1
|
Egner M, Busch R, López-García Ú, Lewandowski M, Höfner G, Wein T, Marschner JA, Merk D. A Nurr1 Agonist Derived from the Natural Ligand DHI Induces Neuroprotective Gene Expression. J Med Chem 2025; 68:4829-4847. [PMID: 39919139 PMCID: PMC7617521 DOI: 10.1021/acs.jmedchem.4c03104] [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] [Indexed: 02/09/2025]
Abstract
The dopamine metabolite 5,6-dihydroxyindole (DHI) has been discovered as a natural Nurr1 ligand with potential biological relevance and is an attractive lead for Nurr1 modulator development but exhibits chemical reactivity and weak potency. We have systematically explored the SAR of 5-chloroindole-6-carboxamide as a DHI mimetic scaffold and identified the first high-affinity (Kd 0.08-0.12 μM) ligands of the DHI binding site of Nurr1. An optimized Nurr1 agonist of this scaffold endowed with favorable physicochemical properties, high selectivity, and low toxicity emerges as a chemical tool to explore the biological impact of Nurr1 activation via the DHI binding site. Treatment of neuronal cells with this compound mediated enhanced expression of Nurr1-regulated neuroprotective genes like brain-derived neurotrophic factor (BDNF), supporting the great potential of Nurr1 activation in neurodegeneration.
Collapse
Affiliation(s)
- Markus Egner
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Romy Busch
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Úrsula López-García
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Max Lewandowski
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Georg Höfner
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Thomas Wein
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Julian A. Marschner
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| | - Daniel Merk
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377Munich, Germany
| |
Collapse
|
2
|
Asvos X, El Mubarak MA, Karampelas T, Rampias T, Tamvakopoulos C, Sivolapenko GB, Papakyriakou A, Topouzis S, Vassilatis DK, Fokas D. BRF110, an Orally Active Nurr1-RXRα-Selective Rexinoid, Enhances BDNF Expression without Elevating Triglycerides. J Med Chem 2025; 68:4763-4786. [PMID: 39945195 PMCID: PMC11874024 DOI: 10.1021/acs.jmedchem.4c03046] [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: 12/11/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/28/2025]
Abstract
We report the discovery of a Nurr1-RXRα heterodimer-selective rexinoid which emerged from the structural modification of aminopyrimidine XCT0135908. Although XCT0135908 demonstrated high selectivity for the Nurr1-RXRα heterodimer over other RXRα dimerization partners, its poor in vivo stability and limited brain penetration hindered its utility. Structure-activity relationship (SAR) studies alongside bioactivity evaluations of a diverse series of substituted pyrimidines led to BRF110, a brain-penetrant compound retaining the selective activation of the Nurr1-RXRα heterodimer. BRF110, as XCT0135908, protects dopaminergic cells against the Parkinson's disease-related toxin MPP+ and increases BDNF transcription in mice. Notably, BRF110, in contrast to the market-approved pan-RXR agonist bexarotene, did not elevate triglyceride levels, indicating that enhanced heterodimer selectivity can mitigate off-target in vivo side effects of rexinoids. These findings highlight the potential of heterodimer-selective scaffolds as a strategy for improving the therapeutic profile of rexinoids, addressing significant challenges in the clinical development of RXR-targeting molecules.
Collapse
Affiliation(s)
- Xenophon Asvos
- Department
of Materials Science and Engineering, University
of Ioannina, Ioannina 45110, Greece
| | | | - Theodoros Karampelas
- Center
for Clinical Research, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Theodoros Rampias
- Center
for Clinical Research, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Constantin Tamvakopoulos
- Center
for Clinical Research, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | | | - Athanasios Papakyriakou
- Institute
of Biosciences and Applications, National
Centre for Scientific Research “Demokritos”, Athens 15341, Greece
| | - Stavros Topouzis
- Department
of Pharmacy, University of Patras, Patras 26504, Greece
| | - Demetrios K. Vassilatis
- Center
for Clinical Research, Experimental Surgery, and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Demosthenes Fokas
- Department
of Materials Science and Engineering, University
of Ioannina, Ioannina 45110, Greece
| |
Collapse
|
3
|
Schallmayer E, Isigkeit L, Elson L, Müller S, Knapp S, Marschner JA, Merk D. Chemogenomics for steroid hormone receptors (NR3). Commun Chem 2025; 8:29. [PMID: 39900826 PMCID: PMC11790914 DOI: 10.1038/s42004-025-01427-z] [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: 09/30/2024] [Accepted: 01/23/2025] [Indexed: 02/05/2025] Open
Abstract
The nine human NR3 nuclear receptors translate steroid hormone signals in transcriptomic responses and operate multiple highly important processes ranging from development over reproductive tissue function to inflammatory and metabolic homeostasis. Although several NR3 ligands such as glucocorticoids are invaluable drugs, this family is only partially explored, for example, in autoimmune diseases and neurodegeneration, but may hold therapeutic potential in new areas. Here we report a chemogenomics (CG) library to reveal elusive effects of NR3 receptor modulation in phenotypic settings. 34 highly annotated and chemically diverse ligands covering all NR3 receptors were selected considering complementary modes of action and activity, selectivity and lack of toxicity. Endoplasmic reticulum stress resolving effects of N3 CG subsets in proof-of-concept application validate suitability of the set to connect phenotypic outcomes with targets and to explore NR3 receptors from a translational perspective.
