1
|
Quancard J, Bach A, Borsari C, Craft R, Gnamm C, Guéret SM, Hartung IV, Koolman HF, Laufer S, Lepri S, Messinger J, Ritter K, Sbardella G, Unzue Lopez A, Willwacher MK, Cox B, Young RJ. The European Federation for Medicinal Chemistry and Chemical Biology (EFMC) Best Practice Initiative: Hit to Lead. ChemMedChem 2025; 20:e202400931. [PMID: 39957306 DOI: 10.1002/cmdc.202400931] [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: 11/18/2024] [Revised: 01/26/2025] [Indexed: 02/18/2025]
Abstract
The Hit to Lead (H2L) process is an integral part of contemporary drug discovery, encompassing the optimisation of validated Hit structures into Lead molecules. High quality leads build confidence, through activity and property profiles as well as preliminary biological data, which might include validating pharmacologic hypotheses along the way, indicating that further investment in the structure(s) and target would be worthwhile. Leads have line of sight to a development candidate and bring an understanding of what priorities Lead Optimisation should address. In this set of best practices, we detail the essential criteria that characterise a good lead, which include establishing SAR from analogues and assessing preliminary DMPK indicators, selectivity and early safety parameters. We highlight the importance of identifying liabilities of the lead series and demonstrating that each can be individually modulated whilst maintaining on target potency. We make the case for having physicochemical properties as critical optimisation parameters and how ligand efficiency metrics can enable this. Then we go over general tactics that can be used to convert hits into a lead series. These include essential steps that, when performed early, increase the chance of success such as deconstructive SAR, pharmacophore and bioactive conformation determination and scaffold optimisation. Finally, we suggest decision-making criteria to substantiate confidence in further investment or, as importantly, making a recommendation to cease further work on a series.
Collapse
Affiliation(s)
- Jean Quancard
- Global Discovery Chemistry, Novartis Biomedical Research, Novartis Pharma AG, Novartis Campus, 4056, Basel, Switzerland
| | - Anders Bach
- Department of Drug Design & Pharmacology, Faculty of Health & Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Chiara Borsari
- Dipartimento di Scienze Farmaceutiche, Università degli Studi di Milano, via L. Mangiagalli 25, 20133, Milano, Italy
| | - Russell Craft
- Medicinal chemistry, Symeres, Kadijk 3, 9747 AT, Groningen, The Netherlands
| | - Christian Gnamm
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riß, Germany
| | - Stéphanie M Guéret
- Medicinal Chemistry, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, 43183, Gothenburg, Sweden
| | - Ingo V Hartung
- Medicinal Chemistry, Global R&D, Merck Healthcare KGaA, Frankfurter Straße 250, 64293, Darmstadt, Germany
| | - Hannes F Koolman
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riß, Germany
| | - Stefan Laufer
- Pharmaceutical & Medicinal Chemistry, Institute of Pharmacy & Biochemistry, Tübingen Center for Academic Drug Discovery, Auf der Morgenstelle 8, 72070, Tübingen, Germany
| | - Susan Lepri
- Discovery Chemistry, Johnson & Johnson Innovative Medicine, Turnhoutseweg 30, 2340, Beerse, Belgium
| | - Josef Messinger
- Medicine Design, Orionpharma, Orionintie 1, 02101, Espoo, Finland
| | - Kurt Ritter
- Pharmaceutical & Medicinal Chemistry, Institute of Pharmacy & Biochemistry, Tübingen Center for Academic Drug Discovery, Auf der Morgenstelle 8, 72070, Tübingen, Germany
| | - Gianluca Sbardella
- Department of Pharmacy, Epigenetic Med Chem Lab, University of Salerno, Via Giovanni Paolo II 132, 84084, Fisciano (SA), Italy
| | - Andrea Unzue Lopez
- Medicinal Chemistry, Global R&D, Merck Healthcare KGaA, Frankfurter Straße 250, 64293, Darmstadt, Germany
| | - Marina K Willwacher
- Medicinal Chemistry, Boehringer Ingelheim Pharma GmbH & Co. KG, Birkendorfer Straße 65, 88397, Biberach an der Riß, Germany
| | - Brian Cox
- School of Life Sciences, University of Sussex, Brighton, BN1 9RH, UK
| | | |
Collapse
|
2
|
Brvar M, O'Neill TJ, Plettenburg O, Krappmann D. An updated patent review of MALT1 inhibitors (2021-present). Expert Opin Ther Pat 2025:1-18. [PMID: 40209204 DOI: 10.1080/13543776.2025.2484371] [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: 01/08/2025] [Revised: 03/01/2025] [Accepted: 03/21/2025] [Indexed: 04/12/2025]
Abstract
INTRODUCTION MALT1 paracaspase acts as a molecular scaffold and a proteolytic enzyme in immune cells. MALT1 has emerged as a promising drug target for cancer therapy, and especially for targeting MALT1 in aggressive lymphomas. Drug discovery programs have yielded potent and selective MALT1 protease inhibitors. First-in-class MALT1 inhibitors have been moved to early clinical trials to evaluate safety and efficacy. AREAS COVERED This review will provide an update regarding the mode of action, the chemical space and therapeutic use of MALT1 inhibitors based on recent patents and the scientific literature (05/2021-12/2024). EXPERT OPINION Allosteric inhibition is the preferred mode of action to inhibit the MALT1 protease. Chemical advances largely focus on improving binding and inhibition in the allosteric site of MALT1. New composition of matter has been generated, but a clinical proof for the safety and efficacy of allosteric MALT1 inhibitors is still pending. We still lack potent and selective competitive or covalent MALT1 inhibitors, indicating the challenges with targeting the active site. Further, MALT1 protein degraders and MALT1 scaffolding inhibitors have been developed, which may have distinct inhibitory profiles compared to allosteric MALT1 protease inhibitors, but more potent and selective compounds are needed to judge the feasibility and usefulness of these approaches.
