1
|
He Z, Yang W, Yang F, Zhang J, Ma L. Innovative medicinal chemistry strategies for enhancing drug solubility. Eur J Med Chem 2024; 279:116842. [PMID: 39260319 DOI: 10.1016/j.ejmech.2024.116842] [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: 06/24/2024] [Revised: 08/25/2024] [Accepted: 08/26/2024] [Indexed: 09/13/2024]
Abstract
Drug candidates with poor solubility have been recognized as the cause of many drug development failures, owing to the fact that low solubility is unfavorable for physicochemical, pharmacokinetic (PK) and pharmacodynamic (PD) properties. Given the imperative role of solubility during drug development, we herein summarize various strategies for solubility optimizations from a medicinal chemistry perspective, including introduction of polar group, salt formation, structural simplification, disruption of molecular planarity and symmetry, optimizations on the solvent exposed region as well as prodrug design. In addition, methods for solubility assessment and prediction are reviewed. Besides, we have deeply discussed the strategies for solubility improvement. This paper is expected to be beneficial for the development of drug-like molecules with good solubility.
Collapse
Affiliation(s)
- Zhangxu He
- Pharmacy College, Henan University of Chinese Medicine, 450046, Zhengzhou, China
| | - Weiguang Yang
- Children's Hospital Affiliated of Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Henan, Zhengzhou, 450000, China
| | - Feifei Yang
- Pharmacy College, Henan University of Chinese Medicine, 450046, Zhengzhou, China
| | - Jingyu Zhang
- Pharmacy College, Henan University of Chinese Medicine, 450046, Zhengzhou, China.
| | - Liying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China; China Meheco Topfond Pharmaceutical Co., Zhumadian, 463000, China.
| |
Collapse
|
2
|
Desgagné M, Chartier M, Lagard C, Ferková S, Choquette M, Longpré JM, Côté J, Boudreault PL, Sarret P. Development of Macrocyclic Neurotensin Receptor Type 2 (NTS2) Opioid-Free Analgesics. Angew Chem Int Ed Engl 2024; 63:e202405941. [PMID: 39110923 DOI: 10.1002/anie.202405941] [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: 03/27/2024] [Accepted: 08/06/2024] [Indexed: 10/15/2024]
Abstract
The opioid crisis has highlighted the urgent need to develop non-opioid alternatives for managing pain, with an effective, safe, and non-addictive pharmacotherapeutic profile. Using an extensive structure-activity relationship approach, here we have identified a new series of highly selective neurotensin receptor type 2 (NTS2) macrocyclic compounds that exert potent, opioid-independent analgesia in various experimental pain models. To our knowledge, the constrained macrocycle in which the Ile12 residue of NT(7-12) was substituted by cyclopentylalanine, Pro7 and Pro10 were replaced by allyl-glycine followed by side-chain to side-chain cyclization is the most selective analog targeting NTS2 identified to date (Ki 2.9 nM), showing 30,000-fold selectivity over NTS1. Of particular importance, this macrocyclic analog is also able to potentiate the analgesic effects of morphine in a dose- and time-dependent manner. Exerting complementary analgesic actions via distinct mechanisms of nociceptive transmission, NTS2-selective macrocycles can therefore be exploited as opioid-free analgesics or as opioid-sparing therapeutics, offering superior pain relief with reduced adverse effects to pain patients.
Collapse
Affiliation(s)
- Michael Desgagné
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Magali Chartier
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Camille Lagard
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Sára Ferková
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Mathieu Choquette
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Jean-Michel Longpré
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Jérôme Côté
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Pierre-Luc Boudreault
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| | - Philippe Sarret
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Institut de Pharmacologie de Sherbrooke, Université de Sherbrooke, 3001, 12e Avenue Nord, J1H 5N4, Sherbrooke, Québec, Canada
| |
Collapse
|
3
|
Ter Laak A, Hillig RC, Ferrara SJ, Korr D, Barak N, Lienau P, Herbert S, Fernández-Montalván AE, Neuhaus R, Gorjánácz M, Puetter V, Badock V, Bone W, Strathdee C, Siegel F, Schatz C, Nowak-Reppel K, Doehr O, Gradl S, Hartung IV, Meyerson M, Bouché L. Discovery and Characterization of BAY-184: A New Potent and Selective Acylsulfonamide-Benzofuran In Vivo-Active KAT6AB Inhibitor. J Med Chem 2024; 67:19282-19303. [PMID: 39450890 DOI: 10.1021/acs.jmedchem.4c01709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2024]
Abstract
KAT6A and KAT6B genes are two closely related lysine acetyltransferases that transfer an acetyl group from acetyl coenzyme A (AcCoA) to lysine residues of target histone substrates, hence playing a key role in chromatin regulation. KAT6A and KAT6B genes are frequently amplified in various cancer types. In breast cancer, the 8p11-p12 amplicon occurs in 12-15% of cases, resulting in elevated copy numbers and expression levels of chromatin modifiers like KAT6A. Here, we report the discovery of a new acylsulfonamide-benzofuran series as a novel structural class for KAT6A/B inhibition. These compounds were identified through high-throughput screening and subsequently optimized using molecular modeling and cocrystal structure determination. The final tool compound, BAY-184 (29), was successfully validated in an in vivo proof-of-concept study.
Collapse
Affiliation(s)
- Antonius Ter Laak
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Roman C Hillig
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Steven J Ferrara
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, 415 Main St., Cambridge, Massachusetts 02142, United States
| | - Daniel Korr
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Naomi Barak
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Philip Lienau
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Simon Herbert
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | | | - Roland Neuhaus
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Mátyás Gorjánácz
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Vera Puetter
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Volker Badock
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Wilhelm Bone
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Craig Strathdee
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, 415 Main St., Cambridge, Massachusetts 02142, United States
| | - Franziska Siegel
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Christoph Schatz
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Katrin Nowak-Reppel
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Olaf Doehr
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Stefan Gradl
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Ingo V Hartung
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| | - Matthew Meyerson
- Broad Institute of MIT and Harvard, Center for the Development of Therapeutics, 415 Main St., Cambridge, Massachusetts 02142, United States
| | - Léa Bouché
- Bayer AG, Pharmaceuticals, Research and Development, Müllerstrasse 178, Berlin 13353, Germany
| |
Collapse
|
4
|
Gallego RA, Scales S, Toledo C, Auth M, Bernier L, Berry M, Brun S, Chung L, Davis C, Diehl W, Dress K, Eisele K, Elleraas J, Ewanicki J, Fobian Y, Greasley S, Greenwald EC, Johnson TW, Khamphavong P, Lafontaine J, Li J, Linton A, Maestre M, Miller N, Murtaza A, Patman RL, Quinlan CL, Ramms DJ, Richardson P, Sach N, Salomon-Ferrer R, Silva F, Timofeevski S, Tran P, Tran-Dubé M, Wang F, Wang W, Wythes M, Yang S, Zou A, VanArsdale T, McAlpine I. Discovery of Highly Selective Inhibitors of Microtubule-Associated Serine/Threonine Kinase-like (MASTL). J Med Chem 2024; 67:19234-19246. [PMID: 39499084 DOI: 10.1021/acs.jmedchem.4c01659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
By virtue of its role in cellular proliferation, microtubule-associated serine/threonine kinase-like (MASTL) represents a novel target and a first-in-class (FIC) opportunity to provide a new impactful therapeutic agent to oncology patients. Herein, we describe a hit-to-lead optimization effort that resulted in the delivery of two highly selective MASTL inhibitors. Key strategies leveraged to enable this work included structure-based drug design (SBDD), analysis of lipophilic efficiency (LipE) and novel synthesis. The resulting advanced lead compounds enabled a tumor growth inhibition study which was pivotal in assessing the potential value of MASTL as an oncology therapeutic target.
Collapse
Affiliation(s)
- Rebecca A Gallego
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Stephanie Scales
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Chad Toledo
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Marin Auth
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Louise Bernier
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Madeline Berry
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Sonja Brun
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Loanne Chung
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Carl Davis
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Wade Diehl
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Klaus Dress
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Koleen Eisele
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jeff Elleraas
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jason Ewanicki
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Yvette Fobian
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Samantha Greasley
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Eric C Greenwald
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Ted W Johnson
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Penney Khamphavong
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jennifer Lafontaine
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Jian Li
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Angelica Linton
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Michael Maestre
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Nichol Miller
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Anwar Murtaza
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Ryan L Patman
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Casey L Quinlan
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Dana J Ramms
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Paul Richardson
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Neal Sach
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Romelia Salomon-Ferrer
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Francisco Silva
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Sergei Timofeevski
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Phuong Tran
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Michelle Tran-Dubé
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Fen Wang
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Wei Wang
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Martin Wythes
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Shouliang Yang
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Aihua Zou
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Todd VanArsdale
- Oncology Research Unit, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| | - Indrawan McAlpine
- Oncology Medicinal Chemistry, Pfizer Worldwide Research and Development, 10770 Science Center Drive, La Jolla, California 92121, United States
| |
Collapse
|
5
|
Guglielmi G, Zamagni C, Del Re M, Danesi R, Fogli S. Targeting HER2 in breast cancer with brain metastases: A pharmacological point of view with special focus on the permeability of blood-brain barrier to targeted treatments. Eur J Pharmacol 2024; 985:177076. [PMID: 39486766 DOI: 10.1016/j.ejphar.2024.177076] [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: 07/08/2024] [Revised: 10/15/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024]
Abstract
Understanding the capability of a drug to penetrate the blood-brain barrier (BBB) is an unmet medical need in patients with positive human epidermal growth factor receptor 2 (HER2 positive) and brain metastases. The National Comprehensive Cancer Network (NCCN) guidelines recommend the use of tyrosine kinase inhibitors (TKIs) lapatinib, neratinib, and tucatinib in co-administration with monoclonal antibodies or chemotherapy drugs and the antibody-drug conjugates (ADCs) trastuzumab-deruxtecan and trastuzumab-emtansine. Predicting the BBB permeability of these therapeutic agents is a pharmacological challenge due to the various factors involved in the barrier functions. In this review article, we discuss about the molecular and cellular features of the barriers located in the central nervous system and the pharmacological parameters found to be important in predicting BBB permeability in human normal brain, and in the presence of brain metastases. Finally, we reported the clinical outcomes and intracranial response of patients with HER2-positive breast cancer with brain metastases treated with targeted TKIs and ADCs.
Collapse
Affiliation(s)
- Giorgio Guglielmi
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy
| | | | - Marzia Del Re
- Saint Camillus International University of Medical and Health Sciences, Rome, Italy; Direzione Scientifica, Fondazione Policlinico A. Gemelli IRCCS, Rome, Italy
| | - Romano Danesi
- Department of Oncology and Hemato-Oncology, University of Milano, Italy
| | - Stefano Fogli
- Clinical Pharmacology and Pharmacogenetics Unit, Department of Clinical and Experimental Medicine, University of Pisa, Italy.
| |
Collapse
|
6
|
Kadayat TM, Kwiatkowski S, Ortiz D, Shoeran G, Hammill JT, Kim HS, Cholewo J, Landfear SM, Kiplin Guy R. Synthesis and biological evaluation of 4,7,9-trisubstituted benzoxazepines as antileishmanial agents. Bioorg Med Chem Lett 2024; 114:130003. [PMID: 39477128 DOI: 10.1016/j.bmcl.2024.130003] [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: 07/31/2024] [Revised: 10/10/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024]
Abstract
Herein we report a series of antileishmanial analogues derived from 4-[(3,5-dimethyl-4-isoxazolyl)acetyl]-9-[(1-methyl-3-piperidinyl)methoxy]-7-(5-methyl-2-thienyl)-2,3,4,5-tetrahydro-1,4-benzoxazepine (1), which was identified through a previously reported high-throughput phenotypic screen. The analogue series was designed, synthesized, and evaluated for antileishmanial activity to establish pharmacophore elements and preliminary structure-activity relationships as key steps in validating the series for further optimization. This study led to identification of the early lead compound 46, which exhibited sub-micromolar proliferation inhibitory activity against intra-macrophage L. mexicana amastigotes, modest selectivity towards host macrophages (J774A.1 line), and good aqueous solubility.
Collapse
Affiliation(s)
- Tara Man Kadayat
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Stefan Kwiatkowski
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Diana Ortiz
- Department of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Gaurav Shoeran
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Jared T Hammill
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Ho Shin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Joanna Cholewo
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA
| | - Scott M Landfear
- Department of Molecular Microbiology & Immunology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - R Kiplin Guy
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536-0509 USA.
| |
Collapse
|
7
|
Bojovic D, Nikolic M, Nedeljkovic N, Vesovic M, Zivanovic A, Karovic M. Medicinal Chemistry Insights in Neuronal Nitric Oxide Synthase Inhibitors Containing Nitrogen Heterocyclic Compounds: A Mini Review. Chem Biodivers 2024:e202402637. [PMID: 39436922 DOI: 10.1002/cbdv.202402637] [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: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024]
Abstract
Many scientific reports over the last two decades have focused on the discovery and development of novel nNOS inhibitors. The structural identity of isoforms, bioavailability, pharmacokinetic, and safety profile issues remain major obstacles in the discovery of more potent and selective nNOS inhibitors. This review aims to provide an in-depth overview of the molecular interaction patterns between nNOS active site and inhibitors containing structurally diverse nitrogen heterocyclic compounds and highlight the structural properties needed to develop selective nNOS inhibitors. Previously published data allowed the usage of the structure-driven approach in the designing of selective nNOS inhibitors, which relies on the specific structural features required to achieve isoform-selectivity towards nNOS. The incorporation of chiral pyrrolidine ring, two aminopyridine heads, or a specific amino tail group, along with the inhibitor's capacity to adopt the curled conformation in the nNOS environment significantly strengthens the molecular interaction between the inhibitor and nNOS residues by forming specific electrostatic interactions and non-bonded contacts that are vital for isoform selectivity. Additional structure-activity relationship investigations are necessary to elucidate more structural characteristics that will ultimately resolve the exact structural basis required for isoform-selective inhibition of nNOS.
Collapse
Affiliation(s)
- Dijana Bojovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Milos Nikolic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Nikola Nedeljkovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Marina Vesovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Ana Zivanovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| | - Marko Karovic
- Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, 34000, Kragujevac, Serbia
| |
Collapse
|
8
|
Zhu P, Wu Y, Du Z, Li S, Li J, Lu X, Jiang X. Identification of 3-methyl-1-(3-methylpyridin-2-yl)-1H-pyrazol-5-ol as promising neuroprotective agent. Bioorg Med Chem Lett 2024; 114:129983. [PMID: 39395634 DOI: 10.1016/j.bmcl.2024.129983] [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: 07/27/2024] [Revised: 09/19/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024]
Abstract
Pyrazolol derivatives are gaining significant attention for their diverse pharmacological effects, such as analgesic, anti-inflammatory, antioxidant, and anticancer activities. In this study, 20 pyrazolol derivatives were designed and synthesized to develop an anti-ischemic stroke formulation with free radical scavenging activity. Most of these synthesized compounds demonstrated antioxidant capabilities in DPPH, ABTS radical scavenging, and ORACFL assays. The methyl-substituted compound Y12, in particular, showed exceptional antioxidant capacity. Additionally, these compounds showed excellent neurocytoprotective effects in the SH-SY5Y cell injury model subjected to oxygen-glucose deprivation/reoxygenation (OGD/R). Notably, Y12 exhibited significant metal chelating activity with Cu2+. In vivo studies confirmed that compound Y12 has neuroprotective effects and can significantly reduce the infarct area in a mouse model of focal cerebral ischemia induced by transient middle cerebral artery occlusion (tMCAO).