Collapse
Affiliation(s)
- Espen Schallmayer
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Laura Isigkeit
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Lewis Elson
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany
| | - Julian A Marschner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe-Universität Frankfurt, Frankfurt, Germany.
- Department of Pharmacy, Ludwig-Maximilians-Universität München, Munich, Germany.
| |
Collapse
|
4
|
Cordsmeier A, Herrmann A, Gege C, Kohlhof H, Korn K, Ensser A. Molecular analysis of the 2022 mpox outbreak and antiviral activity of dihydroorotate dehydrogenase inhibitors against orthopoxviruses. Antiviral Res 2025; 233:106043. [PMID: 39608644 DOI: 10.1016/j.antiviral.2024.106043] [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: 06/10/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 11/30/2024]
Abstract
Monkeypox virus (MPXV) has caused a large pandemic outbreak in 2022 with more than 90.000 confirmed cases and 181 deaths. Notably, signs of microevolution and host adaption have been observed. Here, we demonstrate that viral genomes from Franconia, Bavaria acquired different mutations. Three isolates obtained from diagnostic samples, submitted from suspected Mpox cases, show differences in their replication capacities. One MPXV isolate which shows the fastest replication kinetics and higher viral loads, possesses a unique non-synonymous mutation (D616L) in the A11L protein (gene OPG136), which encodes for a protein that is part of a major viral core structure. In regard to pandemic preparedness and future outbreaks, we analyzed the antiviral activity of dihydroorotate dehydrogenase (DHODH) inhibitors, and show that they are active against MPXV, vaccinia virus (VACV), and cowpox virus (CPXV) and therefore likely against orthopoxviruses in general. In agreement with that, we also demonstrated that chemical optimization leads to compounds with EC50 values in the sub-nanomolar range, associated with low cytotoxicity, which forms a good basis for future drug development from this chemical series.
Collapse
Affiliation(s)
- Arne Cordsmeier
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | | | - Christian Gege
- Immunic AG, Lochhamer Schlag 21, 82166 Gräfelfing, Germany
| | - Hella Kohlhof
- Immunic AG, Lochhamer Schlag 21, 82166 Gräfelfing, Germany
| | - Klaus Korn
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany
| | - Armin Ensser
- Institute of Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
5
|
Häusler D, Weber MS. Towards Treating Multiple Sclerosis Progression. Pharmaceuticals (Basel) 2024; 17:1474. [PMID: 39598386 PMCID: PMC11597358 DOI: 10.3390/ph17111474] [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: 10/08/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/29/2024] Open
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating disease of the central nervous system (CNS). In most patients, the disease starts with an acute onset followed by a remission phase, subsequent relapses and a later transition to steady chronic progression. In a minority of patients, this progressive phase develops from the beginning. MS relapses are characterized predominantly by the de novo formation of an inflammatory CNS lesion and the infiltration of immune cells, whereas the pathological features of MS progression include slowly expanding lesions, global brain atrophy and an inflammatory response predominantly mediated by macrophages/microglia. Importantly, this CNS-intrinsic pathophysiology appears to initiate early during the relapsing-remitting disease phase, while it turns into the key clinical MS feature in later stages. Currently approved disease-modifying treatments for MS are effective in modulating peripheral immunity by dampening immune cell activity or preventing the migration of immune cells into the CNS, resulting in the prevention of relapses; however, they show limited success in halting MS progression. In this manuscript, we first describe the pathological mechanisms of MS and summarize the approved therapeutics for MS progression. We also review the treatment options for progressive MS (PMS) that are currently under investigation. Finally, we discuss potential targets for novel treatment strategies in PMS.
