1
|
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
|
2
|
Loesche C, Picard D, Van Hoorick B, Schuhmann I, Jäger P, Klein K, Schuhler C, Thoma G, Markert C, Poller B, Zamurovic N, Weiss HM, Otto H, Fink M, Röhn TA. LTA4H inhibitor LYS006: Clinical PK/PD and safety in a randomized phase I clinical trial. Clin Transl Sci 2024; 17:e13724. [PMID: 38407540 PMCID: PMC10837484 DOI: 10.1111/cts.13724] [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: 07/20/2023] [Revised: 12/13/2023] [Accepted: 12/26/2023] [Indexed: 02/27/2024] Open
Abstract
LYS006 is a novel, highly potent and selective, new-generation leukotriene A4 hydrolase (LTA4H) inhibitor in clinical development for the treatment of neutrophil-driven inflammatory diseases. We describe the complex pharmacokinetic to pharmacodynamic (PD) relationship in blood, plasma, and skin of LYS006-treated nonclinical species and healthy human participants. In a randomized first in human study, participants were exposed to single ascending doses up to 100 mg and multiple ascending doses up to 80 mg b.i.d.. LYS006 showed rapid absorption, overall dose proportional plasma exposure and nonlinear blood to plasma distribution caused by saturable target binding. The compound efficiently inhibited LTB4 production in human blood and skin blister cells, leading to greater than 90% predose target inhibition from day 1 after treatment initiation at doses of 20 mg b.i.d. and above. Slow re-distribution from target expressing cells resulted in a long terminal half-life and a long-lasting PD effect in ex vivo stimulated blood and skin cells despite low plasma exposures. LYS006 was well-tolerated and demonstrated a favorable safety profile up to highest doses tested, without any dose-limiting toxicity. This supported further clinical development in phase II studies in predominantly neutrophil-driven inflammatory conditions, such as hidradenitis suppurativa, inflammatory acne, and ulcerative colitis.
Collapse
Affiliation(s)
- Christian Loesche
- Translational MedicineNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | - Damien Picard
- Translational MedicineNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
- Present address:
Vaderis Therapeutics AGBaselSwitzerland
| | | | - Imelda Schuhmann
- Biomarker DevelopmentNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | - Petra Jäger
- Immunology Disease AreaNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | - Kai Klein
- PK SciencesNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | - Carole Schuhler
- Early Development Analytics, Novartis Pharma AGBaselSwitzerland
| | - Gebhard Thoma
- Global Discovery ChemistryNovartis Biomedical Research, Novartis Pharma AGBaselSwitzerland
| | - Christian Markert
- Global Discovery ChemistryNovartis Biomedical Research, Novartis Pharma AGBaselSwitzerland
| | - Birk Poller
- PK SciencesNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | | | - H. Markus Weiss
- PK SciencesNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | - Heike Otto
- Immunology Disease AreaNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| | - Martin Fink
- Early Development Analytics, Novartis Pharma AGBaselSwitzerland
| | - Till A. Röhn
- Immunology Disease AreaNovartis BioMedical Research, Novartis Pharma AGBaselSwitzerland
| |
Collapse
|
3
|
Stanger L, Holinstat M. Bioactive lipid regulation of platelet function, hemostasis, and thrombosis. Pharmacol Ther 2023; 246:108420. [PMID: 37100208 PMCID: PMC11143998 DOI: 10.1016/j.pharmthera.2023.108420] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 04/15/2023] [Accepted: 04/17/2023] [Indexed: 04/28/2023]
Abstract
Platelets are small, anucleate cells in the blood that play a crucial role in the hemostatic response but are also implicated in the pathophysiology of cardiovascular disease. It is widely appreciated that polyunsaturated fatty acids (PUFAs) play an integral role in the function and regulation of platelets. PUFAs are substrates for oxygenase enzymes cyclooxygenase-1 (COX-1), 5-lipoxygenase (5-LOX), 12-lipoxygenase (12-LOX) and 15-lipoxygenase (15-LOX). These enzymes generate oxidized lipids (oxylipins) that exhibit either pro- or anti-thrombotic effects. Although the effects of certain oxylipins, such as thromboxanes and prostaglandins, have been studied for decades, only one oxylipin has been therapeutically targeted to treat cardiovascular disease. In addition to the well-known oxylipins, newer oxylipins that demonstrate activity in the platelet have been discovered, further highlighting the expansive list of bioactive lipids that can be used to develop novel therapeutics. This review outlines the known oxylipins, their activity in the platelet, and current therapeutics that target oxylipin signaling.