Collapse
Affiliation(s)
- Peng Zhu
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Yulu Wu
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Zhikang Du
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China
| | - Siyi Li
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jiaming Li
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei 230012, China
| | - Xin Lu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Xueyang Jiang
- Department of Medicinal Chemistry, Anhui University of Chinese Medicine, Hefei 230012, China.
| |
Collapse
|
9
|
Tyagarajan S, Andrews CL, Beshore DC, Buevich AV, Curran PJ, Dandliker P, Greshock TJ, Hoar J, Kim A, Karnachi P, Knemeyer I, Kozlowski J, Liu J, Maletic M, Myers R, Rada V, Sha D, Sauvagnat B, Vachal P, Wolkenberg S, Yu W, Yu Y, Krska SW. Rapid Affinity and Microsomal Stability Ranking of Crude Mixture Libraries of Histone Deacetylase Inhibitors. ACS Med Chem Lett 2024; 15:1787-1794. [PMID: 39411537 PMCID: PMC11472384 DOI: 10.1021/acsmedchemlett.4c00345] [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: 07/22/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
The science of drug discovery involves multiparameter optimization of molecular structures through iterative design-make-test cycles. For medicinal chemistry library synthesis, traditional workflows involve the isolation of each individual compound, gravimetric quantitation, and preparation of a standard concentration solution for biological assays. In this work, we explore ways to expedite this process by testing unpurified library mixtures using a combination of mass spectrometry-based assays for affinity selection and microsomal metabolic stability. Utilizing this approach, microgram quantities of crude library mixtures can be used to identify high affinity, metabolically stable library members for isolation and full characterization. This streamlined approach was demonstrated for the synthesis and evaluation of two libraries of histone deacetylase inhibitors and was shown to generate decision-making data in line with traditional workflows. The advantages of this paradigm include greatly reduced cycle time, reduced material requirements, and concentration of resources on the most promising compounds.
Collapse
Affiliation(s)
- Sriram Tyagarajan
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Christine L. Andrews
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Douglas C. Beshore
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Alexei V. Buevich
- Analytical
Research & Development, Merck &
Co., Inc., Rahway, New Jersey 07065, United States
| | - Patrick J. Curran
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Peter Dandliker
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Thomas J. Greshock
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Jason Hoar
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck
& Co., Inc., Rahway, New Jersey 07065, United States
| | - Alex Kim
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Prabha Karnachi
- Modeling
and Informatics, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Ian Knemeyer
- Pharmacokinetics,
Pharmacodynamics and Drug Metabolism, Merck
& Co., Inc., Boston, Massachusetts 02115, United States
| | - Joseph Kozlowski
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Jian Liu
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Milana Maletic
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Robert Myers
- Department
of Pharmacology, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Vanessa Rada
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Deyou Sha
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Berengere Sauvagnat
- Quantitative
Biosciences, Merck & Co., Inc., Boston, Massachusetts 02115, United States
| | - Petr Vachal
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Scott Wolkenberg
- Discovery
Chemistry, Merck & Co., Inc., West Point, Pennsylvania 19486, United States
| | - Wensheng Yu
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Younong Yu
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| | - Shane W. Krska
- Discovery
Chemistry, Merck & Co., Inc., Rahway, New Jersey 07065, United States
| |
Collapse
|
10
|
Barkan D, Garland K, Zhang L, Eastman RT, Hesse M, Knapp M, Ornelas E, Tang J, Cortopassi WA, Wang Y, King F, Jia W, Nguyen Z, Frank AO, Chan R, Fang E, Fuller D, Busby S, Carias H, Donahue K, Tandeske L, Diagana TT, Jarrousse N, Moser H, Sarko C, Dovala D, Moquin S, Marx VM. Identification of Potent, Broad-Spectrum Coronavirus Main Protease Inhibitors for Pandemic Preparedness. J Med Chem 2024; 67:17454-17471. [PMID: 39332817 PMCID: PMC11472307 DOI: 10.1021/acs.jmedchem.4c01404] [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: 06/20/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/29/2024]
Abstract
The COVID-19 pandemic highlights the ongoing risk of zoonotic transmission of coronaviruses to global health. To prepare for future pandemics, it is essential to develop effective antivirals targeting a broad range of coronaviruses. Targeting the essential and clinically validated coronavirus main protease (Mpro), we constructed a structurally diverse Mpro panel by clustering all known coronavirus sequences by Mpro active site sequence similarity. Through screening, we identified a potent covalent inhibitor that engaged the catalytic cysteine of SARS-CoV-2 Mpro and used structure-based medicinal chemistry to develop compounds in the pyrazolopyrimidine sulfone series that exhibit submicromolar activity against multiple Mpro homologues. Additionally, we solved the first X-ray cocrystal structure of Mpro from the human-infecting OC43 coronavirus, providing insights into potency differences among compound-target pairs. Overall, the chemical compounds described in this study serve as starting points for the development of antivirals with broad-spectrum activity, enhancing our preparedness for emerging human-infecting coronaviruses.
Collapse
Affiliation(s)
- David
T. Barkan
- Discovery
Sciences, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Keira Garland
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Lei Zhang
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Richard T. Eastman
- Global
Health, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Matthew Hesse
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Mark Knapp
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Elizabeth Ornelas
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Jenny Tang
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Wilian Augusto Cortopassi
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Yu Wang
- Discovery
Sciences, Novartis Biomedical Research, La Jolla, California 92121, United States
| | - Frederick King
- Discovery
Sciences, Novartis Biomedical Research, La Jolla, California 92121, United States
| | - Weiping Jia
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Zachary Nguyen
- Discovery
Sciences, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Andreas O. Frank
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Ryan Chan
- Global
Health, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Eric Fang
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Daniel Fuller
- Discovery
Sciences, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Scott Busby
- Discovery
Sciences, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Heidi Carias
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Kristine Donahue
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Laura Tandeske
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Thierry T. Diagana
- Global
Health, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Nadine Jarrousse
- Global
Health, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Heinz Moser
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Christopher Sarko
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| | - Dustin Dovala
- Discovery
Sciences, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Stephanie Moquin
- Global
Health, Novartis Biomedical Research, Emeryville, California 94608, United States
| | - Vanessa M. Marx
- Global
Discovery Chemistry, Novartis Biomedical
Research, Emeryville, California 94608, United States
| |
Collapse
|
11
|
Patel KV, Gadotti VM, Garcia-Caballero A, Antunes FTT, Ali MY, Zamponi GW, Derksen DJ. Development of Tetrahydroquinoline-Based Inhibitors for Chronic Pain. ACS Chem Neurosci 2024. [PMID: 39377454 DOI: 10.1021/acschemneuro.4c00316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/09/2024] Open
Abstract
Chronic pain affects a substantial portion of the population, posing a significant health challenge. Current treatments often come with limitations and side effects, necessitating novel therapeutic approaches. Our study focuses on disrupting the Cav3.2-USP5 interaction as a strategy for chronic pain management. Through structure-activity relationship studies of a tetrahydroquinoline (THQ) scaffold, we identified a family of lead molecules that demonstrated potent inhibition of the Cav3.2-USP5 interaction. In vitro pharmacokinetic assessments and in vivo studies support the efficacy and drug-like properties of the lead compounds in mouse models of acute and chronic pain. Dependence on the Cav3.2 channels was validated in Cav3.2 null mice, consistent with the proposed mode of action of these small molecules. These findings provide a novel chronic pain treatment strategy, highlighting the potential of these small molecules for further development.
Collapse
Affiliation(s)
- Ketul V Patel
- Department of Chemistry, University of Calgary, Calgary T2N 1N4, Alberta, Canada
- Zymedyne Therapeutics, Calgary T2N 4G4, Alberta, Canada
| | - Vinicius M Gadotti
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Agustin Garcia-Caballero
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Flavia T T Antunes
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Md Yousof Ali
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
- Zymedyne Therapeutics, Calgary T2N 4G4, Alberta, Canada
| | - Gerald W Zamponi
- Department of Clinical Neurosciences, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| | - Darren J Derksen
- Department of Chemistry, University of Calgary, Calgary T2N 1N4, Alberta, Canada
- Hotchkiss Brain Institute, University of Calgary, Calgary T2N 4N1, Alberta, Canada
- Alberta Children's Hospital Research Institute, Calgary T2N 4N1, Alberta, Canada
| |
Collapse
|
12
|
Martula E, Morak-Młodawska B, Jeleń M, Okechukwu PN. Analysis of Lipophilicity and Pharmacokinetic Parameters of Dipyridothiazine Dimers with Anticancer Potency. Pharmaceutics 2024; 16:1235. [PMID: 39339271 PMCID: PMC11435374 DOI: 10.3390/pharmaceutics16091235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/09/2024] [Accepted: 09/11/2024] [Indexed: 09/30/2024] Open
Abstract
Lipophilicity is an essential parameter of a compound that determines the solubility and pharmacokinetic properties that determine the transport of the drug to the molecular target. Dimers of dipyridothiazines are diazaphenothiazine derivatives exhibiting diverse anticancer potential in vitro, which is related to their affinity for histone deacetylase. In this study, the lipophilicity of 16 isomeric dipyridothiazine dimers was investigated theoretically and experimentally by reversed-phase thin-layer chromatography (RP-TLC) in an acetone-TRIS buffer (pH = 7.4). The relative lipophilicity parameter RM0 and specific hydrophobic surface area b were significantly intercorrelated, showing congeneric classes of dimers. The parameter RM0 was transformed into parameter logPTLC by use of the calibration curve. Molecular descriptors, ADMET parameters and probable molecular targets were determined in silico for analysis of the pharmacokinetic profile of the tested compounds showing anticancer activity. The analyzed compounds were tested in the context of Lipinski's rule of five, Ghose's rule and Veber's rule, confirming their bioavailability.
Collapse
Affiliation(s)
- Emilia Martula
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, The Medical University of Silesia, Jagiellońska 4, 41-200 Sosnowiec, Poland
| | - Patrick Nwabueze Okechukwu
- Department of Biotechnology, Faculty of Applied Sciences, UCSI University, Cheras, Kuala Lumpur 56000, Malaysia
| |
Collapse
|
13
|
Ling T, Arroyo-Cruz LV, Smither WR, Seighman EK, Martínez-Montemayor MM, Rivas F. Early Preclinical Studies of Ergosterol Peroxide and Biological Evaluation of Its Derivatives. ACS OMEGA 2024; 9:37117-37127. [PMID: 39246459 PMCID: PMC11375702 DOI: 10.1021/acsomega.4c04350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/18/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024]
Abstract
Ganoderma lucidum is a medicinal mushroom that produces various pharmacological compounds, including triterpenoids. A major bioactive component of G. lucidum is ergosterol peroxide (EP), which is attributed to its anticancer effects. The current study focuses on the in vitro ADME (absorption, distribution, metabolism, and elimination), in vivo efficacy and toxicity of EP, and the synthesis of new EP derivatives to improve aqueous solubility. It was found that EP is metabolically stable in liver microsomes and plasma. In vivo studies showed that EP inhibits tumor growth in murine cancer models, and it is well tolerated by mice. The maximum tolerated dose was investigated in mice at escalating doses with a defined maximum amount of 500 mg/kg, which indicated no signs of toxicity, confirmed by plasma chemistry and analysis of harvested tissues. Complementary organ toxicity assays including cardio and hepatotoxicity assays of EP demonstrated no inhibitory effects. Next, a focused library of EP derivatives was developed to investigate the iterative addition of heteroatoms to improve the aqueous solubility properties of EP. Significant solubility improvement was observed by the introduction of hydrogen bonding promoting groups, particularly the sulfate group. Superior aqueous solubility properties are directly correlated with the biological activity of the compound against triple-negative breast cancer cellular (TNBC) models. The EP derivatives maintain ample therapeutic index at the tested concentrations, indicating they engage with the same biological target(s) as the parental compound (EP). The combined studies indicate that EP and its derivatives are selective TNBC cell death inducers, while sparing noncancerous tissue.
Collapse
Affiliation(s)
- Taotao Ling
- Department of Chemistry, Louisiana State University, 133 Chopping Hall, Baton Rouge, Louisiana 70803, United States
| | - Luz V Arroyo-Cruz
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, P.O. Box 60327, Bayamón, Puerto Rico 00960-6032, United States
| | - William R Smither
- Department of Chemistry, Louisiana State University, 133 Chopping Hall, Baton Rouge, Louisiana 70803, United States
| | - Emily K Seighman
- Department of Chemistry, Louisiana State University, 133 Chopping Hall, Baton Rouge, Louisiana 70803, United States
| | - Michelle M Martínez-Montemayor
- Department of Biochemistry, Universidad Central del Caribe, School of Medicine, P.O. Box 60327, Bayamón, Puerto Rico 00960-6032, United States
| | - Fatima Rivas
- Department of Chemistry, Louisiana State University, 133 Chopping Hall, Baton Rouge, Louisiana 70803, United States
| |
Collapse
|
14
|
Bhattacharya S, Dutta A, Khanra PK, Gupta N, Dutta R, Tzvetkov NT, Milella L, Ponticelli M. In silico exploration of 4(α-l-rhamnosyloxy)-benzyl isothiocyanate: A promising phytochemical-based drug discovery approach for combating multi-drug resistant Staphylococcus aureus. Comput Biol Med 2024; 179:108907. [PMID: 39033680 DOI: 10.1016/j.compbiomed.2024.108907] [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: 06/04/2024] [Revised: 07/14/2024] [Accepted: 07/15/2024] [Indexed: 07/23/2024]
Abstract
Multidrug-resistant (MDR) Staphylococcus aureus infections significantly threaten global health. With rising resistance to current antibiotics and limited solutions, the urgent discovery of new, effective, and affordable antibacterials with low toxicity is imperative to combat diverse MDR S. aureus strains. Hence, in this study, we introduce an in silico phytochemical-based approach for discovering novel antibacterial agents, underscoring the potential of computational approaches in therapeutic discovery. Glucomoringin Isothiocyanate (GMG-ITC) from Moringa oleifera Lam. is one of the phytochemical compounds with several biological activities, including antimicrobial, anti-inflammatory, and antioxidant activities, and is also effective against S. aureus. This study focuses on screening GMG-ITC as a potential drug candidate to combat MDR S. aureus infections through a molecular docking approach. Moreover, interaction amino acid analysis, in silico pharmacokinetics, compound target prediction, pathway enrichment analysis and molecular dynamics (MD) simulations were conducted for further investigation. Molecular docking and interaction analysis showed strong binding affinity towards S. aureus lipase, dihydrofolate reductase, and other MDR S. aureus proteins, including penicillin-binding protein 2a, MepR, D-Ala:D-Ala ligase, and RPP TetM, through hydrophilic and hydrophobic interactions. GMG-ITC also showed a strong binding affinity to cyclooxygenase-2 and FAD-dependent NAD(P)H oxidase, suggesting that it is a potential anti-inflammatory and antioxidant candidate that may eliminate inflammation and oxidative stress associated with S. aureus infections. MD simulations validated the stability of the GMG-ITC molecular interactions determined by molecular docking. In silico pharmacokinetic analysis highlights its potency as a drug candidate, showing strong absorption, distribution, and excretion properties in combination with low toxicity. It acts as an active protease and enzyme inhibitor with moderate activity against GPCR ligands, ion channels, nuclear receptor ligands, and kinases. Enrichment analysis further elucidated its involvement in important biological, molecular, and cellular functions with potential therapeutic applications in diseases like cancer, hepatitis B, and influenza. Results suggest that GMG-ITC is an effective antibacterial agent that could treat MDR S. aureus-associated infections.