Collapse
Affiliation(s)
- Matjaz Brvar
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz-University Hannover, Hannover, Germany
| | - Thomas J O'Neill
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
- Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz-University Hannover, Hannover, Germany
- Laboratory of Nano- and Quantum Engineering, Leibniz University Hannover, Hannover, Germany
- Medicinal Chemistry and Drug Design, Institute of Lung Health (ILH), Gießen, Germany
| | - Daniel Krappmann
- Research Unit Signaling and Translation, Group Signaling and Immunity, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
- Faculty of Biology, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| |
Collapse
|
3
|
Zhang RY, Wang ZX, Zhang MY, Wang YF, Zhou SL, Xu JL, Lin WX, Ji TR, Chen YD, Lu T, Li NG, Shi ZH. MALT1 Inhibitors and Degraders: Strategies for NF-κB-Driven Malignancies. J Med Chem 2025; 68:5075-5096. [PMID: 39999563 DOI: 10.1021/acs.jmedchem.4c02873] [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: 02/27/2025]
Abstract
Mucosa-associated lymphoid tissue protein 1 (MALT1), a cysteine protease and the sole paracaspase in humans, plays a pivotal role in the survival and proliferation of NF-κB-dependent malignant cancers, particularly MALT lymphoma and diffuse large B-cell lymphoma (DLBCL). Dysregulated MALT1 activity is implicated in various malignancies, highlighting its importance as a therapeutic target. This Perspective provides an overview of MALT1's structural and functional characteristics, summarizes recent advancements in small-molecule inhibitors and degraders targeting this protein, and discusses compound structures, structure-activity relationship (SAR) analyses, and biological activities. We aim to inform future research efforts to enhance the activity, selectivity, and pharmacological properties of MALT1-targeting compounds, establishing a foundational framework for drug development in this critical area of cancer therapy.
Collapse
Affiliation(s)
- Ru-Yue Zhang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Zi-Xuan Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Meng-Yuan Zhang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Yu-Fan Wang
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Si-Li Zhou
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Jia-Lu Xu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Wen-Xuan Lin
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Tian-Rui Ji
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Ya-Dong Chen
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Tao Lu
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| | - Nian-Guang Li
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, Jiangsu 210023, China
| | - Zhi-Hao Shi
- Laboratory of Molecular Design and Drug Discovery, School of Science, China Pharmaceutical University, 639 Longmian Avenue, Nanjing, Jiangsu 211198, China
| |
Collapse
|
4
|
Plotnik JP, Richardson AE, Yang H, Rojas E, Bontcheva V, Dowell C, Parsons S, Wilson A, Ravanmehr V, Will C, Jung P, Zhu H, Partha SK, Panchal SC, Mali RS, Kohlhapp FJ, McClure RA, Ramathal CY, George MD, Jhala M, Elsen NL, Qiu W, Judge RA, Pan C, Mastracchio A, Henderson J, Meulbroek JA, Green MR, Pappano WN. Inhibition of MALT1 and BCL2 Induces Synergistic Antitumor Activity in Models of B-Cell Lymphoma. Mol Cancer Ther 2024; 23:949-960. [PMID: 38507740 PMCID: PMC11217731 DOI: 10.1158/1535-7163.mct-23-0518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/21/2023] [Accepted: 03/18/2024] [Indexed: 03/22/2024]
Abstract
The activated B cell (ABC) subset of diffuse large B-cell lymphoma (DLBCL) is characterized by chronic B-cell receptor signaling and associated with poor outcomes when treated with standard therapy. In ABC-DLBCL, MALT1 is a core enzyme that is constitutively activated by stimulation of the B-cell receptor or gain-of-function mutations in upstream components of the signaling pathway, making it an attractive therapeutic target. We discovered a novel small-molecule inhibitor, ABBV-MALT1, that potently shuts down B-cell signaling selectively in ABC-DLBCL preclinical models leading to potent cell growth and xenograft inhibition. We also identified a rational combination partner for ABBV-MALT1 in the BCL2 inhibitor, venetoclax, which when combined significantly synergizes to elicit deep and durable responses in preclinical models. This work highlights the potential of ABBV-MALT1 monotherapy and combination with venetoclax as effective treatment options for patients with ABC-DLBCL.