Collapse
Affiliation(s)
- Livia Stanger
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States of America
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States of America; Department of Internal Medicine, Division of Cardiovascular Medicine, University of Michigan Medical School, Ann Arbor, MI, United States of America.
| |
Collapse
|
4
|
Fang M, Tang X, Zhang J, Liao Z, Wang G, Cheng R, Zhang Z, Zhao H, Wang J, Tan Z, Kamau PM, Lu Q, Liu Q, Deng G, Lai R. An inhibitor of leukotriene-A 4 hydrolase from bat salivary glands facilitates virus infection. Proc Natl Acad Sci U S A 2022; 119:e2110647119. [PMID: 35238649 PMCID: PMC8915838 DOI: 10.1073/pnas.2110647119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 12/20/2021] [Indexed: 12/23/2022] Open
Abstract
SignificanceAn immunosuppressant protein (MTX), which facilitates virus infection by inhibiting leukotriene A4 hydrolase (LTA4H) to produce the lipid chemoattractant leukotriene B4 (LTB4), was identified and characterized from the submandibular salivary glands of the bat Myotis pilosus. To the best of our knowledge, this is a report of an endogenous LTA4H inhibitor in animals. MTX was highly concentrated in the bat salivary glands, suggesting a mechanism for the generation of immunological privilege and immune tolerance and providing evidence of viral shedding through oral secretions. Moreover, given that the immunosuppressant MTX selectively inhibited the proinflammatory activity of LTA4H, without affecting its antiinflammatory activity, MTX might be a potential candidate for the development of antiinflammatory drugs by targeting the LTA4-LTA4H-LTB4 inflammatory axis.
Collapse
Affiliation(s)
- Mingqian Fang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
| | - Xiaopeng Tang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
| | - Juan Zhang
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhiyi Liao
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Gan Wang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Ruomei Cheng
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhiye Zhang
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Hongwen Zhao
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jing Wang
- Department of Laboratory Diagnosis, Chongqing Public Health Medical Center, Public Health Hospital of Southwest University, Shapingba District, Chongqing 400038, China
| | - Zhaoxia Tan
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peter Muiruri Kamau
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Qiumin Lu
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| | - Qi Liu
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guohong Deng
- Department of Infectious Diseases, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Ren Lai
- Key Laboratory of Animal Models and Human Disease Mechanisms of the Chinese Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, Kunming Primate Research Center, and National Research Facility for Phenotypic and Genetic Analysis of Model Animals (Primate Facility), Kunming Institute of Zoology, Kunming 650107, Yunnan, China
- Kunming College of Life Science, University of Chinese Academy of Sciences, Beijing 100049, China
- Institutes for Drug Discovery and Development, Chinese Academy of Sciences, Shanghai 201203, China
- Sino-African Joint Research Center, Chinese Academy of Sciences, Wuhan, Hubei 430074, China
| |
Collapse
|
5
|
Röhn TA, Numao S, Otto H, Loesche C, Thoma G. Drug discovery strategies for novel leukotriene A4 hydrolase inhibitors. Expert Opin Drug Discov 2021; 16:1483-1495. [PMID: 34191664 DOI: 10.1080/17460441.2021.1948998] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
IntroductionLeukotriene A4 hydrolase (LTA4H) is the final and rate limiting enzyme regulating the biosynthesis of leukotriene B4 (LTB4), a pro-inflammatory lipid mediator implicated in a large number of inflammatory pathologies. Inhibition of LTA4H not only prevents LTB4 biosynthesis but also induces a lipid mediator class-switch within the 5-lipoxygenase pathway, elevating biosynthesis of the anti-inflammatory lipid mediator Lipoxin A4. Ample preclinical evidence advocates LTA4H as attractive drug target for the treatment of chronic inflammatory diseases.Areas coveredThis review covers details about the biochemistry of LTA4H and describes its role in regulating pro- and anti-inflammatory mediator generation. It summarizes recent efforts in medicinal chemistry toward novel LTA4H inhibitors, recent clinical trials testing LTA4H inhibitors in pulmonary inflammatory diseases, and potential reasons for the discontinuation of former development programs.Expert opinionGiven the prominent role of LTB4 in initiating and perpetuating inflammation, LTA4H remains an appealing drug target. The reason former attempts targeting this enzyme have not met with success in the clinic can be attributed to compound-specific liabilities of first-generation inhibitors and/or choice of target indications to test this mode of action. A new generation of highly potent and selective LTA4H inhibitors is currently undergoing clinical testing in indications with a strong link to LTB4 biology.