Collapse
Affiliation(s)
- Soham Bhattacharya
- Department of Agroecology and Crop Production, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, Prague 6, Suchdol, 165 00, Czech Republic
| | - Adrish Dutta
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, 165 00, Prague 6, Czech Republic
| | - Pijush Kanti Khanra
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati, 39, Assam, India
| | - Neha Gupta
- Department of Crop Sciences and Agroforestry, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Kamýcká 129, Suchdol, 165 00, Prague 6, Czech Republic
| | - Ritesh Dutta
- Environmental Biotechnology & Genomics Division, CSIR-National Environmental Engineering Research Institute (CSIR-NEERI), Nehru Marg, Nagpur, 440020, India
| | - Nikolay T Tzvetkov
- Department of Biochemical Pharmacology & Drug Design, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences (BAS), Acad. G. Bonchev Str., Bl. 21, 1113, Sofia, Bulgaria
| | - Luigi Milella
- Department of Science, University of Basilicata, Via Dell'Ateneo Lucano 10, 85100, Potenza, Italy.
| | - Maria Ponticelli
- Department of Biochemical Pharmacology & Drug Design, Institute of Molecular Biology "Roumen Tsanev", Bulgarian Academy of Sciences (BAS), Acad. G. Bonchev Str., Bl. 21, 1113, Sofia, Bulgaria; Department of Science, University of Basilicata, Via Dell'Ateneo Lucano 10, 85100, Potenza, Italy
| |
Collapse
|
15
|
Bernkop-Schnürch AD, Huber K, Clauser A, Cziferszky M, Leitner D, Talasz H, Hermann M, Hohloch S, Gust R, Kircher B. Design, synthesis, and biological evaluation of novel halogenated chlorido[N,N'-bis(salicylidene)-1,2-bis(3-methoxyphenyl)ethylenediamine]iron(III) complexes as anticancer agents. J Biol Inorg Chem 2024; 29:583-599. [PMID: 39133326 PMCID: PMC11390779 DOI: 10.1007/s00775-024-02067-9] [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: 01/19/2024] [Accepted: 07/17/2024] [Indexed: 08/13/2024]
Abstract
Iron(III) complexes based on N,N´-bis(salicylidene)ethylenediamine (salene) scaffolds have demonstrated promising anticancer features like induction of ferroptosis, an iron dependent cell death. Since poor cellular uptake limits their therapeutical potential, this study aimed to enhance the lipophilic character of chlorido[N,N'-bis(salicylidene)-1,2-bis(3-methoxyphenyl)ethylenediamine]iron(III) complexes by introducing lipophilicity improving ligands such as fluorine (X1), chlorine (X2) and bromine (X3) in 5-position in the salicylidene moieties. After detailed characterization the binding to nucleophiles, logP values and cellular uptake were determined. The complexes were further evaluated regarding their biological activity on MDA-MB 231 mammary carcinoma, the non-tumorous SV-80 fibroblast, HS-5 stroma and MCF-10A mammary gland cell lines. Stability of the complexes in aqueous and biological environments was proven by the lack of interactions with amino acids and glutathione. Cellular uptake was positively correlated with the logP values, indicating that higher lipophilicity enhanced cellular uptake. The complexes induced strong antiproliferative and antimetabolic effects on MDA-MB 231 cells, but were inactive on all non-malignant cells tested. Generation of mitochondrial reactive oxygen species, increase of lipid peroxidation and induction of both ferroptosis and necroptosis were identified as mechanisms of action. In conclusion, halogenation of chlorido[N,N'-bis(salicylidene)-1,2-bis(3-methoxyphenyl)ethylenediamine]iron(III) complexes raises their lipophilic character resulting in improved cellular uptake.
Collapse
Affiliation(s)
- Astrid Dagmar Bernkop-Schnürch
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Klaus Huber
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Armida Clauser
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
- Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology), Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Monika Cziferszky
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Daniel Leitner
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Heribert Talasz
- Biocenter, Institute of Medical Biochemistry, Protein Core Facility, Medical University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Martin Hermann
- Department of Anesthesiology and Critical Care Medicine, Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria
| | - Stephan Hohloch
- Department of General, Inorganic and Theoretical Chemistry, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Ronald Gust
- Department of Pharmaceutical Chemistry, Institute of Pharmacy, CMBI-Center for Molecular Biosciences Innsbruck, CCB-Center for Chemistry and Biomedicine, University of Innsbruck, Innrain 80-82, 6020, Innsbruck, Austria
| | - Brigitte Kircher
- Immunobiology and Stem Cell Laboratory, Department of Internal Medicine V (Hematology and Oncology), Medical University of Innsbruck, Anichstraße 35, 6020, Innsbruck, Austria.
- Tyrolean Cancer Research Institute, Innrain 66, 6020, Innsbruck, Austria.
| |
Collapse
|
16
|
Gallego RA, Edwards MP, Montgomery TP. An update on lipophilic efficiency as an important metric in drug design. Expert Opin Drug Discov 2024; 19:917-931. [PMID: 38919130 DOI: 10.1080/17460441.2024.2368744] [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: 05/10/2024] [Accepted: 06/12/2024] [Indexed: 06/27/2024]
Abstract
INTRODUCTION Lipophilic efficiency (LipE) and lipophilic metabolic efficiency (LipMetE) are valuable tools that can be utilized as part of a multiparameter optimization process to advance a hit to a clinical quality compound. AREAS COVERED This review covers recent, effective use cases of LipE and LipMetE that have been published in the literature over the past 5 years. These use cases resulted in the delivery of high-quality molecules that were brought forward to in vivo work and/or to clinical studies. The authors discuss best-practices for using LipE and LipMetE analysis, combined with lipophilicity-focused compound design strategies, to increase the speed and effectiveness of the hit to clinical quality compound optimization process. EXPERT OPINION It has become well established that increasing LipE and LipMetE within a series of analogs facilitates the improvement of broad selectivity, clearance, solubility, and permeability and, through this optimization, also facilitates the achievement of desired pharmacokinetic properties, efficacy, and tolerability. Within this article, we discuss lipophilic efficiency-focused optimization as a tool to yield high-quality potential clinical candidates. It is suggested that LipE/LipMetE-focused optimization can facilitate and accelerate the drug-discovery process.
Collapse
|
17
|
Gobbo A, Pereira SAP, Mota FAR, Sinenko I, Glinkina K, Rocchi D, Guelfi M, Biver T, Donati C, Zacchini S, Saraiva MLMFS, Dyson PJ, Marchetti F. Anticancer potential of NSAID-derived tris(pyrazolyl)methane ligands in iron(II) sandwich complexes. Dalton Trans 2024. [PMID: 39072444 DOI: 10.1039/d4dt00920g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Tris(pyrazolyl)methane (tpm), 2,2,2-tris(pyrazolyl)ethanol (tpmOH) and its esterification derivatives with ibuprofen and flurbiprofen (tpmIBU and tpmFLU) were used as ligands to obtain complexes of the type [Fe(tpmX)2]Cl2 (1-4). The tpmIBU and tpmFLU ligands and corresponding complexes 3 and 4 were characterized by IR and multinuclear NMR spectroscopy, and the structure of tpmIBU was elucidated by single crystal X-ray diffraction. Complexes 1-4 were also assessed for their behaviour in aqueous media (solubility in D2O, octanol/water partition coefficient, stability in physiological-like conditions). The antiproliferative activity of ligands and complexes was determined on A2780, A2780cis and A549 cancer cell lines and the non-cancerous HEK 293T and BJ cell lines. The ligands and complexes were investigated for their ability to inhibit COX-2 (cyclooxygenase) and HNE (4-hydroxynonenal) enzymes. Complexes 3 and 4 exhibited cytotoxicity that may be attributed predominantly to their bioactive fragments, while DNA binding and enhancement of ROS production do not appear to play any significant role.
Collapse
Affiliation(s)
- Alberto Gobbo
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy.
| | - Sarah A P Pereira
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia da Universidade do Porto, Portugal
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Switzerland
| | - Fátima A R Mota
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia da Universidade do Porto, Portugal
| | - Irina Sinenko
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Switzerland
| | - Kseniya Glinkina
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Switzerland
| | - Dario Rocchi
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy.
| | - Massimo Guelfi
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy.
| | - Tarita Biver
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy.
| | - Chiara Donati
- University of Padova, Department of Pharmaceutical and Pharmacological Sciences, Via F. Marzolo 5, I-35131 Padova, Italy
| | - Stefano Zacchini
- University of Bologna, Department of Industrial Chemistry "Toso Montanari", Via P. Gobetti 85, I-40129 Bologna, Italy
| | - M Lúcia M F S Saraiva
- LAQV, REQUIMTE, Laboratório de Química Aplicada, Faculdade de Farmácia da Universidade do Porto, Portugal
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH 1015, Switzerland
| | - Fabio Marchetti
- University of Pisa, Department of Chemistry and Industrial Chemistry, Via G. Moruzzi 13, I-56124 Pisa, Italy.
| |
Collapse
|
18
|
De Franco M, Biancalana L, Zappelli C, Zacchini S, Gandin V, Marchetti F. 1,3,5-Triaza-7-phosphaadamantane and Cyclohexyl Groups Impart to Di-Iron(I) Complex Aqueous Solubility and Stability, and Prominent Anticancer Activity in Cellular and Animal Models. J Med Chem 2024; 67:11138-11151. [PMID: 38951717 DOI: 10.1021/acs.jmedchem.4c00641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/03/2024]
Abstract
Using a multigram-scalable synthesis, we obtained nine dinuclear complexes based on nonendogenous iron(I) centers and featuring variable aminocarbyne and P-ligands. One compound from the series (FEACYP) emerged for its strong cytotoxicity in vitro against four human cancer cell lines, surpassing the activity of cisplatin by 3-6 times in three cell lines, with an average selectivity index of 6.2 compared to noncancerous HEK293 cells. FEACYP demonstrated outstanding water solubility (15 g/L) and stability in physiological-like solutions. It confirmed its superior antiproliferative activity when tested in 3D spheroids of human pancreatic cancer cells and showed a capacity to inhibit thioredoxin reductase (TrxR) similar to auranofin. In vivo treatment of murine LLC carcinoma with FEACYP (8 mg kg-1 dose) led to excellent tumor growth suppression (88%) on day 15, with no signs of systemic toxicity and only limited body weight loss.
Collapse
Affiliation(s)
- Michele De Franco
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, I-35131 Padova, Italy
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Chiara Zappelli
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy
| | - Stefano Zacchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Via P. Gobetti 85, I-40129 Bologna, Italy
| | - Valentina Gandin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, I-35131 Padova, Italy
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via G. Moruzzi 13, I-56124 Pisa, Italy
| |
Collapse
|
19
|
Chiodi D, Ishihara Y. The role of the methoxy group in approved drugs. Eur J Med Chem 2024; 273:116364. [PMID: 38781921 DOI: 10.1016/j.ejmech.2024.116364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/12/2024] [Accepted: 03/23/2024] [Indexed: 05/25/2024]
Abstract
The methoxy substituent is prevalent in natural products and, consequently, is present in many natural product-derived drugs. It has also been installed in modern drug molecules with no remnant of natural product features because medicinal chemists have been taking advantage of the benefits that this small functional group can bestow on ligand-target binding, physicochemical properties, and ADME parameters. Herein, over 230 methoxy-containing small-molecule drugs, as well as several fluoromethoxy-containing drugs, are presented from the vantage point of the methoxy group. Biochemical mechanisms of action, medicinal chemistry SAR studies, and numerous X-ray cocrystal structures are analyzed to identify the precise role of the methoxy group for many of the drugs and drug classes. Although the methoxy substituent can be considered as the hybridization of a hydroxy and a methyl group, the combination of these functionalities often results in unique effects that can amount to more than the sum of the individual parts.
Collapse
Affiliation(s)
- Debora Chiodi
- Department of Chemistry, Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, CA, 92121, USA
| | - Yoshihiro Ishihara
- Department of Chemistry, Vividion Therapeutics, 5820 Nancy Ridge Drive, San Diego, CA, 92121, USA.
| |
Collapse
|
20
|
Hirst DJ, Bamborough P, Al-Mahdi N, Angell DC, Barnett HA, Baxter A, Bit RA, Brown JA, Chung CW, Craggs PD, Davis RP, Demont EH, Ferrie A, Gordon LJ, Harada I, Ho TCT, Holyer ID, Hooper-Greenhill E, Jones KL, Lindon MJ, Lovatt C, Lugo D, Maller C, McGonagle G, Messenger C, Mitchell DJ, Pascoe DD, Patel VK, Patten C, Poole DL, Shah RR, Rioja I, Stafford KAJ, Tape D, Taylor S, Theodoulou NH, Tomlinson L, Wall ID, Wellaway CR, White G, Prinjha RK, Humphreys PG. Structure- and Property-Based Optimization of Efficient Pan-Bromodomain and Extra Terminal Inhibitors to Identify Oral and Intravenous Candidate I-BET787. J Med Chem 2024; 67:10464-10489. [PMID: 38866424 DOI: 10.1021/acs.jmedchem.4c00959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The bromodomain and extra terminal (BET) family of bromodomain-containing proteins are important epigenetic regulators that elicit their effect through binding histone tail N-acetyl lysine (KAc) post-translational modifications. Recognition of such markers has been implicated in a range of oncology and immune diseases and, as such, small-molecule inhibition of the BET family bromodomain-KAc protein-protein interaction has received significant interest as a therapeutic strategy, with several potential medicines under clinical evaluation. This work describes the structure- and property-based optimization of a ligand and lipophilic efficient pan-BET bromodomain inhibitor series to deliver candidate I-BET787 (70) that demonstrates efficacy in a mouse model of inflammation and suitable properties for both oral and intravenous (IV) administration. This focused two-phase explore-exploit medicinal chemistry effort delivered the candidate molecule in 3 months with less than 100 final compounds synthesized.