Collapse
Affiliation(s)
- Darius Häusler
- Institute of Neuropathology, University Medical Centre, 37075 Goettingen, Germany;
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, 37075 Goettingen, Germany
| | - Martin S. Weber
- Institute of Neuropathology, University Medical Centre, 37075 Goettingen, Germany;
- Fraunhofer-Institute for Translational Medicine and Pharmacology ITMP, 37075 Goettingen, Germany
- Department of Neurology, University Medical Centre, 37075 Goettingen, Germany
| |
Collapse
|
6
|
Herrmann A, Gege C, Wangen C, Wagner S, Kögler M, Cordsmeier A, Irrgang P, Ip WH, Weil T, Hunszinger V, Groß R, Heinen N, Pfaender S, Reuter S, Klopfleisch R, Uhlig N, Eberlein V, Issmail L, Grunwald T, Hietel B, Cynis H, Münch J, Sparrer KMJ, Ensser A, Tenbusch M, Dobner T, Vitt D, Kohlhof H, Hahn F. Orally bioavailable RORγ/DHODH dual host-targeting small molecules with broad-spectrum antiviral activity. Antiviral Res 2024; 231:106008. [PMID: 39306285 DOI: 10.1016/j.antiviral.2024.106008] [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: 07/19/2024] [Revised: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
Host-directed antivirals (HDAs) represent an attractive treatment option and a strategy for pandemic preparedness, especially due to their potential broad-spectrum antiviral activity and high barrier to resistance development. Particularly, dual-targeting HDAs offer a promising approach for antiviral therapy by simultaneously disrupting multiple pathways essential for viral replication. Izumerogant (IMU-935) targets two host proteins, (i) the retinoic acid receptor-related orphan receptor γ isoform 1 (RORγ1), which modulates cellular cholesterol metabolism, and (ii) the enzyme dihydroorotate dehydrogenase (DHODH), which is involved in de novo pyrimidine synthesis. Here, we synthesized optimized derivatives of izumerogant and characterized their antiviral activity in comparison to a recently described structurally distinct RORγ/DHODH dual inhibitor. Cell culture-based infection models for enveloped and non-enveloped DNA and RNA viruses, as well as a retrovirus, demonstrated high potency and broad-spectrum activity against human viral pathogens for RORγ/DHODH dual inhibitors at nanomolar concentrations. Comparative analyses with equipotent single-target inhibitors in metabolite supplementation approaches revealed that the dual-targeting mode represents the mechanistic basis for the potent antiviral activity. For SARS-CoV-2, an optimized dual inhibitor completely blocked viral replication in human airway epithelial cells at 5 nM and displayed a synergistic drug interaction with the nucleoside analog molnupiravir. In a SARS-CoV-2 mouse model, treatment with a dual inhibitor alone, or in combination with molnupiravir, reduced the viral load by 7- and 58-fold, respectively. Considering the clinical safety, oral bioavailability, and tolerability of izumerogant in a recent Phase I study, izumerogant-like drugs represent potent dual-targeting antiviral HDAs with pronounced broad-spectrum activity for further clinical development.
Collapse
Affiliation(s)
| | | | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Sabrina Wagner
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Melanie Kögler
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Arne Cordsmeier
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Pascal Irrgang
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Wing-Hang Ip
- Leibniz Institute of Virology, Hamburg, Germany.
| | - Tatjana Weil
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Victoria Hunszinger
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Rüdiger Groß
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | - Natalie Heinen
- Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, Germany.
| | - Stephanie Pfaender
- Leibniz Institute of Virology, Hamburg, Germany; Ruhr-University Bochum, Department of Molecular and Medical Virology, Bochum, Germany; University of Luebeck, Department of Natural Sciences, Institute of Virology and Cell Biology, Lübeck, Germany.
| | - Sebastian Reuter
- University Hospital Essen - Ruhrlandklinik, Department of Pulmonary Medicine, Experimental Pneumology, Essen, Germany.
| | - Robert Klopfleisch
- Institute for Animal Pathology, Freie Universität Berlin, Berlin, Germany.
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy and Immunology, Preclinical Validation, Leipzig, Germany.
| | - Benjamin Hietel
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle, Germany.
| | - Holger Cynis
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Drug Design and Target Validation, Halle, Germany; Junior Research Group "Immunomodulation in Pathophysiological Processes", Faculty of Medicine, Martin Luther University Halle-Wittenberg, Halle, Germany.
| | - Jan Münch
- Institute of Molecular Virology, Ulm University Medical Center, Ulm, Germany.
| | | | - Armin Ensser
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | - Matthias Tenbusch
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| | | | | | | | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany.
| |
Collapse
|
7
|
Sai M, van Herwijnen N, Merk D. Azologs of the Fatty Acid Mimetic Drug Cinalukast Enable Light-Induced PPARα Activation. ChemMedChem 2024; 19:e202400327. [PMID: 38895744 DOI: 10.1002/cmdc.202400327] [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: 04/30/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 06/21/2024]
Abstract
Photo-switchable nuclear receptor modulators ("photohormones") enable spatial and temporal control over transcription factor activity and are valuable precision tools for biological studies. We have developed a new photohormone chemotype by incorporating a light-switchable motif in the scaffold of a cinalukast-derived PPARα ligand and tuned light-controlled activity by systematic structural variation. An optimized photohormone exhibited PPARα agonism in its light-induced (Z)-configuration and strong selectivity over related lipid-activated transcription factors representing a valuable addition to the collection of light-controlled tools to study nuclear receptor activity.