Collapse
Affiliation(s)
- Till A Röhn
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Shin Numao
- Chemical Biology and Therapeutics, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Heike Otto
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Christian Loesche
- Translational Medicine, Novartis Institutes for Biomedical Research, Basel, Switzerland
| | - Gebhard Thoma
- Global Discovery Chemistry, Novartis Institutes for Biomedical Research, Basel, Switzerland
| |
Collapse
|
6
|
Markert C, Thoma G, Srinivas H, Bollbuck B, Lüönd RM, Miltz W, Wälchli R, Wolf R, Hinrichs J, Bergsdorf C, Azzaoui K, Penno CA, Klein K, Wack N, Jäger P, Hasler F, Beerli C, Loetscher P, Dawson J, Wieczorek G, Numao S, Littlewood-Evans A, Röhn TA. Discovery of LYS006, a Potent and Highly Selective Inhibitor of Leukotriene A 4 Hydrolase. J Med Chem 2021; 64:1889-1903. [PMID: 33592148 DOI: 10.1021/acs.jmedchem.0c01955] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The cytosolic metalloenzyme leukotriene A4 hydrolase (LTA4H) is the final and rate-limiting enzyme in the biosynthesis of pro-inflammatory leukotriene B4 (LTB4). Preclinical studies have validated this enzyme as an attractive drug target in chronic inflammatory diseases. Despite several attempts, no LTA4H inhibitor has reached the market, yet. Herein, we disclose the discovery and preclinical profile of LYS006, a highly potent and selective LTA4H inhibitor. A focused fragment screen identified hits that could be cocrystallized with LTA4H and inspired a fragment merging. Further optimization led to chiral amino acids and ultimately to LYS006, a picomolar LTA4H inhibitor with exquisite whole blood potency and long-lasting pharmacodynamic effects. Due to its high selectivity and its ability to fully suppress LTB4 generation at low exposures in vivo, LYS006 has the potential for a best-in-class LTA4H inhibitor and is currently investigated in phase II clinical trials in inflammatory acne, hidradenitis suppurativa, ulcerative colitis, and NASH.
Collapse
|
7
|
Audat SA, Al-Shar’i NA, Al-Oudat BA, Bryant-Friedrich A, Bedi MF, Zayed AL, Al-Balas QA. Identification of Human Leukotriene A4 Hydrolase Inhibitors Using Structure-Based Pharmacophore Modeling and Molecular Docking. Molecules 2020; 25:molecules25122871. [PMID: 32580506 PMCID: PMC7356593 DOI: 10.3390/molecules25122871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 06/14/2020] [Accepted: 06/19/2020] [Indexed: 12/30/2022] Open
Abstract
Leukotriene B4 (LTB4) is a potent, proinflammatory lipid mediator implicated in the pathologies of an array of inflammatory diseases and cancer. The biosynthesis of LTB4 is regulated by the leukotriene A4 hydrolase (LTA4H). Compounds capable of limiting the formation of LTB4, through selective inhibition of LTA4H, are expected to provide potent anti-inflammatory and anti-cancer agents. The aim of the current study is to obtain potential LTA4H inhibitors using computer-aided drug design. A hybrid 3D structure-based pharmacophore model was generated based on the crystal structure of LTA4H in complex with bestatin. The generated pharmacophore was used in a virtual screen of the Maybridge database. The retrieved hits were extensively filtered, then docked into the active site of the enzyme. Finally, they were consensually scored to yield five hits as potential LTA4H inhibitors. Consequently, the selected hits were purchased and their biological activity assessed in vitro against the epoxide hydrolase activity of LTA4H. The results were very promising, with the most active compound showing 73.6% inhibition of the basal epoxide hydrolase activity of LTA4H. The results from this exploratory study provide valuable information for the design and development of more potent and selective inhibitors.