Collapse
MESH Headings
- Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/antagonists & inhibitors
- Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism
- Humans
- Animals
- Mice
- Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Proto-Oncogene Proteins c-bcl-2/genetics
- Drug Synergism
- Xenograft Model Antitumor Assays
- Cell Line, Tumor
- Sulfonamides/pharmacology
- Sulfonamides/therapeutic use
- Cell Proliferation/drug effects
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/genetics
- Lymphoma, Large B-Cell, Diffuse/pathology
- Bridged Bicyclo Compounds, Heterocyclic/pharmacology
- Bridged Bicyclo Compounds, Heterocyclic/therapeutic use
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Disease Models, Animal
Collapse
Affiliation(s)
| | | | - Haopeng Yang
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Estela Rojas
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | | | - Sydney Parsons
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Ashley Wilson
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Vida Ravanmehr
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Paul Jung
- AbbVie Inc., North Chicago, Illinois.
| | | | | | | | | | | | | | | | | | | | | | - Wei Qiu
- AbbVie Inc., North Chicago, Illinois.
| | | | - Chin Pan
- AbbVie Bay Area, South San Francisco, California.
| | | | - Jared Henderson
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | | - Michael R. Green
- Department of Lymphoma and Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas.
| | | |
Collapse
|
5
|
Evteev S, Ivanenkov Y, Semenov I, Malkov M, Mazaleva O, Bodunov A, Bezrukov D, Sidorenko D, Terentiev V, Malyshev A, Zagribelnyy B, Korzhenevskaya A, Aliper A, Zhavoronkov A. Quantum-assisted fragment-based automated structure generator (QFASG) for small molecule design: an in vitro study. Front Chem 2024; 12:1382512. [PMID: 38633987 PMCID: PMC11021760 DOI: 10.3389/fchem.2024.1382512] [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: 02/05/2024] [Accepted: 03/12/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction: The significance of automated drug design using virtual generative models has steadily grown in recent years. While deep learning-driven solutions have received growing attention, only a few modern AI-assisted generative chemistry platforms have demonstrated the ability to produce valuable structures. At the same time, virtual fragment-based drug design, which was previously less popular due to the high computational costs, has become more attractive with the development of new chemoinformatic techniques and powerful computing technologies. Methods: We developed Quantum-assisted Fragment-based Automated Structure Generator (QFASG), a fully automated algorithm designed to construct ligands for a target protein using a library of molecular fragments. QFASG was applied to generating new structures of CAMKK2 and ATM inhibitors. Results: New low-micromolar inhibitors of CAMKK2 and ATM were designed using the algorithm. Discussion: These findings highlight the algorithm's potential in designing primary hits for further optimization and showcase the capabilities of QFASG as an effective tool in this field.
Collapse
Affiliation(s)
- Sergei Evteev
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Yan Ivanenkov
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Ivan Semenov
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Maxim Malkov
- Insilico Medicine AI Limited, Abu Dhabi, United Arab Emirates
| | - Olga Mazaleva
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Artem Bodunov
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Dmitry Bezrukov
- Insilico Medicine AI Limited, Abu Dhabi, United Arab Emirates
| | - Denis Sidorenko
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Victor Terentiev
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | - Alex Malyshev
- Insilico Medicine Hong Kong Ltd., Hong Kong, Hong Kong SAR, China
| | | | | | - Alex Aliper
- Insilico Medicine AI Limited, Abu Dhabi, United Arab Emirates
| | | |
Collapse
|
6
|
VanDyke D, Xu L, Sargunas PR, Gilbreth RN, Baca M, Gao C, Hunt J, Spangler JB. Redirecting the specificity of tripartite motif containing-21 scaffolds using a novel discovery and design approach. J Biol Chem 2023; 299:105381. [PMID: 37866632 PMCID: PMC10694607 DOI: 10.1016/j.jbc.2023.105381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/30/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023] Open
Abstract
Hijacking the ubiquitin proteasome system to elicit targeted protein degradation (TPD) has emerged as a promising therapeutic strategy to target and destroy intracellular proteins at the post-translational level. Small molecule-based TPD approaches, such as proteolysis-targeting chimeras (PROTACs) and molecular glues, have shown potential, with several agents currently in clinical trials. Biological PROTACs (bioPROTACs), which are engineered fusion proteins comprised of a target-binding domain and an E3 ubiquitin ligase, have emerged as a complementary approach for TPD. Here, we describe a new method for the evolution and design of bioPROTACs. Specifically, engineered binding scaffolds based on the third fibronectin type III domain of human tenascin-C (Tn3) were installed into the E3 ligase tripartite motif containing-21 (TRIM21) to redirect its degradation specificity. This was achieved via selection of naïve yeast-displayed Tn3 libraries against two different oncogenic proteins associated with B-cell lymphomas, mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) and embryonic ectoderm development protein (EED), and replacing the native substrate-binding domain of TRIM21 with our evolved Tn3 domains. The resulting TRIM21-Tn3 fusion proteins retained the binding properties of the Tn3 as well as the E3 ligase activity of TRIM21. Moreover, we demonstrated that TRIM21-Tn3 fusion proteins efficiently degraded their respective target proteins through the ubiquitin proteasome system in cellular models. We explored the effects of binding domain avidity and E3 ligase utilization to gain insight into the requirements for effective bioPROTAC design. Overall, this study presents a versatile engineering approach that could be used to design and engineer TRIM21-based bioPROTACs against therapeutic targets.