Collapse
Affiliation(s)
- Minh Sai
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Niels van Herwijnen
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Daniel Merk
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| |
Collapse
|
8
|
Gege C, Hahn F, Wangen C, Häge S, Herrmann A, Uhlig N, Eberlein V, Issmail L, Klopfleisch R, Grunwald T, Marschall M, Kohlhof H, Vitt D. Synthesis and Characterization of DHODH Inhibitors Based on the Vidofludimus Scaffold with Pronounced Anti-SARS-CoV-2 Activity. ChemMedChem 2024; 19:e202400292. [PMID: 38887198 DOI: 10.1002/cmdc.202400292] [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: 04/22/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/20/2024]
Abstract
New strategies for the rapid development of broad-spectrum antiviral therapies are urgently required for emerging and re-emerging viruses like the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Host-directed antivirals that target universal cellular metabolic pathways necessary for viral replication present a promising approach with broad-spectrum activity and low potential for development of viral resistance. Dihydroorotate dehydrogenase (DHODH) was identified as one of those universal host factors essential for the replication of many clinically relevant human pathogenic viruses. DHODH is the rate-limiting enzyme catalyzing the fourth step in the de novo pyrimidine synthesis. Therefore, it is also developed as a therapeutic target for many diseases relying on cellular pyrimidine resources, such as cancer, autoimmune diseases and viral or bacterial infection. Thus, several DHODH inhibitors, including vidofludimus calcium (VidoCa, IMU-838), are currently in development or have been investigated in clinical trials for the treatment of virus infections such as SARS-CoV-2-mediated coronavirus disease 19 (COVID-19). Here, we report the medicinal chemistry optimization of VidoCa that resulted in metabolically more stable derivatives with improved DHODH target inhibition in various mammalian species, which translated into improved efficacy against SARS-CoV-2.
Collapse
Affiliation(s)
- Christian Gege
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| | - Friedrich Hahn
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Christina Wangen
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Sigrun Häge
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | | | - Nadja Uhlig
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Valentina Eberlein
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Leila Issmail
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Robert-von-Ostertag-Str. 15, 14163, Berlin, Germany
| | - Thomas Grunwald
- Department of Vaccines and Infection Models, Unit Preclinical Validation, Fraunhofer-Institute for Cell Therapy and Immunology IZI, Perlickstrasse 1, 04103, Leipzig, Germany
| | - Manfred Marschall
- Institute for Clinical and Molecular Virology, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Schlossgarten 4, 91054, Erlangen, Germany
| | - Hella Kohlhof
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| | - Daniel Vitt
- Immunic AG, Lochhamer Schlag 21, 82166, Gräfelfing, Germany
| |
Collapse
|
9
|
Giovannoni G, Hawkes CH, Lechner-Scott J, Levy M, Yeh EA, Pepper G, Schmierer K. Can placebo-controlled phase 2 disease-modifying therapy trials in MS still be justified? Mult Scler Relat Disord 2024; 87:105698. [PMID: 38850685 DOI: 10.1016/j.msard.2024.105698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2024]
Affiliation(s)
- Gavin Giovannoni
- The Blizard Institute, Centre for Neuroscience, Surgery & Trauma, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK.
| | - Christopher H Hawkes
- The Blizard Institute, Centre for Neuroscience, Surgery & Trauma, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK
| | | | - Michael Levy
- Massachusetts General Hospital and Harvard Medical School, Massachusetts, USA
| | - E Ann Yeh
- Department of Paediatrics, Dalla Lana School of Public Health, University of Toronto
| | - George Pepper
- Shift.ms, Platform, New Station Street, LS1 4JB, United Kingdom
| | - Klaus Schmierer
- The Blizard Institute, Centre for Neuroscience, Surgery & Trauma, Faculty of Medicine and Dentistry, Queen Mary University of London, London, UK; Clinical Board Medicine (Neuroscience), The Royal London Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
10
|
Sai M, Hank EC, Tai HM, Kasch T, Lewandowski M, Vincendeau M, Marschner JA, Merk D. Development of Nurr1 agonists from amodiaquine by scaffold hopping and fragment growing. Commun Chem 2024; 7:149. [PMID: 38951694 PMCID: PMC11217349 DOI: 10.1038/s42004-024-01224-0] [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: 12/28/2023] [Accepted: 06/14/2024] [Indexed: 07/03/2024] Open
Abstract
The neuroprotective transcription factor nuclear receptor-related 1 (Nurr1) has shown great promise as a therapeutic target in Parkinson's and Alzheimer's disease as well as multiple sclerosis but high-quality chemical tools for pharmacological target validation of Nurr1 are rare. We have employed the weak Nurr1 modulator amodiaquine (AQ) and AQ-derived fragments as templates to design a new Nurr1 agonist chemotype by scaffold hopping and fragment growing strategies. Systematic structural optimization of this scaffold yielded Nurr1 agonists with nanomolar potency and binding affinity. Comprehensive in vitro profiling revealed efficient cellular target engagement and compliance with the highest probe criteria. In human midbrain organoids bearing a Parkinson-driving LRRK2 mutation, a novel Nurr1 agonist rescued tyrosine hydroxylase expression highlighting the potential of the new Nurr1 modulator chemotype as lead and as a chemical tool for biological studies.
