degeneration.
Differentiation of resident oligodendrocyte precursor cells (OPCs) leading to
remyelination of denuded axons occurs regularly in early stages of MS but halts as
the pathology transitions into progressive MS. Pharmacological potentiation of
endogenous OPC maturation and remyelination is now recognized as a promising
therapeutic approach for MS. In this study, we analyzed the effects of modulating the
Rho-A/Rho-associated kinase (ROCK) signaling pathway, by the use of selective
inhibitors of ROCK, on the transformation of OPCs into mature, myelinating
oligodendrocytes. Here we demonstrate, with the use of cellular cultures from rodent
and human origin, that ROCK inhibition in OPCs results in a significant generation of
branches and cell processes in early differentiation stages, followed by accelerated
production of myelin protein as an indication of advanced maturation. Furthermore,
inhibition of ROCK enhanced myelin formation in cocultures of human OPCs and neurons
and remyelination in rat cerebellar tissue explants previously demyelinated with
lysolecithin. Our findings indicate that by direct inhibition of this signaling
molecule, the OPC differentiation program is activated resulting in morphological and
functional cell maturation, myelin formation, and regeneration. Altogether, we show
evidence of modulation of the Rho-A/ROCK signaling pathway as a viable target for the
induction of remyelination in demyelinating pathologies.
Collapse
Affiliation(s)
- Carlos E Pedraza
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | | | - Albertina Pereira
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Michelle Seng
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | - Chui-Se Tham
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| | | | - Michael Webb
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA
| |
Collapse
|
14
|
Quantitative phosphoproteomic analysis reveals system-wide signaling pathways downstream of SDF-1/CXCR4 in breast cancer stem cells. Proc Natl Acad Sci U S A 2014; 111:E2182-90. [PMID: 24782546 DOI: 10.1073/pnas.1404943111] [Citation(s) in RCA: 93] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Breast cancer is the leading cause of cancer-related mortality in women worldwide, with an estimated 1.7 million new cases and 522,000 deaths around the world in 2012 alone. Cancer stem cells (CSCs) are essential for tumor reoccurrence and metastasis which is the major source of cancer lethality. G protein-coupled receptor chemokine (C-X-C motif) receptor 4 (CXCR4) is critical for tumor metastasis. However, stromal cell-derived factor 1 (SDF-1)/CXCR4-mediated signaling pathways in breast CSCs are largely unknown. Using isotope reductive dimethylation and large-scale MS-based quantitative phosphoproteome analysis, we examined protein phosphorylation induced by SDF-1/CXCR4 signaling in breast CSCs. We quantified more than 11,000 phosphorylation sites in 2,500 phosphoproteins. Of these phosphosites, 87% were statistically unchanged in abundance in response to SDF-1/CXCR4 stimulation. In contrast, 545 phosphosites in 266 phosphoproteins were significantly increased, whereas 113 phosphosites in 74 phosphoproteins were significantly decreased. SDF-1/CXCR4 increases phosphorylation in 60 cell migration- and invasion-related proteins, of them 43 (>70%) phosphoproteins are unrecognized. In addition, SDF-1/CXCR4 upregulates the phosphorylation of 44 previously uncharacterized kinases, 8 phosphatases, and 1 endogenous phosphatase inhibitor. Using computational approaches, we performed system-based analyses examining SDF-1/CXCR4-mediated phosphoproteome, including construction of kinase-substrate network and feedback regulation loops downstream of SDF-1/CXCR4 signaling in breast CSCs. We identified a previously unidentified SDF-1/CXCR4-PKA-MAP2K2-ERK signaling pathway and demonstrated the feedback regulation on MEK, ERK1/2, δ-catenin, and PPP1Cα in SDF-1/CXCR4 signaling in breast CSCs. This study gives a system-wide view of phosphorylation events downstream of SDF-1/CXCR4 signaling in breast CSCs, providing a resource for the study of CSC-targeted cancer therapy.