Collapse
Affiliation(s)
- David J Hirst
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Paul Bamborough
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Niam Al-Mahdi
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Davina C Angell
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Heather A Barnett
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Andrew Baxter
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Rino A Bit
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Jack A Brown
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Chun-Wa Chung
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Peter D Craggs
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Robert P Davis
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Emmanuel H Demont
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Alan Ferrie
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Laurie J Gordon
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Isobel Harada
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Tim C T Ho
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Ian D Holyer
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Katherine L Jones
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Matthew J Lindon
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Cerys Lovatt
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - David Lugo
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Claire Maller
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Grant McGonagle
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Cassie Messenger
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Darren J Mitchell
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - David D Pascoe
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | | | - Darren L Poole
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Rishi R Shah
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Inmaculada Rioja
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Daniel Tape
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Simon Taylor
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Laura Tomlinson
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Ian D Wall
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | | - Gemma White
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Rab K Prinjha
- GSK Medicines Research Centre, Stevenage, Hertfordshire SG1 2NY, U.K
| | | |
Collapse
|
21
|
Mammoliti O, Martina S, Claes P, Coti G, Blanque R, Jagerschmidt C, Shoji K, Borgonovi M, De Vos S, Marsais F, Oste L, Quinton E, López-Ramos M, Amantini D, Brys R, Jimenez JM, Galien R, van der Plas S. Discovery of GLPG3667, a Selective ATP Competitive Tyrosine Kinase 2 Inhibitor for the Treatment of Autoimmune Diseases. J Med Chem 2024; 67:8545-8568. [PMID: 38805213 PMCID: PMC11181332 DOI: 10.1021/acs.jmedchem.4c00769] [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: 04/02/2024] [Revised: 05/08/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024]
Abstract
Tyrosine kinase 2 (TYK2) mediates cytokine signaling through type 1 interferon, interleukin (IL)-12/IL-23, and the IL-10 family. There appears to be an association between TYK2 genetic variants and inflammatory conditions, and clinical evidence suggests that selective inhibition of TYK2 could produce a unique therapeutic profile. Here, we describe the discovery of compound 9 (GLPG3667), a reversible and selective TYK2 adenosine triphosphate competitive inhibitor in development for the treatment of inflammatory and autoimmune diseases. The preclinical pharmacokinetic profile was favorable, and TYK2 selectivity was confirmed in peripheral blood mononuclear cells and whole blood assays. Dermal ear inflammation was reduced in an IL-23-induced in vivo mouse model of psoriasis. GLPG3667 also completed a phase 1b study (NCT04594928) in patients with moderate-to-severe psoriasis where clinical effect was shown within the 4 weeks of treatment and it is now in phase 2 trials for the treatment of dermatomyositis (NCT05695950) and systemic lupus erythematosus (NCT05856448).
Collapse
Affiliation(s)
- Oscar Mammoliti
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | | | - Pieter Claes
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Ghjuvanni Coti
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Roland Blanque
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - Kenji Shoji
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Monica Borgonovi
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Steve De Vos
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Florence Marsais
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Line Oste
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | - Evelyne Quinton
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | | - David Amantini
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | - Reginald Brys
- Galapagos
NV, Generaal De Wittelaan
L11, A3, 2800 Mechelen, Belgium
| | | | - René Galien
- Galapagos
SASU, 102 Avenue Gaston Roussel, 93230 Romainville, France
| | | |
Collapse
|
22
|
Lucas SCC, Blackwell JH, Börjesson U, Hargreaves D, Milbradt AG, Ahmed S, Bostock MJ, Guerot C, Gohlke A, Kinzel O, Lamb ML, Selmi N, Stubbs CJ, Su N, Su Q, Luo H, Xiong T, Zuo X, Bazzaz S, Bienstock C, Centrella PA, Denton KE, Gikunju D, Guié MA, Guilinger JP, Hupp C, Keefe AD, Satoh T, Zhang Y, Rivers EL. Identification and Evaluation of Reversible Covalent Binders to Cys55 of Bfl-1 from a DNA-Encoded Chemical Library Screen. ACS Med Chem Lett 2024; 15:791-797. [PMID: 38894895 PMCID: PMC11181504 DOI: 10.1021/acsmedchemlett.4c00113] [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: 03/07/2024] [Revised: 05/02/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Bfl-1 is overexpressed in both hematological and solid tumors; therefore, inhibitors of Bfl-1 are highly desirable. A DNA-encoded chemical library (DEL) screen against Bfl-1 identified the first known reversible covalent small-molecule ligand for Bfl-1. The binding was validated through biophysical and biochemical techniques, which confirmed the reversible covalent mechanism of action and pointed to binding through Cys55. This represented the first identification of a cyano-acrylamide reversible covalent compound from a DEL screen and highlights further opportunities for covalent drug discovery through DEL screening. A 10-fold improvement in potency was achieved through a systematic SAR exploration of the hit. The more potent analogue compound 13 was successfully cocrystallized in Bfl-1, revealing the binding mode and providing further evidence of a covalent interaction with Cys55.
Collapse
Affiliation(s)
- Simon C. C. Lucas
- Hit
Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K.
| | - J. Henry Blackwell
- Hit
Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K.
| | - Ulf Börjesson
- Hit
Discovery, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - David Hargreaves
- Mechanistic
and Structural Biology, Discovery Sciences, R&DAstraZeneca, Cambridge CB2 0AA, U.K.
| | - Alexander G. Milbradt
- Mechanistic
and Structural Biology, Discovery Sciences, R&DAstraZeneca, Cambridge CB2 0AA, U.K.
| | - Samiyah Ahmed
- Discovery
Biology, Discovery Sciences, R&DAstraZeneca, Cambridge CB2 0AA, U.K.
| | - Mark J. Bostock
- Mechanistic
and Structural Biology, Discovery Sciences, R&DAstraZeneca, Cambridge CB2 0AA, U.K.
| | - Carine Guerot
- Medicinal
Chemistry, Oncology, R&D, AstraZeneca, Cambridge CB2 0AA, U.K.
| | - Andrea Gohlke
- Mechanistic
and Structural Biology, Discovery Sciences, R&DAstraZeneca, Cambridge CB2 0AA, U.K.
| | - Olaf Kinzel
- Medicinal
Chemistry, Oncology, R&D, Acerta B.V.,
a member of the AstraZeneca Group, Oss 5349, The Netherlands
| | - Michelle L. Lamb
- Medicinal
Chemistry, Oncology, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Nidhal Selmi
- Compound
Synthesis and Management, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Christopher J. Stubbs
- Mechanistic
and Structural Biology, Discovery Sciences, R&DAstraZeneca, Cambridge CB2 0AA, U.K.
| | - Nancy Su
- Mechanistic
and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Qibin Su
- Medicinal
Chemistry, Oncology, R&D, AstraZeneca, Waltham, Massachusetts 02451, United States
| | - Haiou Luo
- Pharmaron Beijing Co., Ltd., Beijing 100176, P. R. China
| | - Ting Xiong
- Pharmaron Beijing Co., Ltd., Beijing 100176, P. R. China
| | - Xiaoqian Zuo
- Pharmaron Beijing Co., Ltd., Beijing 100176, P. R. China
| | - Sana Bazzaz
- X-Chem Inc., Waltham, Massachusetts 02453, United States
| | | | | | - Kyle E. Denton
- X-Chem Inc., Waltham, Massachusetts 02453, United States
| | - Diana Gikunju
- X-Chem Inc., Waltham, Massachusetts 02453, United States
| | | | | | | | | | - Takashi Satoh
- X-Chem Inc., Waltham, Massachusetts 02453, United States
| | - Ying Zhang
- X-Chem Inc., Waltham, Massachusetts 02453, United States
| | - Emma L. Rivers
- Hit
Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K.
| |
Collapse
|
23
|
Le TM, Njangiru IK, Vincze A, Zupkó I, Balogh GT, Szakonyi Z. Synthesis and medicinal chemical characterisation of antiproliferative O, N-functionalised isopulegol derivatives. RSC Adv 2024; 14:18508-18518. [PMID: 38867736 PMCID: PMC11168086 DOI: 10.1039/d4ra03467h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 06/02/2024] [Indexed: 06/14/2024] Open
Abstract
Benzylation of isopulegol furnished O-benzyl-protected isopulegol, which was transformed into aminodiols via epoxidation followed by ring opening of the corresponding epoxides and subsequent hydrogenolysis. On the other hand, (-)-isopulegol was oxidised to a diol, which was then converted into dibenzyl-protected diol derivatives. The products were then transformed into aminotriols by using a similar method. The antiproliferative activity of aminodiol and aminotriol derivatives was examined. In addition, structure-activity relationships were also explored from the aspects of substituent effects and stereochemistry on the aminodiol and aminotriol systems. The drug-likeness of the compounds was assessed by in silico and experimental physicochemical characterisations, completed by kinetic aqueous solubility and in vitro intestinal-specific parallel artificial membrane permeability assay (PAMPA-GI) measurements.
Collapse
Affiliation(s)
- Tam Minh Le
- Institute of Pharmaceutical Chemistry, University of Szeged Eötvös utca 6 H-6720 Szeged Hungary +36 62 545705 +36 62 546809
- HUN-REN-SZTE Stereochemistry, Research Group, University of Szeged Eötvös u. 6 H-6720 Szeged Hungary
| | - Isaac Kinyua Njangiru
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged H-6720 Eötvös utca 6 Szeged Hungary
| | - Anna Vincze
- Department of Pharmaceutical Chemistry, Semmelweis University Hőgyes Endre u. 9 H-1092 Budapest Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged H-6720 Eötvös utca 6 Szeged Hungary
| | - György T Balogh
- Department of Pharmaceutical Chemistry, Semmelweis University Hőgyes Endre u. 9 H-1092 Budapest Hungary
| | - Zsolt Szakonyi
- Institute of Pharmaceutical Chemistry, University of Szeged Eötvös utca 6 H-6720 Szeged Hungary +36 62 545705 +36 62 546809
| |
Collapse
|
24
|
Klimoszek D, Jeleń M, Dołowy M, Morak-Młodawska B. Study of the Lipophilicity and ADMET Parameters of New Anticancer Diquinothiazines with Pharmacophore Substituents. Pharmaceuticals (Basel) 2024; 17:725. [PMID: 38931392 PMCID: PMC11206290 DOI: 10.3390/ph17060725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 05/28/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Lipophilicity is one of the principal parameters that describe the pharmacokinetic behavior of a drug, including its absorption, distribution, metabolism, elimination, and toxicity. In this study, the lipophilicity and other physicochemical, pharmacokinetic, and toxicity properties that affect the bioavailability of newly synthesized dialkylaminoalkyldiquinothiazine hybrids as potential drug candidates are presented. The lipophilicity, as RM0, was determined experimentally by the RP-TLC method using RP18 plates and acetone-TRIS buffer (pH 7.4) as the mobile phase. The chromatographic parameters of lipophilicity were compared to computationally calculated partition coefficients obtained by various types of programs such as iLOGP, XLOGP3, WLOGP, MLOGP, SILCOS-IT, LogP, logP, and milogP. In addition, the selected ADMET parameters were determined in silico using the SwissADME and pkCSM platforms and correlated with the experimental lipophilicity descriptors. The results of the lipophilicity study confirm that the applied algorithms can be useful for the rapid prediction of logP values during the first stage of study of the examined drug candidates. Of all the algorithms used, the biggest similarity to the chromatographic value (RM0) for certain compounds was seen with iLogP. It was found that both the SwissADME and pkCSM web tools are good sources of a wide range of ADMET parameters that describe the pharmacokinetic profiles of the studied compounds and can be fast and low-cost tools in the evaluation of examined drug candidates during the early stages of the development process.
Collapse
Affiliation(s)
- Daria Klimoszek
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland; (D.K.); (M.D.)
| | - Małgorzata Jeleń
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland;
| | - Małgorzata Dołowy
- Department of Analytical Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland; (D.K.); (M.D.)
| | - Beata Morak-Młodawska
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, Jagiellońska Street 4, 41-200 Sosnowiec, Poland;
| |
Collapse
|
25
|
Morrison KM, Stradiotto M. The development of cage phosphine 'DalPhos' ligands to enable nickel-catalyzed cross-couplings of (hetero)aryl electrophiles. Chem Sci 2024; 15:7394-7407. [PMID: 38784740 PMCID: PMC11110136 DOI: 10.1039/d4sc01253d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Nickel-catalyzed cross-couplings of (hetero)aryl electrophiles with a diversity of nucleophiles (nitrogen, oxygen, carbon, and others) have evolved into competitive alternatives to well-established palladium- and copper-based protocols for the synthesis of (hetero)aryl products, including (hetero)anilines and (hetero)aryl ethers. A survey of the literature reveals that the use of cage phosphine (CgP) 'DalPhos' (DALhousie PHOSphine) bisphosphine-type ligands operating under thermal conditions currently offers the most broad substrate scope in nickel-catalyzed cross-couplings of this type, especially involving (hetero)aryl chlorides and phenol-derived electrophiles. The development and application of these DalPhos ligands is described in a ligand-specific manner that is intended to serve as a guide for the synthetic chemistry end-user.
Collapse
Affiliation(s)
- Kathleen M Morrison
- Department of Chemistry, Dalhousie University 6274 Coburg Road, P.O. 15000 Halifax Nova Scotia B3H 4R2 Canada
| | - Mark Stradiotto
- Department of Chemistry, Dalhousie University 6274 Coburg Road, P.O. 15000 Halifax Nova Scotia B3H 4R2 Canada
| |
Collapse
|
26
|
Mihajlović E, Biancalana L, Jelača S, Chiaverini L, Dojčinović B, Dunđerović D, Zacchini S, Mijatović S, Maksimović-Ivanić D, Marchetti F. FETPY: a Diiron(I) Thio-Carbyne Complex with Prominent Anticancer Activity In Vitro and In Vivo. J Med Chem 2024; 67:7553-7568. [PMID: 38639401 DOI: 10.1021/acs.jmedchem.4c00377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
FETPY, an organo-diiron(I) complex, showed strong cytotoxicity across a panel of human and mouse cancer cell lines, combined with an outstanding selectivity compared to nonmalignant cells. Enhanced iron uptake in aggressive, low-differentiated cell lines, caused membrane lipid peroxidation, which resulted in ferroptosis in human ovarian cancer cells. FETPY induced significant morphological changes in murine B16-F1 and B16-F10 melanoma cells, leading to senescence and/or trans-differentiation into Schwann-like cells, thus significantly reducing their tumorigenic potential. Additionally, FETPY substantially suppressed tumor growth in low- and high-grade syngeneic melanoma models when administered in a therapeutic regimen. FETPY is featured by satisfactory water solubility (millimolar range), an amphiphilic character (Log Pow = -0.17), and excellent stability in a biological medium (DMEM). These important requisites for drug development are rarely met in iron complexes investigated so far as possible anticancer agents. Overall, FETPY holds promise as a safe and potent targeted antitumor agent.