Collapse
Affiliation(s)
- Minh Sai
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Emily C Hank
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Hin-Man Tai
- Helmholtz Munich, Institute of Virology, 85764, Munich, Germany
| | - Till Kasch
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Max Lewandowski
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Michelle Vincendeau
- Helmholtz Munich, Institute of Virology, 85764, Munich, Germany
- Technical University of Munich, Institute of Virology, School of Medicine, 81675, Munich, Germany
| | - Julian A Marschner
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany
| | - Daniel Merk
- Ludwig-Maximilians-Universität München, Department of Pharmacy, 81377, Munich, Germany.
| |
Collapse
|
11
|
Sequeira L, Benfeito S, Fernandes C, Lima I, Peixoto J, Alves C, Machado CS, Gaspar A, Borges F, Chavarria D. Drug Development for Alzheimer's and Parkinson's Disease: Where Do We Go Now? Pharmaceutics 2024; 16:708. [PMID: 38931832 PMCID: PMC11206728 DOI: 10.3390/pharmaceutics16060708] [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: 04/04/2024] [Revised: 05/15/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Neurodegenerative diseases (NDs) are a set of progressive, chronic, and incurable diseases characterized by the gradual loss of neurons, culminating in the decline of cognitive and/or motor functions. Alzheimer's disease (AD) and Parkinson's disease (PD) are the most common NDs and represent an enormous burden both in terms of human suffering and economic cost. The available therapies for AD and PD only provide symptomatic and palliative relief for a limited period and are unable to modify the diseases' progression. Over the last decades, research efforts have been focused on developing new pharmacological treatments for these NDs. However, to date, no breakthrough treatment has been discovered. Hence, the development of disease-modifying drugs able to halt or reverse the progression of NDs remains an unmet clinical need. This review summarizes the major hallmarks of AD and PD and the drugs available for pharmacological treatment. It also sheds light on potential directions that can be pursued to develop new, disease-modifying drugs to treat AD and PD, describing as representative examples some advances in the development of drug candidates targeting oxidative stress and adenosine A2A receptors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fernanda Borges
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Daniel Chavarria
- CIQUP-IMS—Centro de Investigação em Química da Universidade do Porto, Institute of Molecular Sciences, Department of Chemistry and Biochemistry, Faculty of Sciences, University of Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| |
Collapse
|
12
|
Gabaldon-Albero A, Mayo S, Martinez F. NR4A2 as a Novel Target Gene for Developmental and Epileptic Encephalopathy: A Systematic Review of Related Disorders and Therapeutic Strategies. Int J Mol Sci 2024; 25:5198. [PMID: 38791237 PMCID: PMC11120677 DOI: 10.3390/ijms25105198] [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: 04/03/2024] [Revised: 05/04/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
The NR4A2 gene encodes an orphan transcription factor of the steroid-thyroid hormone-retinoid receptor superfamily. This review focuses on the clinical findings associated with the pathogenic variants so far reported, including three unreported cases. Also, its role in neurodegenerative diseases, such as Parkinson's or Alzheimer's disease, is examined, as well as a brief exploration on recent proposals to develop novel therapies for these neurological diseases based on small molecules that could modulate NR4A2 transcriptional activity. The main characteristic shared by all patients is mild to severe developmental delay/intellectual disability. Moderate to severe disorder of the expressive and receptive language is present in at least 42%, while neuro-psychiatric issues were reported in 53% of patients. Movement disorders, including dystonia, chorea or ataxia, are described in 37% patients, although probably underestimated because of its frequent onset in late adolescence-young adulthood. Finally, epilepsy was surprisingly present in 42% of patients, being drug-resistant in three of them. The age at onset varied widely, from five months to twenty-six years, as did the classification of epilepsy, which ranged from focal epilepsy to infantile spasms or Lennox-Gastaut syndrome. Accordingly, we propose that NR4A2 should be considered as a first-tier target gene for the genetic diagnosis of developmental and epileptic encephalopathy.