Collapse
Affiliation(s)
- Derek VanDyke
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Linda Xu
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Paul R Sargunas
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Ryan N Gilbreth
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Manuel Baca
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - Changshou Gao
- Biologics Engineering, R&D, AstraZeneca, Gaithersburg, Maryland, USA
| | - James Hunt
- Biologics Engineering, R&D, AstraZeneca, Cambridge, UK
| | - Jamie B Spangler
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, Maryland, USA; Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Bloomberg∼Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University, Baltimore, Maryland, USA; Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
7
|
Daoui O, Elkhattabi S, Chtita S. Rational design of novel pyridine-based drugs candidates for lymphoma therapy. J Mol Struct 2022. [DOI: 10.1016/j.molstruc.2022.133964] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
8
|
Tian Y, Li S, Yang P, Su X, Liu J, Lv X, Dong K, Yang T, Duan M, Hu G, Yue H, Sun Y, Sun Y, Zhang H, Du Z, Miao Z, Tong M, Hou Y, Gao Z, Zhao Y. Synthesis and biological evaluation of new series of benzamide derivatives containing urea moiety as sEH inhibitors. Bioorg Med Chem Lett 2022; 70:128805. [PMID: 35598794 DOI: 10.1016/j.bmcl.2022.128805] [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: 04/19/2022] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/02/2022]
Abstract
The pharmacological inhibition of soluble epoxide hydrolase (sEH) was shown to reduce inflammation and pain. Herein, we described a series of newly synthesized sEH inhibitors with the trident-shaped skeleton. Intensive structural modifications led to the identification of compound B15 as a potent sEH inhibitor with an IC50 value of 0.03 ± 0.01 nM. Furthermore, compound B15 showed satisfactory metabolic stability in human liver microsomes with a half-time of 197 min. In carrageenan-induced inflammatory pain rat model, compound B15 exhibited a better therapeutic effect compared to t-AUCB and Celecoxib, which demonstrated the proof of potential as anti-inflammatory agents for pain relief.
Collapse
Affiliation(s)
- Ye Tian
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Shuo Li
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Peiyao Yang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xiaolu Su
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Jialu Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Xuening Lv
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Kuan Dong
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Ting Yang
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Meibo Duan
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Guangda Hu
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Hao Yue
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Yanping Sun
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Yongjun Sun
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Huimin Zhang
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Zhidian Du
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China
| | - Zhenyu Miao
- 3D BioOptima Co,. Ltd., Suzhou 215104, PR China
| | | | - Yunlei Hou
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China
| | - Zibin Gao
- State Key Laboratory Breeding Base-Hebei Province Key Laboratory of Molecular Chemistry for Drug, Hebei University of Science and Technology, Shijiazhuang, PR China.
| | - Yanfang Zhao
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, PR China.
| |
Collapse
|
9
|
Chintawar CC, Bhoyare VW, Mane MV, Patil NT. Enantioselective Au(I)/Au(III) Redox Catalysis Enabled by Chiral (P,N)-Ligands. J Am Chem Soc 2022; 144:7089-7095. [PMID: 35436097 DOI: 10.1021/jacs.2c02799] [Citation(s) in RCA: 50] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Presented herein is the first report of enantioselective Au(I)/Au(III) redox catalysis, enabled by a newly designed hemilabile chiral (P,N)-ligand (ChetPhos). The potential of this concept has been demonstrated by the development of enantioselective 1,2-oxyarylation and 1,2-aminoarylation of alkenes which provided direct access to the medicinally relevant 3-oxy- and 3-aminochromans (up to 88% yield and 99% ee). DFT studies were carried out to unravel the enantiodetermining step, which revealed that the stronger trans influence of phosphorus allows selective positioning of the substrate in the C2-symmetric chiral environment present around nitrogen, imparting a high level of enantioselectivity.