Collapse
|
15
|
Munteanu B, Meyer B, von Reitzenstein C, Burgermeister E, Bog S, Pahl A, Ebert MP, Hopf C. Label-Free in Situ Monitoring of Histone Deacetylase Drug Target Engagement by Matrix-Assisted Laser Desorption Ionization-Mass Spectrometry Biotyping and Imaging. Anal Chem 2014; 86:4642-7. [DOI: 10.1021/ac500038j] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
| | | | | | - Elke Burgermeister
- Department
of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | - Susanne Bog
- Heidelberg Pharma
GmbH, Schriesheimer Strasse 101, 68526 Ladenburg, Germany
| | - Andreas Pahl
- Heidelberg Pharma
GmbH, Schriesheimer Strasse 101, 68526 Ladenburg, Germany
| | - Matthias P. Ebert
- Department
of Medicine II, Medical Faculty Mannheim, University of Heidelberg, Theodor-Kutzer-Ufer 1-3, 68167 Mannheim, Germany
| | | |
Collapse
|
16
|
Anderson D, Kodukula K. Biomarkers in pharmacology and drug discovery. Biochem Pharmacol 2014; 87:172-88. [DOI: 10.1016/j.bcp.2013.08.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2013] [Accepted: 08/19/2013] [Indexed: 12/21/2022]
|
17
|
Morris MK, Chi A, Melas IN, Alexopoulos LG. Phosphoproteomics in drug discovery. Drug Discov Today 2013; 19:425-32. [PMID: 24141136 DOI: 10.1016/j.drudis.2013.10.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 09/05/2013] [Accepted: 10/10/2013] [Indexed: 12/20/2022]
Abstract
Several important aspects of the drug discovery process, including target identification, mechanism of action determination and biomarker identification as well as drug repositioning, require complete understanding of the effects of drugs on protein phosphorylation in relevant biological systems. Novel high-throughput phosphoproteomic technologies can be employed to measure these phosphorylation events. In this review, we describe the advantages and limitations of state-of-the-art phosphoproteomic approaches such as mass spectrometry and antibody-based technologies in terms of sample and data throughput as well as data quality. We then discuss how datasets from each technology can be analyzed and how the results can be and have been applied to advance different aspects of the drug discovery process.
Collapse
Affiliation(s)
| | - An Chi
- Merck & Co., Boston, MA, USA
| | - Ioannis N Melas
- ProtATonce Ltd, Athens, Greece; Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece
| | - Leonidas G Alexopoulos
- ProtATonce Ltd, Athens, Greece; Department of Mechanical Engineering, National Technical University of Athens, Athens, Greece.
| |
Collapse
|
18
|
Animal models of human disease: challenges in enabling translation. Biochem Pharmacol 2013; 87:162-71. [PMID: 23954708 DOI: 10.1016/j.bcp.2013.08.006] [Citation(s) in RCA: 327] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 02/07/2023]
Abstract
Animal models have historically played a critical role in the exploration and characterization of disease pathophysiology, target identification, and in the in vivo evaluation of novel therapeutic agents and treatments. In the wake of numerous clinical trial failures of new chemical entities (NCEs) with promising preclinical profiles, animal models in all therapeutic areas have been increasingly criticized for their limited ability to predict NCE efficacy, safety and toxicity in humans. The present review discusses some of the challenges associated with the evaluation and predictive validation of animal models, as well as methodological flaws in both preclinical and clinical study designs that may contribute to the current translational failure rate. The testing of disease hypotheses and NCEs in multiple disease models necessitates evaluation of pharmacokinetic/pharmacodynamic (PK/PD) relationships and the earlier development of validated disease-associated biomarkers to assess target engagement and NCE efficacy. Additionally, the transparent integration of efficacy and safety data derived from animal models into the hierarchical data sets generated preclinically is essential in order to derive a level of predictive utility consistent with the degree of validation and inherent limitations of current animal models. The predictive value of an animal model is thus only as useful as the context in which it is interpreted. Finally, rather than dismissing animal models as not very useful in the drug discovery process, additional resources, like those successfully used in the preclinical PK assessment used for the selection of lead NCEs, must be focused on improving existing and developing new animal models.