Collapse
Affiliation(s)
- Alba Gabaldon-Albero
- Translational Research Group in Genetics, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain;
| | - Sonia Mayo
- Genetics and Inheritance Research Group, Instituto de Investigación Sanitaria Hospital 12 de Octubre, 28041 Madrid, Spain
- Department of Genetics, Hospital Universitario 12 de Octubre, 28041 Madrid, Spain
| | - Francisco Martinez
- Translational Research Group in Genetics, Instituto de Investigación Sanitaria La Fe, 46026 Valencia, Spain;
- Genetics Unit, Hospital Universitario y Politecnico La Fe, 46026 Valencia, Spain
| |
Collapse
|
13
|
Fox RJ, Wiendl H, Wolf C, De Stefano N, Sellner J, Gryb V, Rejdak K, Bozhinov PS, Vitt D, Kohlhof H, Slizgi J, Ondrus M, Sciacca V, Muehler AR. Safety and Dose-Response of Vidofludimus Calcium in Relapsing Multiple Sclerosis: Extended Results of a Placebo-Controlled Phase 2 Trial. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200208. [PMID: 38662979 PMCID: PMC11087024 DOI: 10.1212/nxi.0000000000200208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 01/19/2024] [Indexed: 05/12/2024]
Abstract
BACKGROUND AND OBJECTIVES Vidofludimus calcium suppressed MRI disease activity compared with placebo in patients with relapsing-remitting multiple sclerosis (RRMS) in the first cohort of the phase 2 EMPhASIS study. Because 30 mg and 45 mg showed comparable activity on multiple end points, the study enrolled an additional low-dose cohort to further investigate a dose-response relationship. METHODS In a randomized, placebo-controlled, phase 2 trial, patients with RRMS, aged 18-55 years, and with ≥2 relapses in the last 2 years or ≥1 relapse in the last year, and ≥1 gadolinium-enhancing brain lesion in the last 6 months. Patients were randomly assigned (1:1:1) vidofludimus calcium (30 or 45 mg) or placebo in cohort 1 and vidofludimus calcium (10 mg) or placebo (4:1) in cohort 2 for 24 weeks. The primary end point was the cumulative number of combined unique active (CUA) lesions at week 24. Secondary end points were clinical outcomes and safety. RESULTS Across cohorts 1 and 2, 268 patients were randomized to placebo (n = 81), 10 mg (n = 47) vidofludimus calcium, 30 mg (n = 71) vidofludimus calcium, or 45 mg (n = 69) vidofludimus calcium. The mean cumulative CUA lesions over 24 weeks was 5.8 (95% CI 4.1-8.2) for placebo, 5.9 (95% CI 3.9-9.0) for 10 mg treatment group, 1.4 (95% CI 0.9-2.1) for 30 mg treatment group, and 1.7 (95% CI 1.1-2.5) for 45 mg treatment group. Serum neurofilament light chain decreased in a dose-dependent manner. The number of patients with confirmed disability worsening after 24 weeks was 3 (3.7%) patients receiving placebo and 3 (1.6%) patients receiving any dose of vidofludimus calcium. Treatment-emergent adverse events occurred in 35 (43%) placebo patients compared with 11 (23%) and 71 (37%) patients in the 10 mg or any dose of vidofludimus calcium groups, respectively. The incidence of liver enzyme elevations and infections were similar between placebo and any dose of vidofludimus calcium. No new safety signals were observed. DISCUSSION Compared with placebo, vidofludimus calcium suppressed the development of new brain lesions with daily doses of 30 mg and 45 mg, but not 10 mg, establishing the lowest efficacious dose is 30 mg. CLASSIFICATION OF EVIDENCE This study provides Class II evidence that among adults with active RRMS and ≥1 Gd+ brain lesion in the past 6 months, the cumulative number of active lesions decreased with vidofludimus calcium. TRIAL REGISTRATION INFORMATION ClinicalTrials.gov (NCT03846219) and EudraCT (2018-001896-19).
Collapse
Affiliation(s)
- Robert J Fox
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Heinz Wiendl
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Christian Wolf
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Nicola De Stefano
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Johann Sellner
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Viktoriia Gryb
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Konrad Rejdak
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Plamen S Bozhinov
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Daniel Vitt
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Hella Kohlhof
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Jason Slizgi
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Matej Ondrus
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Valentina Sciacca
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| | - Andreas R Muehler
- From the Mellen Center for Multiple Sclerosis (R.J.F.), Cleveland Clinic, OH; Department of Neurology with Institute of Translational Neurology (H.W.), University of Münster, Germany; Lycalis sprl (C.W.), Brussels, Belgium; Department of Medicine (N.D.S.), Surgery and Neuroscience, University of Siena, Italy; Department of Neurology (J. Sellner), Landesklinikum Mistelbach-Gänserndorf, Austria; Regional Clinical Hospital Department of Vascular Neurology (V.G.), Ivano-Frankivsk, Ukraine; Department of Neurology (K.R.), Medical University of Lublin, Lublin, Poland; Medical University of Pleven (P.S.B.), Bulgaria; Immunic AG (D.V., H.K., M.O., V.S., A.R.M.), Gräfelfing, Germany; and independent consultant (J. Slizgi), Raleigh, NC
| |
Collapse
|
14
|
Liu Y, Ma C, Li Y, Li M, Cui T, Zhao X, Li Z, Jia H, Wang H, Xiu X, Hu D, Zhang R, Wang N, Liu P, Yang H, Cheng M. Design, synthesis and biological evaluation of carbamate derivatives incorporating multifunctional carrier scaffolds as pseudo-irreversible cholinesterase inhibitors for the treatment of Alzheimer's disease. Eur J Med Chem 2024; 265:116071. [PMID: 38157596 DOI: 10.1016/j.ejmech.2023.116071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/03/2024]
Abstract
In this study, a series of carbamate derivatives incorporating multifunctional carrier scaffolds were designed, synthesized, and evaluated as potential therapeutic agents for Alzheimer's disease (AD). We used tacrine to modify the aliphatic substituent, and employed rivastigmine, indole and sibiriline fragments as carrier scaffolds. The majority of compounds exhibited good inhibitory activity for cholinesterase. Notably, compound C7 with sibiriline fragment exhibited potent inhibitory activities against human acetylcholinesterase (hAChE, IC50 = 30.35 ± 2.07 nM) and human butyrylcholinesterase (hBuChE, IC50 = 48.03 ± 6.41 nM) with minimal neurotoxicity. Further investigations have demonstrated that C7 exhibited a remarkable capacity to safeguard PC12 cells against H2O2-induced apoptosis and effectively suppressed the production of reactive oxygen species (ROS). Moreover, in an inflammation model of BV2 cells induced by lipopolysaccharide (LPS), C7 effectively attenuated the levels of pro-inflammatory cytokines. After 12 h of dialysis, C7 continued to exhibit an inhibitory effect on cholinesterase activity. An acute toxicity test in vivo demonstrated that C7 exhibited a superior safety profile and no hepatotoxicity compared to the parent nucleus tacrine. In the scopolamine-induced AD mouse model, C7 (20 mg/kg) significantly reduced cholinesterase activity in the brain of the mice. C7 was tested in a pharmacological AD mouse model induced by Aβ1-42 and attenuated memory deficits at doses as low as 5 mg/kg. The pseudo-irreversible cholinesterase inhibitory properties and multifunctional therapeutic attributes of C7 render it a promising candidate for further investigation in the treatment of AD.