Collapse
Affiliation(s)
- Chetan C Chintawar
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhauri, Bhopal 462 066, India
| | - Vivek W Bhoyare
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhauri, Bhopal 462 066, India
| | - Manoj V Mane
- Physical Chemistry Division, CSIR-National Chemical Laboratory, Dr. Homi Bhabha Road, Pune 411 008, India.,Academy of Scientific and Innovative Research (AcSIR), New Delhi 110 025, India.,KAUST Catalysis Centre, King Abdullah University of Science and Technology (KAUST), Thuwal 23955-6900, Saudi Arabia
| | - Nitin T Patil
- Department of Chemistry, Indian Institute of Science Education and Research Bhopal, Bhauri, Bhopal 462 066, India
| |
Collapse
|
10
|
Yin H, Karayel O, Chao YY, Seeholzer T, Hamp I, Plettenburg O, Gehring T, Zielinski C, Mann M, Krappmann D. A20 and ABIN-1 cooperate in balancing CBM complex-triggered NF-κB signaling in activated T cells. Cell Mol Life Sci 2022; 79:112. [PMID: 35099607 PMCID: PMC8803816 DOI: 10.1007/s00018-022-04154-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 01/06/2022] [Accepted: 01/15/2022] [Indexed: 11/03/2022]
Abstract
T cell activation initiates protective adaptive immunity, but counterbalancing mechanisms are critical to prevent overshooting responses and to maintain immune homeostasis. The CARD11-BCL10-MALT1 (CBM) complex bridges T cell receptor engagement to NF-κB signaling and MALT1 protease activation. Here, we show that ABIN-1 is modulating the suppressive function of A20 in T cells. Using quantitative mass spectrometry, we identified ABIN-1 as an interactor of the CBM signalosome in activated T cells. A20 and ABIN-1 counteract inducible activation of human primary CD4 and Jurkat T cells. While A20 overexpression is able to silence CBM complex-triggered NF-κB and MALT1 protease activation independent of ABIN-1, the negative regulatory function of ABIN-1 depends on A20. The suppressive function of A20 in T cells relies on ubiquitin binding through the C-terminal zinc finger (ZnF)4/7 motifs, but does not involve the deubiquitinating activity of the OTU domain. Our mechanistic studies reveal that the A20/ABIN-1 module is recruited to the CBM complex via A20 ZnF4/7 and that proteasomal degradation of A20 and ABIN-1 releases the CBM complex from the negative impact of both regulators. Ubiquitin binding to A20 ZnF4/7 promotes destructive K48-polyubiquitination to itself and to ABIN-1. Further, after prolonged T cell stimulation, ABIN-1 antagonizes MALT1-catalyzed cleavage of re-synthesized A20 and thereby diminishes sustained CBM complex signaling. Taken together, interdependent post-translational mechanisms are tightly controlling expression and activity of the A20/ABIN-1 silencing module and the cooperative action of both negative regulators is critical to balance CBM complex signaling and T cell activation.
Collapse
Affiliation(s)
- Hongli Yin
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Ozge Karayel
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Ying-Yin Chao
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute and Friedrich Schiller University Jena, Jena, Germany.,Central Institute for Translational Cancer Research (TranslaTUM), Technical University of Munich, Munich, Germany
| | - Thomas Seeholzer
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Isabel Hamp
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, 30167, Hannover, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, 30167, Hannover, Germany
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, 30167, Hannover, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, 30167, Hannover, Germany
| | - Torben Gehring
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany
| | - Christina Zielinski
- Department of Infection Immunology, Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll-Institute and Friedrich Schiller University Jena, Jena, Germany
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Molecular Targets and Therapeutics Center, Helmholtz Zentrum München-German Research Center for Environmental Health, Ingolstaedter Landstr. 1, 85764, Neuherberg, Germany.
| |
Collapse
|
11
|
Discovery solubility measurement and assessment of small molecules with drug development in mind. Drug Discov Today 2022; 27:1315-1325. [PMID: 35114363 DOI: 10.1016/j.drudis.2022.01.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 01/04/2022] [Accepted: 01/27/2022] [Indexed: 12/24/2022]
Abstract
Solubility is a key physicochemical property for the success of any drug candidate. Although the methods used and their rationales for determining solubility are subject to project needs and stages along the drug discovery-drug development pipeline, an artificial boundary can exist at the discovery-development interface. This boundary results in less effective solubility knowledge sharing and data integration among scientists in both drug discovery and drug development. Herein, we present a refreshed perspective on solubility. Solubility experimentation is not a one-size-fits-all measurement; instead, we stress the importance of constructing a seamless solubility understanding of a molecule as it progresses from a new chemical entity into a drug product.
Collapse
|
12
|
Wang Z, Dai H, Si X, Gao C, Liu L, Zhang L, Zhang Y, Song Y, Zhao P, Zheng J, Ke Y, Liu H, Zhang Q. Synthesis and Antitumor Activity of 2,4,6-Trisubstituted Novel Quinazoline Derivatives Containing Trifluoromethyl. CHINESE J ORG CHEM 2022. [DOI: 10.6023/cjoc202107026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
13
|
Abstract
The activated B-cell (ABC) subtype of diffuse large B-cell lymphoma (DLBCL) has an aggressive course and is associated with poor prognosis in the relapsed or refractory setting. ABC-DLBCL is characterized by chronic active signaling of NF-κB, which is dependent on the CARD11-BCL10-MALT1 (CBM) complex. MALT1 is a key effector of the CBM complex and activates canonical NF-κB and AP-1 among other transcription factors via distinct protease and scaffold functions. There is therefore growing interest in therapeutic targeting of MALT1 for B-cell malignancies. Here, we review recent advances in therapeutic targeting of MALT1 for ABC-DLBCL. Covalent and allosteric MALT1 protease inhibitors have been developed which inhibit growth of ABC-DLBCL in preclinical models, and two clinical MALT1 protease inhibitors are being developed in phase I clinical trials. Importantly, these compounds can overcome resistance to BTK inhibitors in preclinical models. Alternative compounds blocking the scaffold effect of MALT1 are also in early preclinical development. Blockade of MALT1 protease activity may have important implications for anti-lymphoma immunity by increasing immunogenicity of ABC-DLBCL cells and also by potentiating anti-lymphoma activity of other immune cells in the lymphoma microenvironment. Together, early data suggest that MALT1 is a promising target for ABC-DLBCL and possibly other B-cell malignancies, and can have lymphoma cell-intrinsic as well as immune-mediated therapeutic effects.