Collapse
|
19
|
Lauber MA, Koza SM, McCall SA, Alden BA, Iraneta PC, Fountain KJ. High-Resolution Peptide Mapping Separations with MS-Friendly Mobile Phases and Charge-Surface-Modified C18. Anal Chem 2013; 85:6936-44. [DOI: 10.1021/ac401481z] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Matthew A. Lauber
- Waters Corporation, 34 Maple
Street, Milford, Massachusetts 01757-3696, United States
| | - Stephan M. Koza
- Waters Corporation, 34 Maple
Street, Milford, Massachusetts 01757-3696, United States
| | - Scott A. McCall
- Waters Corporation, 34 Maple
Street, Milford, Massachusetts 01757-3696, United States
| | - Bonnie A. Alden
- Waters Corporation, 34 Maple
Street, Milford, Massachusetts 01757-3696, United States
| | - Pamela C. Iraneta
- Waters Corporation, 34 Maple
Street, Milford, Massachusetts 01757-3696, United States
| | - Kenneth J. Fountain
- Waters Corporation, 34 Maple
Street, Milford, Massachusetts 01757-3696, United States
| |
Collapse
|
20
|
|
21
|
Use of PD biomarkers to drive dose selection and early clinical decision making. Bioanalysis 2013; 4:2485-97. [PMID: 23157357 DOI: 10.4155/bio.12.224] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A major challenge facing the development of new therapies is the high level of compound attrition in late-stage clinical studies. A key factor in reducing these unsustainable levels of attrition is the successful evaluation of the level of drug effect on its target pathway in early development, otherwise known as testing the compound mechanism. Incorporation of PD biomarkers into Phase I/II trials to demonstrate compound binding to its molecular target and the subsequent modulation of downstream pathways enables early testing of compound mechanism and provides a data-driven framework for decisions on compound progression. This review will discuss the identification and validation of such 'fit-for-purpose' PD biomarkers, and case studies illustrating their use and value in dose selection and accelerating the clinical development of small-molecule drugs will be described.
Collapse
|
22
|
Slotta-Huspenina J, Berg D, Bauer K, Wolff C, Malinowsky K, Bauer L, Drecoll E, Bettstetter M, Feith M, Walch A, Höfler H, Becker KF, Langer R. Evidence of prognostic relevant expression profiles of heat-shock proteins and glucose-regulated proteins in oesophageal adenocarcinomas. PLoS One 2012; 7:e41420. [PMID: 22911792 PMCID: PMC3404067 DOI: 10.1371/journal.pone.0041420] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 06/21/2012] [Indexed: 02/07/2023] Open
Abstract
A high percentage of oesophageal adenocarcinomas show an aggressive clinical behaviour with a significant resistance to chemotherapy. Heat-shock proteins (HSPs) and glucose-regulated proteins (GRPs) are molecular chaperones that play an important role in tumour biology. Recently, novel therapeutic approaches targeting HSP90/GRP94 have been introduced for treating cancer. We performed a comprehensive investigation of HSP and GRP expression including HSP27, phosphorylated (p)-HSP27((Ser15)), p-HSP27((Ser78)), p-HSP27((Ser82)), HSP60, HSP70, HSP90, GRP78 and GRP94 in 92 primary resected oesophageal adenocarcinomas by using reverse phase protein arrays (RPPA), immunohistochemistry (IHC) and real-time quantitative RT-PCR (qPCR). Results were correlated with pathologic features and survival. HSP/GRP protein and mRNA expression was detected in all tumours at various levels. Unsupervised hierarchical clustering showed two distinct groups of tumours with specific protein expression patterns: The hallmark of the first group was a high expression of p-HSP27((Ser15, Ser78, Ser82)) and low expression of GRP78, GRP94 and HSP60. The second group showed the inverse pattern with low p-HSP27 and high GRP78, GRP94 and HSP60 expression. The clinical outcome for patients from the first group was significantly improved compared to patients from the second group, both in univariate analysis (p = 0.015) and multivariate analysis (p = 0.029). Interestingly, these two groups could not be distinguished by immunohistochemistry or qPCR analysis. In summary, two distinct and prognostic relevant HSP/GRP protein expression patterns in adenocarcinomas of the oesophagus were detected by RPPA. Our approach may be helpful for identifying candidates for specific HSP/GRP-targeted therapies.
Collapse
|