Collapse
Affiliation(s)
- Yaoyang Liu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Chao Ma
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Yingbo Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Mengzhen Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Tao Cui
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China
| | - Xueqi Zhao
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Zhenli Li
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Hongwei Jia
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Hanxun Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Xiaomeng Xiu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Dexiang Hu
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Ruiwen Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Ningwei Wang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China
| | - Peng Liu
- Department of Pharmacology, Shenyang Pharmaceutical University, Shenyang, Liaoning, 110016, China.
| | - Huali Yang
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China.
| | - Maosheng Cheng
- Key Laboratory of Structure-Based Drug Design & Discovery of Ministry of Education, School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, Liaoning, Shenyang, 110016, China.
| |
Collapse
|
15
|
Varshney KK, Gupta JK, Srivastava R. Investigating In silico and In vitro Therapeutic Potential of Diosmetin as the Anti-Parkinson Agent. Protein Pept Lett 2024; 31:714-735. [PMID: 39323333 DOI: 10.2174/0109298665333333240909104354] [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: 05/26/2024] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/27/2024]
Abstract
AIM This study aimed to investigate how diosmetin interacts with seven target receptors associated with oxidative stress (OS) and validate its antioxidant properties for the potential management of Parkinson's disease (PD). BACKGROUND In PD, the degeneration of dopaminergic cells is strongly influenced by OS. This stressor is intricately connected to various mechanisms involved in neurodegeneration, such as mitochondrial dysfunction, neuroinflammation, and excitotoxicity induced by nitric oxide. OBJECTIVE The aim of this research was to establish a molecular connection between diosmetin and OS-associated target receptors was the goal, and it investigated how this interaction can lessen PD. METHODS Seven molecular targets - Adenosine A2A (AA2A), Peroxisome Proliferator-Activated Receptor Gamma (PPARγ), Protein Kinase AKT1, Nucleolar Receptor NURR1, Liver - X Receptor Beta (LXRβ), Monoamine Oxidase - B (MAO-B) and Tropomyosin receptor kinase B (TrkB) were obtained from RCSB. Molecular docking software was employed to determine molecular interactions, while antioxidant activity was assessed through in vitro assays against various free radicals. RESULTS Diosmetin exhibited interactions with all seven target receptors at their binding sites. Notably, it showed superior interaction with AA2A and NURR1 compared to native ligands, with binding energies of -7.55, and -6.34 kcal/mol, respectively. Additionally, significant interactions were observed with PPARγ, AKT1, LXRβ, MAO-B, and TrkB with binding energies of -8.34, -5.42, -7.66, -8.82, -8.45 kcal/mol, respectively. Diosmetin also demonstrated antioxidant activity against various free radicals, particularly against hypochlorous acid (HOCl) and nitric oxide (NO) free radicals. CONCLUSION Diosmetin possibly acts on several target receptors linked to the pathophysiology of PD, demonstrating promise as an OS inhibitor and scavenger.
Collapse
Affiliation(s)
| | | | - Rajnish Srivastava
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| |
Collapse
|
16
|
Adouvi G, Nawa F, Ballarotto M, Rüger LA, Knümann L, Kasch T, Arifi S, Schubert-Zsilavecz M, Willems S, Marschner JA, Pabel J, Merk D. Structural Fusion of Natural and Synthetic Ligand Features Boosts RXR Agonist Potency. J Med Chem 2023; 66:16762-16771. [PMID: 38064686 DOI: 10.1021/acs.jmedchem.3c01435] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
The retinoid X receptors (RXRs) are ligand-activated transcription factors involved in, for example, differentiation and apoptosis regulation. Currently used reference RXR agonists suffer from insufficient specificity and poor physicochemical properties, and improved tools are needed to capture the unexplored therapeutic potential of RXR. Endogenous vitamin A-derived RXR ligands and the natural product RXR agonist valerenic acid comprise acrylic acid residues with varying substitution patterns to engage the critical ionic contact with the binding site arginine. To mimic and exploit this natural ligand motif, we probed its structural fusion with synthetic RXR modulator scaffolds, which had profound effects on agonist activity and remarkably boosted potency of an oxaprozin-derived RXR agonist chemotype. Bioisosteric replacement of the acrylic acid to overcome its pan-assay interference compounds (PAINS) character enabled the development of a highly optimized RXR agonist chemical probe.