Collapse
Affiliation(s)
- Madhav R Seshadri
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Ari M Melnick
- Department of Medicine, Division of Hematology and Oncology, Weill Cornell Medicine, Cornell University, New York, NY, USA
| |
Collapse
|
14
|
Schiesser S, Hajek P, Pople HE, Käck H, Öster L, Cox RJ. Discovery and optimization of cyclohexane-1,4-diamines as allosteric MALT1 inhibitors. Eur J Med Chem 2021; 227:113925. [PMID: 34742013 DOI: 10.1016/j.ejmech.2021.113925] [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: 08/09/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 11/19/2022]
Abstract
Inhibition of mucosa-associated lymphoid tissue lymphoma translocation protein-1 (MALT1) is a promising strategy to modulate NF-κB signaling, with the potential to treat B-cell lymphoma and autoimmune diseases. We describe the discovery and optimization of (1s,4s)-N,N'-diaryl cyclohexane-1,4-diamines, a novel series of allosteric MALT1 inhibitors, resulting in compound 8 with single digit micromolar cell potency. X-ray analysis confirms that this compound binds to an induced allosteric site in MALT1. Compound 8 is highly selective and has an excellent in vivo rat PK profile with low clearance and high oral bioavailability, making it a promising lead for further optimization.
Collapse
Affiliation(s)
- Stefan Schiesser
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden.
| | - Peter Hajek
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Huw E Pople
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden; School of Chemistry, Cardiff University, Main Building, Park Place, Cardiff, CF10 3AT, United Kingdom
| | - Helena Käck
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Linda Öster
- Mechanistic and Structural Biology, Discovery Sciences, R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| | - Rhona J Cox
- Department of Medicinal Chemistry, Research and Early Development, Respiratory & Immunology (R&I), BioPharmaceuticals R&D, AstraZeneca, Pepparedsleden 1, 43183, Mölndal, Sweden
| |
Collapse
|
15
|
Subbaiah MAM, Meanwell NA. Bioisosteres of the Phenyl Ring: Recent Strategic Applications in Lead Optimization and Drug Design. J Med Chem 2021; 64:14046-14128. [PMID: 34591488 DOI: 10.1021/acs.jmedchem.1c01215] [Citation(s) in RCA: 273] [Impact Index Per Article: 68.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The benzene moiety is the most prevalent ring system in marketed drugs, underscoring its historic popularity in drug design either as a pharmacophore or as a scaffold that projects pharmacophoric elements. However, introspective analyses of medicinal chemistry practices at the beginning of the 21st century highlighted the indiscriminate deployment of phenyl rings as an important contributor to the poor physicochemical properties of advanced molecules, which limited their prospects of being developed into effective drugs. This Perspective deliberates on the design and applications of bioisosteric replacements for a phenyl ring that have provided practical solutions to a range of developability problems frequently encountered in lead optimization campaigns. While the effect of phenyl ring replacements on compound properties is contextual in nature, bioisosteric substitution can lead to enhanced potency, solubility, and metabolic stability while reducing lipophilicity, plasma protein binding, phospholipidosis potential, and inhibition of cytochrome P450 enzymes and the hERG channel.
Collapse
Affiliation(s)
- Murugaiah A M Subbaiah
- Department of Medicinal Chemistry, Biocon-Bristol Myers Squibb Research and Development Centre, Biocon Park, Bommasandra IV Phase, Jigani Link Road, Bangalore, Karnataka 560099, India
| | - Nicholas A Meanwell
- Department of Small Molecule Drug Discovery, Bristol Myers Squibb Research and Early Development, P.O. Box 4000, Princeton, New Jersey 08543-4000, United States
| |
Collapse
|
16
|
Hamp I, O'Neill TJ, Plettenburg O, Krappmann D. A patent review of MALT1 inhibitors (2013-present). Expert Opin Ther Pat 2021; 31:1079-1096. [PMID: 34214002 DOI: 10.1080/13543776.2021.1951703] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION MALT1 is the only human paracaspase, a protease with unique cleavage activity and substrate specificity. As a key regulator of immune responses, MALT1 has attracted attention as an immune modulatory target for the treatment of autoimmune/inflammatory diseases. Further, chronic MALT1 protease activation drives survival of lymphomas, suggesting that MALT1 is a suitable drug target for lymphoid malignancies. Recent studies have indicated that MALT1 inhibition impairs immune suppressive function of regulatory T cells in the tumor microenvironment, suggesting that MALT1 inhibitors may boost anti-tumor immunity in the treatment of solid cancers. AREAS COVERED This review summarizes the literature on MALT1 patents and applications. We discuss the potential therapeutic uses for MALT1 inhibitors based on patents and scientific literature. EXPERT OPINION There has been a steep increase in MALT1 inhibitor patents. Compounds with high selectivity and good bioavailability have been developed. An allosteric binding pocket is the preferred site for potent and selective MALT1 targeting. MALT1 inhibitors have moved to early clinical trials, but toxicological studies indicate that long-term MALT1 inhibition can disrupt immune homeostasis and lead to autoimmunity. Even though this poses risks, preventing immune suppression may favor the use of MALT1 inhibitors in cancer immunotherapies.