Collapse
Affiliation(s)
- Ekatarina Mihajlović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11108, Serbia
| | - Lorenzo Biancalana
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa I-56124, Italy
| | - Sanja Jelača
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11108, Serbia
| | - Lorenzo Chiaverini
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa I-56124, Italy
| | - Biljana Dojčinović
- Institute of Chemistry, Technology and Metallurgy University of Belgrade, Njegoševa 12, Belgrade 11000, Serbia
| | - Duško Dunđerović
- Institute of Pathology, School of Medicine University of Belgrade, dr Subotića 1, Belgrade 11000, Serbia
| | - Stefano Zacchini
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, Via P. Gobetti 85, Bologna I-40129, Italy
| | - Sanja Mijatović
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11108, Serbia
| | - Danijela Maksimović-Ivanić
- Department of Immunology, Institute for Biological Research "Siniša Stanković" - National Institute of the Republic of Serbia, University of Belgrade, Belgrade 11108, Serbia
| | - Fabio Marchetti
- Department of Chemistry and Industrial Chemistry, University of Pisa, Via Giuseppe Moruzzi 13, Pisa I-56124, Italy
| |
Collapse
|
27
|
Lucas SCC, Ahmed A, Ashraf SN, Argyrou A, Bauer MR, De Donatis GM, Demanze S, Eisele F, Fusani L, Hock A, Kadamur G, Li S, Macmillan-Jones A, Michaelides IN, Phillips C, Rehnström M, Richter M, Rodrigo-Brenni MC, Shilliday F, Wang P, Storer RI. Optimization of Potent Ligands for the E3 Ligase DCAF15 and Evaluation of Their Use in Heterobifunctional Degraders. J Med Chem 2024; 67:5538-5566. [PMID: 38513086 DOI: 10.1021/acs.jmedchem.3c02136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
Unlocking novel E3 ligases for use in heterobifunctional PROTAC degraders is of high importance to the pharmaceutical industry. Over-reliance on the current suite of ligands used to recruit E3 ligases could limit the potential of their application. To address this, potent ligands for DCAF15 were optimized using cryo-EM supported, structure-based design to improve on micromolar starting points. A potent binder, compound 24, was identified and subsequently conjugated into PROTACs against multiple targets. Following attempts on degrading a number of proteins using DCAF15 recruiting PROTACs, only degradation of BRD4 was observed. Deconvolution of the mechanism of action showed that this degradation was not mediated by DCAF15, thereby highlighting both the challenges faced when trying to expand the toolbox of validated E3 ligase ligands for use in PROTAC degraders and the pitfalls of using BRD4 as a model substrate.
Collapse
Affiliation(s)
- Simon C C Lucas
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Afshan Ahmed
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - S Neha Ashraf
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Argyrides Argyrou
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Matthias R Bauer
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | | | - Sylvain Demanze
- Oncology Chemistry, Oncology R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Frederik Eisele
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83,Sweden
| | - Lucia Fusani
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Andreas Hock
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Ganesh Kadamur
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Shuyou Li
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, People's Republic of China
| | | | | | - Christopher Phillips
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Marie Rehnström
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Gothenburg SE-431 83, Sweden
| | - Magdalena Richter
- Discovery Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Monica C Rodrigo-Brenni
- Safety Innovation and PROTAC Safety, Clinical Pharmacology & Safety Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Fiona Shilliday
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| | - Peng Wang
- Pharmaron Beijing Co., Ltd., 6 Taihe Road BDA, Beijing 100176, People's Republic of China
| | - R Ian Storer
- Hit Discovery, Discovery Sciences, R&D, AstraZeneca, Cambridge CB2 0AA, U.K
| |
Collapse
|
28
|
Mambwe D, Coertzen D, Leshabane M, Mulubwa M, Njoroge M, Gibhard L, Girling G, Wicht KJ, Lee MCS, Wittlin S, Moreira DRM, Birkholtz LM, Chibale K. hERG, Plasmodium Life Cycle, and Cross Resistance Profiling of New Azabenzimidazole Analogues of Astemizole. ACS Med Chem Lett 2024; 15:463-469. [PMID: 38628794 PMCID: PMC11017395 DOI: 10.1021/acsmedchemlett.3c00496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 03/01/2024] [Accepted: 03/06/2024] [Indexed: 04/19/2024] Open
Abstract
Toward addressing the cardiotoxicity liability associated with the antimalarial drug astemizole (AST, hERG IC50 = 0.0042 μM) and its derivatives, we designed and synthesized analogues based on compound 1 (Pf NF54 IC50 = 0.012 μM; hERG IC50 = 0.63 μM), our previously identified 3-trifluoromethyl-1,2,4-oxadiazole AST analogue. Compound 11 retained in vitro multistage antiplasmodium activity (ABS PfNF54 IC50 = 0.017 μM; gametocytes PfiGc/PfLGc IC50 = 1.24/1.39 μM, and liver-stage PbHepG2 IC50 = 2.30 μM), good microsomal metabolic stability (MLM CLint < 11 μL·min-1·mg-1, EH < 0.33), and solubility (150 μM). It shows a ∼6-fold and >6000-fold higher selectivity against human ether-á-go-go-related gene higher selectively potential over hERG relative to 1 and AST, respectively. Despite the excellent in vitro antiplasmodium activity profile, in vivo efficacy in the Plasmodium berghei mouse infection model was diminished, attributable to suboptimal oral bioavailability (F = 14.9%) at 10 mg·kg-1 resulting from poor permeability (log D7.4 = -0.82). No cross-resistance was observed against 44 common Pf mutant lines, suggesting activity via a novel mechanism of action.
Collapse
Affiliation(s)
- Dickson Mambwe
- Department
of Chemistry, University of Cape Town, Rondebosch, 7701 Cape Town, South Africa
| | - Dina Coertzen
- Department
of Biochemistry, Genetics & Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield, 0028 Pretoria, South Africa
| | - Meta Leshabane
- Department
of Biochemistry, Genetics & Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield, 0028 Pretoria, South Africa
| | - Mwila Mulubwa
- Drug
Discovery and Development Centre (H3D), DMPK & Pharmacology, University of Cape Town, Observatory, 7925 Cape Town, South Africa
| | - Mathew Njoroge
- Drug
Discovery and Development Centre (H3D), DMPK & Pharmacology, University of Cape Town, Observatory, 7925 Cape Town, South Africa
| | - Liezl Gibhard
- Drug
Discovery and Development Centre (H3D), DMPK & Pharmacology, University of Cape Town, Observatory, 7925 Cape Town, South Africa
| | - Gareth Girling
- Wellcome
Sanger Institute, Wellcome
Trust Genome Campus, Hinxton CB10 1SA, United Kingdom
| | - Kathryn J. Wicht
- Department
of Chemistry, University of Cape Town, Rondebosch, 7701 Cape Town, South Africa
| | - Marcus C. S. Lee
- Wellcome
Sanger Institute, Wellcome
Trust Genome Campus, Hinxton CB10 1SA, United Kingdom
- Biological
Chemistry and Drug Discovery, School of Life Sciences, University of Dundee, Dundee DD1 4HN, Scotland, United Kingdom
| | - Sergio Wittlin
- Swiss
Tropical and Public Health Institute, Socinstrasse 57, 4002 Basel, Switzerland
- University
of Basel, 4003 Basel, Switzerland
| | | | - Lyn-Marie Birkholtz
- Department
of Biochemistry, Genetics & Microbiology, Institute for Sustainable
Malaria Control, University of Pretoria, Private Bag X20, Hatfield, 0028 Pretoria, South Africa
| | - Kelly Chibale
- Department
of Chemistry, University of Cape Town, Rondebosch, 7701 Cape Town, South Africa
- Drug
Discovery and Development Centre (H3D), DMPK & Pharmacology, University of Cape Town, Observatory, 7925 Cape Town, South Africa
- South
African Medical Research Council Drug Discovery and Development Research
Unit, University of Cape Town, Rondebosch, 7701 Cape Town, South Africa
- Institute
of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, 7701 Cape Town, South Africa
| |
Collapse
|
29
|
Pippione AC, Kovachka S, Vigato C, Bertarini L, Mannella I, Sainas S, Rolando B, Denasio E, Piercy-Mycock H, Romalho L, Salladini E, Adinolfi S, Zonari D, Peraldo-Neia C, Chiorino G, Passoni A, Mirza OA, Frydenvang K, Pors K, Lolli ML, Spyrakis F, Oliaro-Bosso S, Boschi D. Structure-guided optimization of 3-hydroxybenzoisoxazole derivatives as inhibitors of Aldo-keto reductase 1C3 (AKR1C3) to target prostate cancer. Eur J Med Chem 2024; 268:116193. [PMID: 38364714 DOI: 10.1016/j.ejmech.2024.116193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/16/2024] [Accepted: 01/25/2024] [Indexed: 02/18/2024]
Abstract
AKR1C3 is an enzyme that is overexpressed in several types of radiotherapy- and chemotherapy-resistant cancers. Despite AKR1C3 is a validated target for drug development, no inhibitor has been approved for clinical use. In this manuscript, we describe our study of a new series of potent AKR1C3-targeting 3-hydroxybenzoisoxazole based inhibitors that display high selectivity over the AKR1C2 isoform and low micromolar activity in inhibiting 22Rv1 prostate cancer cell proliferation. In silico studies suggested proper substituents to increase compound potency and provided with a mechanistic explanation that could clarify their different activity, later confirmed by X-ray crystallography. Both the in-silico studies and the crystallographic data highlight the importance of 90° rotation around the single bond of the biphenyl group, in ensuring that the inhibitor can adopt the optimal binding mode within the active pocket. The p-biphenyls that bear the meta-methoxy, and the ortho- and meta-trifluoromethyl substituents (in compounds 6a, 6e and 6f respectively) proved to be the best contributors to cellular potency as they provided the best IC50 values in series (2.3, 2.0 and 2.4 μM respectively) and showed no toxicity towards human MRC-5 cells. Co-treatment with scalar dilutions of either compound 6 or 6e and the clinically used drug abiraterone led to a significant reduction in cell proliferation, and thus confirmed that treatment with both CYP171A1-and AKR1C3-targeting compounds possess the potential to intervene in key steps in the steroidogenic pathway. Taken together, the novel compounds display desirable biochemical potency and cellular target inhibition as well as good in-vitro ADME properties, which highlight their potential for further preclinical studies.
Collapse
Affiliation(s)
- Agnese Chiara Pippione
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Sandra Kovachka
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy; The Herbert Wertheim UF Scripps Institute for Biomedical Innovation & Technology, Jupiter, FL, USA
| | - Chiara Vigato
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Laura Bertarini
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy; Department of Life Sciences, University of Modena and Reggio Emilia, via Campi 103, 41125, Modena, Italy
| | - Iole Mannella
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Stefano Sainas
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Barbara Rolando
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Enrica Denasio
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, West Yorkshire, BD7 1DP, UK
| | - Helen Piercy-Mycock
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, West Yorkshire, BD7 1DP, UK
| | - Linda Romalho
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Edoardo Salladini
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Salvatore Adinolfi
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Daniele Zonari
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Caterina Peraldo-Neia
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Giovanna Chiorino
- Laboratory of Cancer Genomics, Fondazione Edo ed Elvo Tempia, via Malta 3, 13900, Biella, Italy
| | - Alice Passoni
- Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Via Mario Negri 2, 20156, Milan, Italy
| | - Osman Asghar Mirza
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Karla Frydenvang
- Department of Drug Design and Pharmacology, University of Copenhagen, Jagtvej 162, DK-2100, Copenhagen, Denmark
| | - Klaus Pors
- Institute of Cancer Therapeutics, Faculty of Life Sciences, University of Bradford, West Yorkshire, BD7 1DP, UK
| | - Marco Lucio Lolli
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Francesca Spyrakis
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy
| | - Simonetta Oliaro-Bosso
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy.
| | - Donatella Boschi
- Department of Science and Drug Technology, University of Turin, via Pietro Giuria 9, 10125, Turin, Italy.
| |
Collapse
|
30
|
Krámos B, Hadady Z, Makó A, Szántó G, Felföldi N, Magdó I, Bobok AÁ, Bata I, Román V, Visegrády A, Keserű G, Greiner I, Éles J. Novel-Type GABA B PAMs: Structure-Activity Relationship in Light of the Protein Structure. ACS Med Chem Lett 2024; 15:396-405. [PMID: 38505850 PMCID: PMC10945541 DOI: 10.1021/acsmedchemlett.3c00560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/16/2024] [Accepted: 02/20/2024] [Indexed: 03/21/2024] Open
Abstract
Selecting a known HTS hit with the pyrazolo[1,5-a]pyrimidine core, our project was started from CMPPE, and its optimization was driven by a ligand-based pharmacophore model developed on the basis of published GABAB positive allosteric modulators (PAMs). Our primary goal was to improve the potency by finding new enthalpic interactions. Therefore, we included the lipophilic ligand efficiency (LLE or LipE) as an objective function in the optimization that led to a carboxylic acid derivative (34). This lead candidate offers the possibility to improve potency without drastically inflating the physicochemical properties. Although the discovery of the novel carboxyl feature was surprising, it turned out to be an important element of the GABAB PAM pharmacophore that can be perfectly explained based on the new protein structures. Rationalizing the binding mode of 34, we analyzed the intersubunit PAM binding site of GABAB receptor using the publicly available experimental structures.