Collapse
Affiliation(s)
- Gustave Adouvi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Felix Nawa
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Marco Ballarotto
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Lorena Andrea Rüger
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | - Loris Knümann
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Till Kasch
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Silvia Arifi
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
| | | | - Sabine Willems
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Julian A Marschner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Jörg Pabel
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Daniel Merk
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt, 60438 Frankfurt, Germany
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
17
|
Stiller T, Merk D. Exploring Fatty Acid Mimetics as NR4A Ligands. J Med Chem 2023; 66:15362-15369. [PMID: 37918435 PMCID: PMC10683012 DOI: 10.1021/acs.jmedchem.3c01467] [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: 08/09/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
The ligand-activated transcription factors Nur77, Nurr1, and NOR-1 forming the NR4A family of nuclear receptors are considered as potential targets in various pathologies, including neurodegeneration and cancer. However, chemical tools for pharmacological NR4A modulation as a prerequisite for target validation are rare. Recent findings suggest that NR4As bind fatty acid metabolites and fatty acid mimetic (FAM) drugs, opening new opportunities for NR4A modulator development. We have explored the chemical space of FAM NR4A ligands by using fragment screening, in silico analysis, and systematic structure-activity relationship evaluation. From a chemically diverse library of 92 fragments, we identified 11 new FAM NR4A agonist and inverse agonist scaffolds. Structural optimization of the most active FAM fragment yielded NR4A agonists with submicromolar potency and binding affinity, demonstrating remarkable potential of FAM as NR4A-modulating tools and drugs.
Collapse
Affiliation(s)
- Tanja Stiller
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| |
Collapse
|
18
|
Sai M, Vietor J, Kornmayer M, Egner M, López-García Ú, Höfner G, Pabel J, Marschner JA, Wein T, Merk D. Structure-Guided Design of Nurr1 Agonists Derived from the Natural Ligand Dihydroxyindole. J Med Chem 2023; 66:13556-13567. [PMID: 37751901 PMCID: PMC10578347 DOI: 10.1021/acs.jmedchem.3c00852] [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: 05/12/2023] [Indexed: 09/28/2023]
Abstract
The neuroprotective transcription factor Nurr1 was recently found to bind the dopamine metabolite 5,6-dihydroxyindole (DHI) providing access to Nurr1 ligand design from a natural template. We screened a custom set of 14 k extended DHI analogues in silico for optimized descendants to select 24 candidates for microscale synthesis and in vitro testing. Three out of six primary hits were validated as novel Nurr1 agonists with up to sub-micromolar binding affinity, highlighting the druggability of the Nurr1 surface region lining helix 12. In vitro profiling confirmed cellular target engagement of DHI descendants and demonstrated remarkable additive effects of combined Nurr1 agonist treatment, indicating diverse binding sites mediating Nurr1 activation, which may open new avenues in Nurr1 modulation.
Collapse
Affiliation(s)
| | | | - Moritz Kornmayer
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Markus Egner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Úrsula López-García
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Georg Höfner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Jörg Pabel
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Julian A. Marschner
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Thomas Wein
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität München, 81377 Munich, Germany
| |
Collapse
|
19
|
Ballarotto M, Willems S, Stiller T, Nawa F, Marschner JA, Grisoni F, Merk D. De Novo Design of Nurr1 Agonists via Fragment-Augmented Generative Deep Learning in Low-Data Regime. J Med Chem 2023. [PMID: 37256819 DOI: 10.1021/acs.jmedchem.3c00485] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Generative neural networks trained on SMILES can design innovative bioactive molecules de novo. These so-called chemical language models (CLMs) have typically been trained on tens of template molecules for fine-tuning. However, it is challenging to apply CLM to orphan targets with few known ligands. We have fine-tuned a CLM with a single potent Nurr1 agonist as template in a fragment-augmented fashion and obtained novel Nurr1 agonists using sampling frequency for design prioritization. Nanomolar potency and binding affinity of the top-ranking design and its structural novelty compared to available Nurr1 ligands highlight its value as an early chemical tool and as a lead for Nurr1 agonist development, as well as the applicability of CLM in very low-data scenarios.
Collapse
Affiliation(s)
- Marco Ballarotto
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
- Department of Pharmaceutical Sciences, Università degli Studi di Perugia, 06123 Perugia, Italy
| | - Sabine Willems
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Tanja Stiller
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Felix Nawa
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Julian A Marschner
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| | - Francesca Grisoni
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Eindhoven University of Technology, 5612AZ Eindhoven, The Netherlands
- Centre for Living Technologies, Alliance TU/e, WUR, UU, UMC Utrecht, 3584CB Utrecht, The Netherlands
| | - Daniel Merk
- Department of Pharmacy, Ludwig-Maximilians-Universität (LMU) München, 81377 Munich, Germany
| |
Collapse
|