Collapse
Affiliation(s)
- Isabel Hamp
- Institute for Medicinal Chemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Thomas J O'Neill
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Oliver Plettenburg
- Institute for Medicinal Chemistry, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Centre of Biomolecular Drug Research (BMWZ), Institute of Organic Chemistry, Leibniz Universität Hannover, Hannover, Germany
| | - Daniel Krappmann
- Research Unit Cellular Signal Integration, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
17
|
Liang X, Sun C, Li C, Yu H, Wei X, Liu X, Bao W, Shi Y, Sun X, Khamrakulov M, Yang C, Liu H. Identification of Novel Fused Heteroaromatics-Based MALT1 Inhibitors by High-Throughput Screening to Treat B Cell Lymphoma. J Med Chem 2021; 64:9217-9237. [PMID: 34181850 DOI: 10.1021/acs.jmedchem.1c00466] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Development of mucosa-associated lymphoid tissue lymphoma translocation protein 1 (MALT1) inhibitors is of great value and significance in the treatment of neoplastic disorders and inflammatory and autoimmune diseases. However, there is a lack of effective MALT1 inhibitors in clinic. Herein, a novel class of potent 5-oxo-1-thioxo-4,5-dihydro-1H-thiazolo[3,4-a]quinazoline-based MALT1 inhibitors and their covalent derivatives were first identified and designed through high-throughput screening. We demonstrated that compounds 15c, 15e, and 20c effectively inhibited the MALT1 protease and displayed selective cytotoxicity to activated B cell-like diffuse large B cell lymphoma with low single-digit micromolar potency. Furthermore, compound 20c specifically repressed NF-κB signaling and induced cell apoptosis in MALT1-dependent TMD8 cells in a dose-dependent manner. More importantly, 20c showed good pharmacokinetic properties and antitumor efficacy with no significant toxicity in the TMD8 xenograft tumor model. Collectively, this study provides valuable lead compounds of MALT1 inhibitors for further structural optimization and antitumor mechanism study.
Collapse
Affiliation(s)
- Xuewu Liang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenxia Sun
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200043, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Chunpu Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Haolan Yu
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200043, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Xiaohui Wei
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuyi Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Bao
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200043, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Yuqiang Shi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaochen Sun
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200043, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Mirzadavlat Khamrakulov
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chenghua Yang
- Department of Urology, Changhai Hospital, Second Military Medical University, Shanghai 200043, China.,CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Hong Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zu Chong Zhi Road, Shanghai 201203, China.,University of Chinese Academy of Sciences, Beijing 100049, China.,School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou 310024, China
| |
Collapse
|
18
|
Kiesewetter B, Copie-Bergman C, Levy M, Wu F, Dupuis J, Barau C, Arcaini L, Paulli M, Lucioni M, Bonometti A, Salar A, Fernández-Rodriguez C, Piris MA, Cucco F, Dobson R, Li Y, Chen Z, Robe C, Simonitsch-Klupp I, Wotherspoon A, Raderer M, Du MQ. Genetic Characterization and Clinical Features of Helicobacter pylori Negative Gastric Mucosa-Associated Lymphoid Tissue Lymphoma. Cancers (Basel) 2021; 13:2993. [PMID: 34203889 PMCID: PMC8232676 DOI: 10.3390/cancers13122993] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In Western countries, the prevalence of gastric mucosa-associated lymphoid tissue (MALT) lymphoma has declined over the last three decades. Contemporaneously, H. pylori negative gastric MALT lymphoma is increasingly encountered, and their genetic basis and clinical features remain elusive. METHODS A total of 57 cases of H. pylori negative gastric MALT lymphoma were reviewed and investigated for chromosome translocation by fluorescence in-situ hybridization and for somatic mutations by the targeted sequencing of 93 genes. RESULTS MALT1 translocation, most likely t(11;18)(q21;q21)/BIRC3-MALT1, was detected in 39% (22/57) cases, and IGH translocation was further seen in 12 MALT1-negative cases, together accounting for 60% of the cohort. Targeted sequencing was successful in 35 cases, and showed frequent mutations in NF-κB signaling pathways (TNFAIP3 = 23%, CARD11 = 9%, MAP3K14 = 9%), together affecting 14 cases (40%). The NF-κB pathway mutations were mutually exclusive from MALT1, albeit not IGH translocation, altogether occurring in 86% of cases. There was no significant correlation between the genetic changes and clinicopathological parameters. The patients showed a median of progression-free survival (PFS) of 66.3 months, and a significant superior PFS when treated with systemic versus antibiotic therapy (p = 0.004). CONCLUSION H. pylori negative gastric MALT lymphoma is characterized by highly frequent genetic changes in the NF-κB signaling pathways.