Collapse
Affiliation(s)
- Balázs Krámos
- Spectroscopic
Research Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Zsuzsa Hadady
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Attila Makó
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Gábor Szántó
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Nóra Felföldi
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Ildikó Magdó
- Spectroscopic
Research Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Amrita Ágnes Bobok
- Pharmacological
and Drug Safety Research, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - Imre Bata
- Chemistry
Department, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| | - Viktor Román
- Pharmacological
and Drug Safety Research, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - András Visegrády
- Pharmacological
and Drug Safety Research, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - György
M. Keserű
- Medicinal
Chemistry Research Group, Research Centre
for Natural Sciences, Magyar tudósok krt. 2, Budapest, 1117 Hungary
| | - István Greiner
- Research
and Development Director, Gedeon Richter
Plc., Gyömrői
út 19-21, Budapest, 1103 Hungary
| | - János Éles
- Head
of Medicinal Chemistry, Gedeon Richter Plc., Gyömrői út
19-21, Budapest, 1103 Hungary
| |
Collapse
|
31
|
Carrillo AK, Kadayat TM, Hwang JY, Chen Y, Zhu F, Holbrook G, Gillingwater K, Connelly MC, Yang L, Kaiser M, Guy RK. Antitrypanosomal Chloronitrobenzamides. J Med Chem 2024; 67:3437-3447. [PMID: 38363074 DOI: 10.1021/acs.jmedchem.3c01680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Human African trypanosomiasis (HAT), a neglected tropical disease caused by Trypanosoma brucei gambiense (Tbg) or Trypanosoma brucei rhodesiense (Tbr), remains a significant public health concern with over 55 million people at risk of infection. Current treatments for HAT face the challenges of poor efficacy, drug resistance, and toxicity. This study presents the synthesis and evaluation of chloronitrobenzamides (CNBs) against Trypanosoma species, identifying previously reported compound 52 as a potent and selective orally bioavailable antitrypanosomal agent. 52 was well tolerated in vivo and demonstrated favorable oral pharmacokinetics, maintaining plasma concentrations surpassing the cellular EC50 for over 24 h and achieving peak brain concentrations exceeding 7 μM in rodents after single oral administration (50 mg/kg). Treatment with 52 significantly extended the lifespan of mice infected with Trypanosoma congolense and T. brucei rhodesiense. These results demonstrate that 52 is a strong antitrypanosomal lead with potential for developing treatments for both human and animal African trypanosomiasis.
Collapse
Affiliation(s)
- Angela K Carrillo
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Tara Man Kadayat
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509, United States
| | - Jong Yeon Hwang
- Korea Research Institute of Chemical Technology, 141 Gajeong-ro, Daejeon, KR 34114, United States
| | - Yizhe Chen
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509, United States
| | - Fangyi Zhu
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Gloria Holbrook
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Kirsten Gillingwater
- Department of Medical Parasitology & Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil 4123, Switzerland
| | - Michele C Connelly
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Lei Yang
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
| | - Marcel Kaiser
- Department of Medical Parasitology & Infection Biology, Swiss Tropical and Public Health Institute, Kreuzstrasse 2, Allschwil 4123, Switzerland
- Faculty of Science, University of Basel, Petersplatz 1, Basel 4003, Switzerland
| | - R Kiplin Guy
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, United States
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536-0509, United States
| |
Collapse
|
32
|
Thoma G, Miltz W, Srinivas H, Penno CA, Kiffe M, Gajewska M, Klein K, Evans A, Beerli C, Röhn TA. Structure-Guided Elaboration of a Fragment-Like Hit into an Orally Efficacious Leukotriene A4 Hydrolase Inhibitor. J Med Chem 2024. [PMID: 38476002 DOI: 10.1021/acs.jmedchem.4c00290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Leukotriene A4 hydrolase (LTA4H) is the final and rate-limiting enzyme in the biosynthesis of pro-inflammatory leukotriene B4 (LTB4). Preclinical studies have provided strong evidence that LTA4H is an attractive drug target for the treatment of chronic inflammatory diseases. Here, we describe the transformation of compound 2, a fragment-like hit, into the potent inhibitor of LTA4H 3. Our strategy involved two key steps. First, we aimed to increase the polarity of fragment 2 to improve its drug-likeness, particularly its solubility, while preserving both its promising potency and low molecular weight. Second, we utilized structural information and incorporated a basic amino function, which allowed for the formation of an essential hydrogen bond with Q136 of LTA4H and consequently enhanced the potency. Compound 3 exhibited exceptional selectivity and showed oral efficacy in a KRN passive serum-induced arthritis model in mice. The anticipated human dose to achieve 90% target engagement at the trough concentration was determined to be 40 mg administered once daily.
Collapse
Affiliation(s)
- Gebhard Thoma
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Wolfgang Miltz
- Global Discovery Chemistry, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Honnappa Srinivas
- Discovery Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Carlos A Penno
- Discovery Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Michael Kiffe
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Monika Gajewska
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Kai Klein
- PK Sciences, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Amanda Evans
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Christian Beerli
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| | - Till A Röhn
- Immunology Disease Area, Biomedical Research, Novartis Pharma AG, 4002 Basel, Switzerland
| |
Collapse
|
33
|
Kaufman B, Williams EC, Underkoffler C, Pederson R, Mardirossian N, Watson I, Parkhill J. COATI: Multimodal Contrastive Pretraining for Representing and Traversing Chemical Space. J Chem Inf Model 2024; 64:1145-1157. [PMID: 38316665 DOI: 10.1021/acs.jcim.3c01753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2024]
Abstract
Creating a successful small molecule drug is a challenging multiparameter optimization problem in an effectively infinite space of possible molecules. Generative models have emerged as powerful tools for traversing data manifolds composed of images, sounds, and text and offer an opportunity to dramatically improve the drug discovery and design process. To create generative optimization methods that are more useful than brute-force molecular generation and filtering via virtual screening, we propose that four integrated features are necessary: large, quantitative data sets of molecular structure and activity, an invertible vector representation of realistic accessible molecules, smooth and differentiable regressors that quantify uncertainty, and algorithms to simultaneously optimize properties of interest. Over the course of 12 months, Terray Therapeutics has collected a data set of 2 billion quantitative binding measurements of small molecules to therapeutic targets, which directly motivates multiparameter generative optimization of molecules conditioned on these data. To this end, we present contrastive optimization for accelerated therapeutic inference (COATI), a pretrained, multimodal encoder-decoder model of druglike chemical space. COATI is constructed without any human biasing of features, using contrastive learning from text and 3D representations of molecules to allow for downstream use with structural models. We demonstrate that COATI possesses many of the desired properties of universal molecular embedding: fixed-dimension, invertibility, autoencoding, accurate regression, and low computation cost. Finally, we present a novel metadynamics algorithm for generative optimization using a small subset of our proprietary data collected for a model protein, carbonic anhydrase, designing molecules that satisfy the multiparameter optimization task of potency, solubility, and drug likeness. This work sets the stage for fully integrated generative molecular design and optimization for small molecules.
Collapse
Affiliation(s)
- Benjamin Kaufman
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| | - Edward C Williams
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| | - Carl Underkoffler
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| | - Ryan Pederson
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| | - Narbe Mardirossian
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| | - Ian Watson
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| | - John Parkhill
- Terray Therapeutics, Inc., 800 Royal Oaks Dr, Monrovia, California 91016, United States
| |
Collapse
|
34
|
Morozova A, Chan SC, Bayle S, Sun L, Grassie D, Iermolaieva A, Kalaga MN, Frydman S, Sansil S, Schönbrunn E, Duckett D, Monastyrskyi A. Development of potent and selective ULK1/2 inhibitors based on 7-azaindole scaffold with favorable in vivo properties. Eur J Med Chem 2024; 266:116101. [PMID: 38232465 DOI: 10.1016/j.ejmech.2023.116101] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/19/2024]
Abstract
The UNC-51-like kinase-1 (ULK1) is one of the central upstream regulators of the autophagy pathway, represents a key target for the development of molecular probes to abrogate autophagy and explore potential therapeutic avenues. Here we report the discovery, structure-activity and structure-property relationships of selective, potent, and cell-active ULK1/2 inhibitors based on a 7-azaindole scaffold. Using structure-based drug design, we have developed a series of analogs with excellent binding affinity and biochemical activity against ULK1/2 (IC50 < 25 nM). The validation of cellular target engagement for these compounds was achieved through the employment of the ULK1 NanoBRET intracellular kinase assay. Notably, we have successfully solved the crystal structure of the lead compound, MR-2088, bound to the active site of ULK1. Moreover, the combination treatment of MR-2088 with known KRAS→RAF→MEK→ERK pathway inhibitors, such as trametinib, showed promising synergistic effect in vitro using H2030 (KRASG12C) cell lines. Lastly, our findings underscore MR-2088's potential to inhibit starvation/stimuli-induced autophagic flux, coupled with its suitability for in vivo studies based on its pharmacokinetic properties.
Collapse
Affiliation(s)
- Alisa Morozova
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Sean Chin Chan
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Simon Bayle
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Luxin Sun
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Dylan Grassie
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Anna Iermolaieva
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Mahalakshmi N Kalaga
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Sylvia Frydman
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Samer Sansil
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Ernst Schönbrunn
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Derek Duckett
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States
| | - Andrii Monastyrskyi
- Department of Drug Discovery, H. Lee Moffitt Cancer Center and Research Institute, 12902 USF Magnolia Dr, Tampa, FL, United States.
| |
Collapse
|
35
|
Brocklehurst CE, Altmann E, Bon C, Davis H, Dunstan D, Ertl P, Ginsburg-Moraff C, Grob J, Gosling DJ, Lapointe G, Marziale AN, Mues H, Palmieri M, Racine S, Robinson RI, Springer C, Tan K, Ulmer W, Wyler R. MicroCycle: An Integrated and Automated Platform to Accelerate Drug Discovery. J Med Chem 2024; 67:2118-2128. [PMID: 38270627 DOI: 10.1021/acs.jmedchem.3c02029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2024]
Abstract
We herein describe the development and application of a modular technology platform which incorporates recent advances in plate-based microscale chemistry, automated purification, in situ quantification, and robotic liquid handling to enable rapid access to high-quality chemical matter already formatted for assays. In using microscale chemistry and thus consuming minimal chemical matter, the platform is not only efficient but also follows green chemistry principles. By reorienting existing high-throughput assay technology, the platform can generate a full package of relevant data on each set of compounds in every learning cycle. The multiparameter exploration of chemical and property space is hereby driven by active learning models. The enhanced compound optimization process is generating knowledge for drug discovery projects in a time frame never before possible.
Collapse
Affiliation(s)
- Cara E Brocklehurst
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Eva Altmann
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Corentin Bon
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Holly Davis
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - David Dunstan
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Peter Ertl
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Carol Ginsburg-Moraff
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Jonathan Grob
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Daniel J Gosling
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Guillaume Lapointe
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Alexander N Marziale
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Heinrich Mues
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Marco Palmieri
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Sophie Racine
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| | - Richard I Robinson
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Clayton Springer
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - Kian Tan
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - William Ulmer
- Global Discovery Chemistry, Novartis Biomedical Research, Cambridge, Massachusetts 02139, United States
| | - René Wyler
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Basel 4033, Switzerland
| |
Collapse
|
36
|
Pal M, Upadhyay A, Masarkar N, Bera A, Mukherjee S, Roy M. Folate-assisted targeted photocytotoxicity of red-light-activable iron(III) complex co-functionalized gold nanoconjugates (Fe@FA-AuNPs) against HeLa and triple-negative MDA-MB-231 cancer cells. Dalton Trans 2024; 53:2108-2119. [PMID: 38180438 DOI: 10.1039/d3dt03581f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2024]
Abstract
Photo-redox chemistry resulting from ligand to metal charge transfer in red-light-activable iron(III) complexes could be a potent strategic tool for next-generation photochemotherapeutic applications. Herein, we developed an iron(III) complex and folate co-functionalized gold nanoconjugate (Fe@FA-AuNPs) and thoroughly characterized it with NMR, ESI MS, UV-visible, EPR, EDX, XPS, powder X-ray diffraction, TEM and DLS studies. There was a remarkable shift in the SPR band of AuNPs to 680 nm, and singlet oxygen (1O2) and hydroxyl radicals were potently generated upon red-light activation, which were probed by UV-visible and EPR spectroscopic assays. Cellular uptake studies of the nanoconjugate (Fe@FA-AuNPs) revealed significantly higher uptake in folate(+) cancer cells (HeLa and MDA-MB-231) than folate(-) (A549) cancer cells or normal cells (HPL1D), indicating the targeting potential of the nanoconjugate. Confocal imaging indicated primarily mitochondrial localization. The IC50 values of the nanoconjugate determined from a cell viability assay in HeLa, MDA-MB-231, and A549 cells were 27.83, 39.91, and 69.54 μg mL-1, respectively in red light, while in the dark the values were >200 μg mL-1; the photocytotoxicity was correlated with the cellular uptake of the nanoconjugate. The nanocomposite exhibited similar photocytotoxicity (IC50 in red light, 37.35 ± 8.29 μg mL-1 and IC50 in the dark, >200 μg mL-1). Mechanistic studies revealed that intracellular generation of ROS upon red-light activation led to apoptosis in HeLa cells. Scratch-wound-healing assays indicated the inhibition of the migration of MDA-MB-231 cells treated with the nanoconjugate and upon photo-activation. Overall, the nanoconjugate has emerged as a potent tool for next-generation photo-chemotherapeutics in the clinical arena of targeted cancer therapy.
Collapse
Affiliation(s)
- Maynak Pal
- Department of Chemistry, National Institute of Technology Manipur, Langol 795004, Imphal (Manipur), India.
| | - Aarti Upadhyay
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore-560012, India
| | - Neha Masarkar
- Department of Biochemistry, All India Institute of Medical Science (AIIMS), Saket Nagar, Bhopal, Madhya Pradesh, 462026, India
| | - Arpan Bera
- Department of Inorganic and Physical Chemistry, Indian Institute of Science, Bangalore-560012, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Science (AIIMS), Saket Nagar, Bhopal, Madhya Pradesh, 462026, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol 795004, Imphal (Manipur), India.
| |
Collapse
|
37
|
Fleetwood TD, Kerr WJ, Mason J. Copper-Mediated N-Trifluoromethylation of O-Benzoylhydroxylamines. Chemistry 2024; 30:e202303314. [PMID: 38018464 PMCID: PMC10952365 DOI: 10.1002/chem.202303314] [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: 10/09/2023] [Revised: 11/24/2023] [Accepted: 11/28/2023] [Indexed: 11/30/2023]
Abstract
The use of trifluoromethyl containing compounds is well established within medicinal chemistry, with a range of approved drugs containing C-CF3 and O-CF3 moieties. However, the utilisation of the N-CF3 functional group remains relatively unexplored. This may be attributed to the challenging synthesis of this unit, with many current methods employing harsh conditions or less accessible reagents. A robust methodology for the N-trifluoromethylation of secondary amines has been developed, which employs an umpolung strategy in the form of a copper-catalysed electrophilic amination. The method is operationally simple, uses mild, inexpensive reagents, and has been used to synthesise a range of novel, structurally complex N-CF3 containing compounds.
Collapse
Affiliation(s)
- Thomas D. Fleetwood
- Medicinal ChemistryGSK Medicines Research CentreGunnels Wood RoadSG1 2NYStevenageEnglandU.K.