Collapse
Affiliation(s)
- Barbara Kiesewetter
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
- Department of Medicine I, Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Christiane Copie-Bergman
- Groupe Henri Mondor-Albert Chenevier, Département de Pathologie, APHP, INSERM U955, Université Paris Est, 94010 Créteil, France; (C.C.-B.); (C.R.)
| | - Michael Levy
- Groupe Henri Mondor-Albert Chenevier and EC2M3 EA7375 Research Unit, Department of Gastroenterology, APHP, 94010 Créteil, France;
| | - Fangtian Wu
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
- Department of Hematology, Pukou CLL Center, the First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jehan Dupuis
- Lymphoid Malignancies Unit, APHP, Groupe Hospitalier Henri Mondor-Albert Chenevier, 94010 Créteil, France;
| | - Caroline Barau
- Plateforme de Ressources Biologiques BB-0033-00021, APHP, Groupe Hospitalier Henri Mondor-Albert Chenevier, 94010 Créteil, France;
| | - Luca Arcaini
- Division of Hematology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
| | - Marco Paulli
- Department of Pathology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.P.); (M.L.)
| | - Marco Lucioni
- Department of Pathology, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy; (M.P.); (M.L.)
| | - Arturo Bonometti
- Anatomic Pathology, Department of Molecular Medicine University of Pavia & Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy;
| | - Antonio Salar
- Department of Hematology, Hospital del Mar & Instituto de investigaciones médicas Hospital del Mar (IMIM), 08003 Barcelona, Spain;
| | | | - Miguel A. Piris
- Pathology Department, Hospital Fundación Jiménez Díaz, 28040 Madrid, Spain;
| | - Francesco Cucco
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
| | - Rachel Dobson
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
| | - Yan Li
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
- Department of Haematology, Hebei General Hospital, Shijiazhuang 050000, China
| | - Zi Chen
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
| | - Cyrielle Robe
- Groupe Henri Mondor-Albert Chenevier, Département de Pathologie, APHP, INSERM U955, Université Paris Est, 94010 Créteil, France; (C.C.-B.); (C.R.)
| | | | - Andrew Wotherspoon
- Department of Histopathology, The Royal Marsden Hospital, 203 Fulham Rd, London SW3 6JJ, UK;
| | - Markus Raderer
- Department of Medicine I, Division of Oncology, Medical University of Vienna, 1090 Vienna, Austria;
| | - Ming Qing Du
- Department of Pathology, Division of Cellular and Molecular Pathology, University of Cambridge, Cambridge CB2 1TN, UK; (F.W.); (F.C.); (R.D.); (Y.L.); (Z.C.)
| |
Collapse
|
19
|
Nelson KM, Walters MA. The Communication of Hit Quality Using Natural History Visualizations (NHVs). SLAS DISCOVERY 2021; 26:862-869. [PMID: 34111995 DOI: 10.1177/24725552211017518] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
High-throughput screening (HTS) often yields a list of compounds that requires prioritization before further work is performed. Prioritization criteria typically include activity, selectivity, physicochemical properties, and other absolute or calculated measurements of compound "value." One critical method of compound prioritization is often not discussed in published accounts of HTS. We have referred to this oft-overlooked metric as "compound natural history." These natural histories are observational evaluations of how a compound has been reported in the historical literature or compound databases. The purpose of this work was to develop a useful natural history visualization (NHV) that could form a standard, important part of hit reporting and evaluation. In this case report, we propose an efficient and effective NHV that will assist in the prioritization of active compounds and demonstrate its utility using a retrospective analysis of reported hits. We propose that this method of compound natural history evaluation be adopted in HTS triage and become an integral component of published reports of HTS outcomes.
Collapse
Affiliation(s)
- Kathryn M Nelson
- Department of Medicinal Chemistry, Institute for Therapeutic Discovery and Development, University of Minnesota, Minneapolis, MN, USA
| | - Michael A Walters
- Department of Medicinal Chemistry, Institute for Therapeutic Discovery and Development, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
20
|
Fedoseev SV, Ershov OE. Reaction of 4-Halo-3-hydroxyfuro[3,4-c]pyridin-1(3H)-ones with Morpholine and Thiomorpholine. RUSSIAN JOURNAL OF ORGANIC CHEMISTRY 2021. [DOI: 10.1134/s1070428021030234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
21
|
Quancard J, Simic O, Pissot Soldermann C, Aichholz R, Blatter M, Renatus M, Erbel P, Melkko S, Endres R, Sorge M, Kieffer L, Wagner T, Beltz K, Mcsheehy P, Wartmann M, Régnier CH, Calzascia T, Radimerski T, Bigaud M, Weiss A, Bornancin F, Schlapbach A. Optimization of the In Vivo Potency of Pyrazolopyrimidine MALT1 Protease Inhibitors by Reducing Metabolism and Increasing Potency in Whole Blood. J Med Chem 2020; 63:14594-14608. [DOI: 10.1021/acs.jmedchem.0c01246] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Jean Quancard
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Oliver Simic
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Carole Pissot Soldermann
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Reiner Aichholz
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Markus Blatter
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Martin Renatus
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paulus Erbel
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Samu Melkko
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Ralf Endres
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Mickael Sorge
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Laurence Kieffer
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Trixie Wagner
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Karen Beltz
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Paul Mcsheehy
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Markus Wartmann
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Catherine H. Régnier
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Thomas Calzascia
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Thomas Radimerski
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Marc Bigaud
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Andreas Weiss
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Frédéric Bornancin
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| | - Achim Schlapbach
- Novartis Pharma AG, Novartis Institutes for BioMedical Research, Novartis Campus, CH-4056 Basel, Switzerland
| |
Collapse
|