- Department of Pure and Applied ChemistryUniversity of StrathclydeG1 1XLGlasgowScotlandU.K.
| | - William J. Kerr
- Department of Pure and Applied ChemistryUniversity of StrathclydeG1 1XLGlasgowScotlandU.K.
| | - Joseph Mason
- Medicinal ChemistryGSK Medicines Research CentreGunnels Wood RoadSG1 2NYStevenageEnglandU.K.
| |
Collapse
|
38
|
Miyajima Y, Noguchi-Yachide T, Ochiai K, Fujii S. Physicochemical characterization of B-hydroxyphenyl phosphine borane derivatives and their evaluation as nuclear estrogen receptor ligands. RSC Med Chem 2024; 15:119-126. [PMID: 38283218 PMCID: PMC10809333 DOI: 10.1039/d3md00350g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/27/2023] [Indexed: 01/30/2024] Open
Abstract
Increasing the structural options in medicinal chemistry is a promising approach to develop new drug candidates. In this research, we designed and synthesized a series of B-hydroxyphenyl phosphine borane derivatives and investigated their structure-property and structure-activity relationships. The synthesized B-phenylphosphine borane derivatives exhibited sufficient stability in aqueous media, weaker hydrophobicity than the corresponding alkanes and silanes, and sufficient affinity for lipid membranes to enable permeability. Several B-hydroxyphenyl phosphine borane derivatives exhibited significant estrogen receptor (ER) agonistic activity with superior ligand-lipophilicity efficiency (LLE). The phosphine borane framework appears to be a promising option for structural development in drug discovery studies.
Collapse
Affiliation(s)
- Yu Miyajima
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University 2-3-10 Kanda-Surugadai Chiyoda-ku Tokyo 101-0062 Japan
| | - Tomomi Noguchi-Yachide
- Institute for Quantitative Biosciences, The University of Tokyo 1-1-1 Yayoi Bunkyo-ku Tokyo 113-0032 Japan
| | - Kotaro Ochiai
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University 2-3-10 Kanda-Surugadai Chiyoda-ku Tokyo 101-0062 Japan
| | - Shinya Fujii
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University 2-3-10 Kanda-Surugadai Chiyoda-ku Tokyo 101-0062 Japan
| |
Collapse
|
39
|
Takarada JE, Cunha MR, Almeida VM, Vasconcelos SNS, Santiago AS, Godoi PH, Salmazo A, Ramos PZ, Fala AM, de Souza LR, Da Silva IEP, Bengtson MH, Massirer KB, Couñago RM. Discovery of pyrazolo[3,4-d]pyrimidines as novel mitogen-activated protein kinase kinase 3 (MKK3) inhibitors. Bioorg Med Chem 2024; 98:117561. [PMID: 38157838 DOI: 10.1016/j.bmc.2023.117561] [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: 10/14/2023] [Revised: 12/06/2023] [Accepted: 12/17/2023] [Indexed: 01/03/2024]
Abstract
The dual-specificity protein kinase MKK3 has been implicated in tumor cell proliferation and survival, yet its precise role in cancer remains inconclusive. A critical step in elucidating the kinase's involvement in disease biology is the identification of potent, cell-permeable kinase inhibitors. Presently, MKK3 lacks a dedicated tool compound for these purposes, along with validated methods for the facile screening, identification, and optimization of inhibitors. In this study, we have developed a TR-FRET-based enzymatic assay for the detection of MKK3 activity in vitro and a BRET-based assay to assess ligand binding to this enzyme within intact human cells. These assays were instrumental in identifying hit compounds against MKK3 that share a common chemical scaffold, sourced from a library of bioactive kinase inhibitors. Initial hits were subsequently expanded through the synthesis of novel analogs. The resulting structure-activity relationship (SAR) was rationalized using molecular dynamics simulations against a homology model of MKK3. We expect our findings to expedite the development of novel, potent, selective, and bioactive inhibitors, thus facilitating investigations into MKK3's role in various cancers.
Collapse
Affiliation(s)
- Jéssica E Takarada
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Micael R Cunha
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Vitor M Almeida
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Stanley N S Vasconcelos
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - André S Santiago
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Paulo H Godoi
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Anita Salmazo
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Priscila Z Ramos
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Angela M Fala
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Lucas R de Souza
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Italo E P Da Silva
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP 13083-862, Brazil
| | - Mario H Bengtson
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil; Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas (UNICAMP), Campinas, SP 13083-862, Brazil
| | - Katlin B Massirer
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil
| | - Rafael M Couñago
- Center of Medicinal Chemistry (CQMED), Center for Molecular Biology and Genetic Engineering (CBMEG), University of Campinas, Av. Dr. André Tosello 550, 13083-886 Campinas, Brazil; Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, United States.
| |
Collapse
|
40
|
Kavyani B, Saffari F, Afgar A, Kavyani S, Rezaie M, Sharifi F, Ahmadrajabi R. Gallocin-derived Engineered Peptides Targeting EGFR and VEGFR in Colorectal Cancer: A Bioinformatic Approach. Curr Top Med Chem 2024; 24:1599-1614. [PMID: 38840394 DOI: 10.2174/0115680266295587240522050712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/11/2024] [Accepted: 04/16/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Colorectal cancer (CRC) treatment using time-saving and cost-effective targeted therapies with high selectivity and low toxicity drugs, is a great challenge. In primary investigations on Gallocin, as the most proposed factor in CRC pathogenesis caused by Streptococcus gallolyticus, it was surprisingly found that this bacteriocin has four α-helix structures and some anti-cancer sequences. OBJECTIVE The aim of this study was to determine the ability of Gallocin-based anticancer peptides (ACPs) against epidermal growth factor receptor (EGFR) and vascular epidermal growth factor receptor (VEGFR) and the evaluation of their pharmacokinetic properties using bioinformatic approaches. METHODS Support vector machine algorithm web-based tools were used for predicting ACPs. The physicochemical characteristics and the potential of anti-cancer activity of Gallocin-derived ACPs were determined by in silico tools. The 3D structure of predicted ACPs was modeled using modeling tools. The interactions between predicted ACPs and targets were investigated by molecular docking exercises. Then, the stability of ligand-receptor interactions was determined by molecular dynamic simulation. Finally, ADMET analysis was carried out to check the pharmacokinetic properties and toxicity of ACPs. RESULTS Four amino acid sequences with anti-cancer potential were selected. Through molecular docking, Pep2, and Pep3 gained the best scores, more binding affinity, and strong attachments by the formation of reasonable H-bonds with both EGFR and VEGFR. Molecular simulation confirmed the stability of Pep3- EGFR. According to pharmacokinetic analysis, the ACPs were safe and truthful. CONCLUSION Designed peptides can be nominated as drugs for CRC treatment. However, different in-vitro and in-vivo assessments are required to approve this claim.
Collapse
Affiliation(s)
- Batoul Kavyani
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fereshteh Saffari
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Ali Afgar
- Research Center for Hydatid Disease in Iran, Kerman University of Medical Sciences, Kerman, Iran
| | - Sajjad Kavyani
- Department of Chemical and Materials Engineering, University of Alberta, Edmonton, Alberta, T6G 1H9, Canada
| | - Masoud Rezaie
- Student Research Committee, Afzalipour School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Fatemeh Sharifi
- Research Center of Tropical and Infectious Diseases, Kerman University of Medical Sciences, Kerman, Iran
| | - Roya Ahmadrajabi
- Department of Medical Microbiology (Bacteriology and Virology), Afzalipour Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| |
Collapse
|
41
|
Menke MJ, Schneider P, Badenhorst CPS, Kunzendorf A, Heinz F, Dörr M, Hayes MA, Bornscheuer UT. A Universal, Continuous Assay for SAM-dependent Methyltransferases. Angew Chem Int Ed Engl 2023; 62:e202313912. [PMID: 37917964 DOI: 10.1002/anie.202313912] [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: 09/21/2023] [Revised: 10/30/2023] [Accepted: 11/02/2023] [Indexed: 11/04/2023]
Abstract
Enzyme-catalyzed late-stage functionalization (LSF), such as methylation of drug molecules and lead structures, enables direct access to more potent active pharmaceutical ingredients (API). S-adenosyl-l-methionine-dependent methyltransferases (MTs) can play a key role in the development of new APIs, as they catalyze the chemo- and regioselective methylation of O-, N-, S- and C-atoms, being superior to traditional chemical routes. To identify suitable MTs, we developed a continuous fluorescence-based, high-throughput assay for SAM-dependent methyltransferases, which facilitates screening using E. coli cell lysates. This assay involves two enzymatic steps for the conversion of S-adenosyl-l-homocysteine into H2 S to result in a selective fluorescence readout via reduction of an azidocoumarin sulfide probe. Investigation of two O-MTs and an N-MT confirmed that this assay is suitable for the determination of methyltransferase activity in E. coli cell lysates.
Collapse
Affiliation(s)
- Marian J Menke
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Pascal Schneider
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Christoffel P S Badenhorst
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Andreas Kunzendorf
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Florian Heinz
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Mark Dörr
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| | - Martin A Hayes
- Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Uwe T Bornscheuer
- Department of Biotechnology and Enzyme Catalysis, Institute of Biochemistry, University of Greifswald, Felix-Hausdorff-Str. 4, 17487, Greifswald, Germany
| |
Collapse
|
42
|
Bradley E, Fusani L, Chung CW, Craggs PD, Demont EH, Humphreys PG, Mitchell DJ, Phillipou A, Rioja I, Shah RR, Wellaway CR, Prinjha RK, Palmer DS, Kerr WJ, Reid M, Wall ID, Cookson R. Structure-Guided Design of a Domain-Selective Bromodomain and Extra Terminal N-Terminal Bromodomain Chemical Probe. J Med Chem 2023; 66:15728-15749. [PMID: 37967462 PMCID: PMC10726358 DOI: 10.1021/acs.jmedchem.3c00906] [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: 05/19/2023] [Revised: 08/11/2023] [Accepted: 10/26/2023] [Indexed: 11/17/2023]
Abstract
Small-molecule-mediated disruption of the protein-protein interactions between acetylated histone tails and the tandem bromodomains of the bromodomain and extra-terminal (BET) family of proteins is an important mechanism of action for the potential modulation of immuno-inflammatory and oncology disease. High-quality chemical probes have proven invaluable in elucidating profound BET bromodomain biology, with seminal publications of both pan- and domain-selective BET family bromodomain inhibitors enabling academic and industrial research. To enrich the toolbox of structurally differentiated N-terminal bromodomain (BD1) BET family chemical probes, this work describes an analysis of the GSK BRD4 bromodomain data set through a lipophilic efficiency lens, which enabled identification of a BD1 domain-biased benzimidazole series. Structure-guided growth targeting a key Asp/His BD1/BD2 switch enabled delivery of GSK023, a high-quality chemical probe with 300-1000-fold BET BD1 domain selectivity and a phenotypic cellular fingerprint consistent with BET bromodomain inhibition.
Collapse
Affiliation(s)
- Erin Bradley
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K.
| | - Lucia Fusani
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K.
| | - Chun-wa Chung
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | - Peter D. Craggs
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | | | | | | | - Alex Phillipou
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | - Inmaculada Rioja
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | - Rishi R. Shah
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | | | - Rab K. Prinjha
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | - David S. Palmer
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K.
| | - William J. Kerr
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K.
| | - Marc Reid
- Department
of Pure and Applied Chemistry, University
of Strathclyde, Thomas
Graham Building, 295 Cathedral Street, Glasgow G1 1XL, U.K.
| | - Ian D. Wall
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| | - Rosa Cookson
- GSK,
Medicines Research Centre, Stevenage SG1 2NY, Hertfordshire, U.K.
| |
Collapse
|
43
|
Zhao H. Kinetic modelling of the P-glycoprotein mediated efflux with a large-scale matched molecular pair analysis. Eur J Med Chem 2023; 261:115830. [PMID: 37774507 DOI: 10.1016/j.ejmech.2023.115830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/19/2023] [Accepted: 09/21/2023] [Indexed: 10/01/2023]
Abstract
P-glycoprotein (Pgp) mediated efflux impacts on the drug absorption, distribution, metabolism and excretion, and confers multidrug resistance to cancer cells. Kinetic modelling provides mechanistic insights into the relationship between the substrate-Pgp interactions and efflux, and bridges the gap between the preference of polar compounds as Pgp substrates and the hydrophobic nature of its drug-binding site. Matched molecular pair analysis supports the guidelines of controlling H-bond donors and polar surface area in the efflux mitigation, but also reveals insufficiency of this type of rule-based approach. Contrary to the rule-of-five compliant compounds, proteolysis-targeting chimeras (PROTACs) have shown the opposite preference of physicochemical properties to evade efflux. Our analysis reiterates the critical role of intrinsic passive permeability in the efflux ratio, and indeed, its mitigation is often driven by increased passive permeability. It is thus useful to separate the passive permeability from the structural context-specific substrate-Pgp interactions in the design cycle.
Collapse
Affiliation(s)
- Hongtao Zhao
- Medicinal Chemistry, Research and Early Development, Respiratory and Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden.
| |
Collapse
|
44
|
Pervez MN, Mishu MMR, Tanvir NP, Talukder ME, Cai Y, Telegin FY, Zhao Y, Naddeo V. Insights into the structures and properties of dyes in the Fenton catalytic process for treating wastewater effluent. WATER ENVIRONMENT RESEARCH : A RESEARCH PUBLICATION OF THE WATER ENVIRONMENT FEDERATION 2023; 95:e10948. [PMID: 38062884 DOI: 10.1002/wer.10948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/19/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023]
Abstract
A notable level of apprehension exists over the adverse impacts of dye pollution on aquatic ecosystems and human well-being. The primary objective of this research is to assess the effectiveness of Fenton catalytic reactions in degrading 14 different commercial azo dyes (both single and double) present in aqueous solutions. The investigation focused on the function of dye structures, using a combination of experimental data and examination of theoretical factors. Dye degradation process was carried out at pH 3, and the concentrations of Fe2+ (10-4 mol/L), H2 O2 (2 × 10-3 mol/L), and dye (0.05 g/L). The findings revealed that dyes with a larger molecular weight were more effective at degrading (D%), with the overall degradation efficiency varying from 0% to 94%. Functional groups played an important role in degradation efficiency; for example, dyes with higher aromatic rings led to less D%, while a higher number of sulfonic, methyl, and nitro groups was responsible for better D%. Notably, the presence of OH groups in the backbone of dyes (AB 24, ABE 113, and MB 9) formed the Fe complex during the catalytic process, and the D% was minimal. On the other hand, theoretical quantum calculations such as the greater the JCLogP, highest occupied molecular orbital, and Dipole moment value, the higher the degradation efficiency. And dyes with low lowest unoccupied molecular orbital tended to have a better degradation efficiency. To some extent, UV-Vis spectral analysis was investigated to determine the degradation pathway, and the pseudo-second-order kinetic model fitted better in the degradation process. The overall experimental and theoretical findings suggested that dye degradation efficiency by the Fenton process is structure-dependent. PRACTITIONER POINTS: Insights into the role of azo dye structures-properties on degradation efficiency. Higher molecular weight and sulfonic groups containing dyes showed better degradation efficiency. Hydroxyl groups play the formation of the Fe complex during the degradation process. Higher values of HOMO and lower values of LUMO enhanced degradation efficiency. The pseudo-second-order (PSO) kinetic model obeyed the Fenton process.
Collapse
Affiliation(s)
- Md Nahid Pervez
- Hubei Provincial Engineering Laboratory for Clean Production and High Value Utilization of Bio-based Textile Materials, Wuhan Textile University, Wuhan, China
- Sanitary Environmental Engineering Division (SEED), Department of Civil Engineering, University of Salerno, Fisciano, Italy
| | - Mst Monira Rahman Mishu
- Faculty of Nutrition and Food Science, Patuakhali Science and Technology University, Patuakhali, Bangladesh
| | - Naim Pervez Tanvir
- Department of Chemistry, Patuakhali Govt. College, Patuakhali, Bangladesh
| | - Md Eman Talukder
- Hubei Provincial Engineering Laboratory for Clean Production and High Value Utilization of Bio-based Textile Materials, Wuhan Textile University, Wuhan, China
| | - Yingjie Cai
- Hubei Provincial Engineering Laboratory for Clean Production and High Value Utilization of Bio-based Textile Materials, Wuhan Textile University, Wuhan, China
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, China
| | - Felix Y Telegin
- G.A. Krestov Institute of Solution Chemistry, Russian Academy of Sciences, Ivanovo, Russia
| | - Yaping Zhao
- Shanghai Engineering Research Center of Biotransformation of Organic Solid Waste, School of Ecological and Environmental Sciences, East China Normal University and Institute of Eco-Chongming, Shanghai, China
| | - Vincenzo Naddeo
- Sanitary Environmental Engineering Division (SEED), Department of Civil Engineering, University of Salerno, Fisciano, Italy
| |
Collapse
|
45
|
Cano-González L, Espinosa-Mendoza JD, Matadamas-Martínez F, Romero-Velásquez A, Flores-Ramos M, Colorado-Pablo LF, Cerbón-Cervantes MA, Castillo R, González-Sánchez I, Yépez-Mulia L, Hernández-Campos A, Aguayo-Ortiz R. Structure-Based Optimization of Carbendazim-Derived Tubulin Polymerization Inhibitors through Alchemical Free Energy Calculations. J Chem Inf Model 2023; 63:7228-7238. [PMID: 37947759 DOI: 10.1021/acs.jcim.3c01379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2023]
Abstract
Carbendazim derivatives, commonly used as antiparasitic drugs, have shown potential as anticancer agents due to their ability to induce cell cycle arrest and apoptosis in human cancer cells by inhibiting tubulin polymerization. Crystallographic structures of α/β-tubulin multimers complexed with nocodazole and mebendazole, two carbendazim derivatives with potent anticancer activity, highlighted the possibility of designing compounds that occupy both benzimidazole- and colchicine-binding sites. In addition, previous studies have demonstrated that the incorporation of a phenoxy group at position 5/6 of carbendazim increases the antiproliferative activity in cancer cell lines. Despite the significant progress made in identifying new tubulin-targeting anticancer compounds, further modifications are needed to enhance their potency and safety. In this study, we explored the impact of modifying the phenoxy substitution pattern on antiproliferative activity. Alchemical free energy calculations were used to predict the binding free energy difference upon ligand modification and define the most viable path for structure optimization. Based on these calculations, seven compounds were synthesized and evaluated against lung and colon cancer cell lines. Our results showed that compound 5a, which incorporates an α-naphthyloxy substitution, exhibits the highest antiproliferative activity against both cancer lines (SK-LU-1 and SW620, IC50 < 100 nM) and induces morphological changes in the cells associated with mitotic arrest and mitotic catastrophe. Nevertheless, the tubulin polymerization assay showed that 5a has a lower inhibitory potency than nocodazole. Molecular dynamics simulations suggested that this low antitubulin activity could be associated with the loss of the key H-bond interaction with V236. This study provides insights into the design of novel carbendazim derivatives with anticancer activity.
Collapse
Affiliation(s)
- Lucia Cano-González
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Johan D Espinosa-Mendoza
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Félix Matadamas-Martínez
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Ariana Romero-Velásquez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Miguel Flores-Ramos
- Escuela Nacional de Estudios Superiores, Unidad Mérida, Universidad Nacional Autónoma de México, Yucatán 97357, Mexico
| | - Luis Fernando Colorado-Pablo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | | | - Rafael Castillo
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Ignacio González-Sánchez
- Departamento de Biología, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Lilián Yépez-Mulia
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Hospital de Pediatría, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City 06720, Mexico
| | - Alicia Hernández-Campos
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| | - Rodrigo Aguayo-Ortiz
- Departamento de Farmacia, Facultad de Química, Universidad Nacional Autónoma de México, Mexico City 04510, Mexico
| |
Collapse
|
46
|
Du BX, Xu Y, Yiu SM, Yu H, Shi JY. ADMET property prediction via multi-task graph learning under adaptive auxiliary task selection. iScience 2023; 26:108285. [PMID: 38026198 PMCID: PMC10654589 DOI: 10.1016/j.isci.2023.108285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/18/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
It is a critical step in lead optimization to evaluate the absorption, distribution, metabolism, excretion, and toxicity (ADMET) properties of drug-like compounds. Classical single-task learning (STL) has effectively predicted individual ADMET endpoints with abundant labels. Conversely, multi-task learning (MTL) can predict multiple ADMET endpoints with fewer labels, but ensuring task synergy and highlighting key molecular substructures remain challenges. To tackle these issues, this work elaborates a multi-task graph learning framework for predicting multiple ADMET properties of drug-like small molecules (MTGL-ADMET) by holding a new paradigm of MTL, "one primary, multiple auxiliaries." It first adeptly combines status theory with maximum flow for auxiliary task selection. The subsequent phase introduces a primary-task-centric MTL model with integrated modules. MTGL-ADMET not only outstrips existing STL and MTL methods but also offers a transparent lens into crucial molecular substructures. It is anticipated that this work can promote lead compound finding and optimization in drug discovery.
Collapse
Affiliation(s)
- Bing-Xue Du
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Yi Xu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Siu-Ming Yiu
- Department of Computer Science, The University of Hong Kong, Hong Kong, China
| | - Hui Yu
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| | - Jian-Yu Shi
- School of Life Sciences, Northwestern Polytechnical University, Xi’an 710072, China
| |
Collapse
|
47
|
Murphy ST, Atienza J, Brown JW, Cheruvallath ZS, Cukierski MJ, Fabrey R, Keung W, Kwok L, O’Connell S, Tang M, Vanderpool DL, Vincent PW, Zhang L, Marx MA. Optimization of mTOR Inhibitors Using Property-Based Drug Design and Free-Wilson Analysis for Improved In Vivo Efficacy. ACS Med Chem Lett 2023; 14:1544-1550. [PMID: 37970587 PMCID: PMC10641921 DOI: 10.1021/acsmedchemlett.3c00351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/17/2023] Open
Abstract
The mTOR kinase regulates a variety of critical cellular processes and has become a target for the treatment of various cancers. Using a combination of property-based drug design and Free-Wilson analysis, we further optimized a series of selective mTOR inhibitors based on the (S)-6a-methyl-6a,7,9,10-tetrahydro[1,4]oxazino[3,4-h]pteridin-6(5H)-one scaffold. Our efforts resulted in 14c, which showed similar in vivo efficacy compared to previous lead 1 at 1/15 the dose, a result of its improved drug-like properties.
Collapse
Affiliation(s)
- Sean T. Murphy
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Joy Atienza
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jason W. Brown
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | | | - Matthew J. Cukierski
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Robyn Fabrey
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Walter Keung
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Lily Kwok
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Shawn O’Connell
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Mingnam Tang
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Darin L. Vanderpool
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Patrick W. Vincent
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Lilly Zhang
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Matthew A. Marx
- Takeda California, 9625 Towne Centre Drive, San Diego, California 92121, United States
| |
Collapse
|
48
|
Higashi T, Yoshida C, Hachiro Y, Nakata C, Takechi A, Yagi T, Miyashita K, Kitada N, Obata R, Hirano T, Hara T, Maki SA. Synthesis and anti-tumor activities in human leukemia-derived cells of polyenylpyrroles with a methyl group at the conjugated polyene terminus. Bioorg Med Chem Lett 2023; 95:129471. [PMID: 37717362 DOI: 10.1016/j.bmcl.2023.129471] [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: 08/08/2023] [Revised: 09/04/2023] [Accepted: 09/06/2023] [Indexed: 09/19/2023]
Abstract
To develop novel drugs for treating T-cell acute lymphoblastic leukemia (T-ALL) and acute myeloid leukemia (AML) which are highly malignant hematological tumors, a series of analogs having a polyenylpyrrole structure of natural compounds (rumbrin and auxarconjugatin B) were synthesized and investigated their structure-activity relationships (SAR) of in vitro anti-T-ALL and anti-AML activities. We obtained three findings: (1) introduction of a methyl group at the conjugated polyene terminus enhanced anti-T-ALL activity, (2) analogs with a 3-chloropyrrole moiety had even higher selectivity for T-ALL cells, and (3) some analogs were effective against AML-derived cells. Among the studied compounds, 3-chloro-2-(8-ethoxycarbonylnona-1,3,5,7-tetraenyl) pyrrole 4e was the most promising candidate of T-ALL- and AML-treating drug. This study provides useful structural information for designing novel drugs treating T-ALL and AML.
Collapse
Affiliation(s)
- Tomoya Higashi
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Chihiro Yoshida
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Yoshifumi Hachiro
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Chihiro Nakata
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Azusa Takechi
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Takuya Yagi
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Kazuya Miyashita
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Division of Cellular Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | - Nobuo Kitada
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Rika Obata
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Takashi Hirano
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan
| | - Takahiko Hara
- Stem Cell Project, Tokyo Metropolitan Institute of Medical Science, 2-1-6 Kamikitazawa, Setagaya-ku, Tokyo 156-8506, Japan; Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan; Graduate School of Science, Department of Biological Science, Tokyo Metropolitan University, 1-1 Minami-Osawa, Hachioji-shi, Tokyo 192-0397, Japan
| | - Shojiro A Maki
- Department of Engineering Science, Graduate School of Informatics and Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan; Center for Neuroscience and Biomedical Engineering, The University of Electro-Communications, 1-5-1 Chofugaoka, Chofu-shi, Tokyo 182-8585, Japan.
| |
Collapse
|
49
|
Josa-Culleré L, Galan SRG, Cogswell TJ, Jackson TR, Jay-Smith M, Mola L, Greaves CR, Carter TS, Madden KS, Trott S, Zhang D, Bataille CJR, Davies SG, Vyas P, Milne TA, Naylor A, Wynne GM, Russell AJ. Phenotypic screening identifies a trisubstituted imidazo[1,2-a]pyridine series that induces differentiation in multiple AML cell lines. Eur J Med Chem 2023; 258:115509. [PMID: 37343464 DOI: 10.1016/j.ejmech.2023.115509] [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: 02/08/2023] [Revised: 05/19/2023] [Accepted: 05/21/2023] [Indexed: 06/23/2023]
Abstract
Acute myeloid leukaemia (AML) is an aggressive type of leukaemia with low rates of long-term survival. While the current standard of care is based on cytotoxic chemotherapy, a promising emerging approach is differentiation therapy. However, most current differentiating agents target specific mutations and are effective only in certain patient subtypes. To identify agents which may be effective in wider population cohorts, we performed a phenotypic screen with the myeloid marker CD11b and identified a compound series that was able to differentiate AML cell lines in vitro regardless of their mutation status. Structure-activity relationship studies revealed that replacing the formamide and catechol methyl ether groups with sulfonamide and indazole respectively improved the in vitro metabolic profile of the series while maintaining the differentiation profile in multiple cell lines. This optimisation exercise enabled progression of a lead compound to in vivo efficacy testing. Our work supports the promise of phenotypic screening to identify novel small molecules that induce differentiation in a wide range of AML subtypes.
Collapse
Affiliation(s)
- Laia Josa-Culleré
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Sébastien R G Galan
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Thomas J Cogswell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Thomas R Jackson
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Morgan Jay-Smith
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Laura Mola
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Christopher R Greaves
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Tom S Carter
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Katrina S Madden
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Sophie Trott
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Douzi Zhang
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Carole J R Bataille
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK
| | - Stephen G Davies
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Thomas A Milne
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DS, UK.
| | - Alan Naylor
- Alan Naylor Consultancy Ltd., Harston, Cambridge, CB22 7QJ, UK
| | - Graham M Wynne
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK
| | - Angela J Russell
- Department of Chemistry, Chemistry Research Laboratory, University of Oxford, Mansfield Road, Oxford, OX1 3TA, UK; Department of Pharmacology, University of Oxford, Mansfield Road, Oxford, OX1 3QT, UK.
| |
Collapse
|
50
|
Aspnes GE, Bagley SW, Coffey SB, Conn EL, Curto JM, Edmonds DJ, Genovino J, Griffith DA, Ingle G, Jiao W, Limberakis C, Mathiowetz AM, Piotrowski DW, Rose CR, Ruggeri RB, Wei L. 6-Azaspiro[2.5]octanes as small molecule agonists of the human glucagon-like peptide-1 receptor. Bioorg Med Chem Lett 2023; 94:129454. [PMID: 37591316 DOI: 10.1016/j.bmcl.2023.129454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/05/2023] [Accepted: 08/14/2023] [Indexed: 08/19/2023]
Abstract
Activation of the glucagon-like peptide-1 (GLP-1) receptor stimulates insulin release, lowers plasma glucose levels, delays gastric emptying, increases satiety, suppresses food intake, and affords weight loss in humans. These beneficial attributes have made peptide-based agonists valuable tools for the treatment of type 2 diabetes mellitus and obesity. However, efficient, and consistent delivery of peptide agents generally requires subcutaneous injection, which can reduce patient utilization. Traditional orally absorbed small molecules for this target may offer improved patient compliance as well as the opportunity for co-formulation with other oral therapeutics. Herein, we describe an SAR investigation leading to small-molecule GLP-1 receptor agonists that represent a series that parallels the recently reported clinical candidate danuglipron. In the event, identification of a benzyloxypyrimidine lead, using a sensitized high-throughput GLP-1 agonist assay, was followed by optimization of the SAR using substituent modifications analogous to those discovered in the danuglipron series. A new series of 6-azaspiro[2.5]octane molecules was optimized into potent GLP-1 agonists. Information gleaned from cryogenic electron microscope structures was used to rationalize the SAR of the optimized compounds.
Collapse
Affiliation(s)
- Gary E Aspnes
- Pfizer Medicine Design, Cambridge, MA 02139, United States
| | | | | | - Edward L Conn
- Pfizer Medicine Design, Groton, CT 06340, United States
| | - John M Curto
- Pfizer Medicine Design, Groton, CT 06340, United States
| | | | | | | | | | - Wenhua Jiao
- Pfizer Medicine Design, Groton, CT 06340, United States
| | | | | | | | - Colin R Rose
- Pfizer Medicine Design, Groton, CT 06340, United States
| | | | - Liuqing Wei
- Pfizer Medicine Design, Groton, CT 06340, United States
| |
Collapse
|