Collapse
Affiliation(s)
- Suaad A. Audat
- Department of Chemistry, College of Science and Arts, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
- Correspondence: ; Tel.: +962-772046922; Fax: +962-7201071
| | - Nizar A. Al-Shar’i
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan; (N.A.A.-S.); (B.A.A.-O.); (A.L.Z.); (Q.A.A.-B.)
| | - Buthina A. Al-Oudat
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan; (N.A.A.-S.); (B.A.A.-O.); (A.L.Z.); (Q.A.A.-B.)
| | - Amanda Bryant-Friedrich
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA; (A.B.-F.); (M.F.B.)
| | - Mel F. Bedi
- Department of Medicinal and Biological Chemistry, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH 43606, USA; (A.B.-F.); (M.F.B.)
| | - Aref L. Zayed
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan; (N.A.A.-S.); (B.A.A.-O.); (A.L.Z.); (Q.A.A.-B.)
| | - Qosay A. Al-Balas
- Department of Medicinal Chemistry and Pharmacognosy, Faculty of Pharmacy, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan; (N.A.A.-S.); (B.A.A.-O.); (A.L.Z.); (Q.A.A.-B.)
| |
Collapse
|
8
|
Numao S, Hasler F, Laguerre C, Srinivas H, Wack N, Jäger P, Schmid A, Osmont A, Röthlisberger P, Houguenade J, Bergsdorf C, Dawson J, Carte N, Hofmann A, Markert C, Hardaker L, Billich A, Wolf RM, Penno CA, Bollbuck B, Miltz W, Röhn TA. Feasibility and physiological relevance of designing highly potent aminopeptidase-sparing leukotriene A4 hydrolase inhibitors. Sci Rep 2017; 7:13591. [PMID: 29051536 PMCID: PMC5648829 DOI: 10.1038/s41598-017-13490-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/25/2017] [Indexed: 01/01/2023] Open
Abstract
Leukotriene A4 Hydrolase (LTA4H) is a bifunctional zinc metalloenzyme that comprises both epoxide hydrolase and aminopeptidase activity, exerted by two overlapping catalytic sites. The epoxide hydrolase function of the enzyme catalyzes the biosynthesis of the pro-inflammatory lipid mediator leukotriene (LT) B4. Recent literature suggests that the aminopeptidase function of LTA4H is responsible for degradation of the tripeptide Pro-Gly-Pro (PGP) for which neutrophil chemotactic activity has been postulated. It has been speculated that the design of epoxide hydrolase selective LTA4H inhibitors that spare the aminopeptidase pocket may therefore lead to more efficacious anti-inflammatory drugs. In this study, we conducted a high throughput screen (HTS) for LTA4H inhibitors and attempted to rationally design compounds that would spare the PGP degrading function. While we were able to identify compounds with preference for the epoxide hydrolase function, absolute selectivity was not achievable for highly potent compounds. In order to assess the relevance of designing such aminopeptidase-sparing LTA4H inhibitors, we studied the role of PGP in inducing inflammation in different settings in wild type and LTA4H deficient (LTA4H KO) animals but could not confirm its chemotactic potential. Attempting to design highly potent epoxide hydrolase selective LTA4H inhibitors, therefore seems to be neither feasible nor relevant.
Collapse
Affiliation(s)
- Shin Numao
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Franziska Hasler
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Claire Laguerre
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Honnappa Srinivas
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Nathalie Wack
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Petra Jäger
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Andres Schmid
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Arnaud Osmont
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Patrik Röthlisberger
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Jeremy Houguenade
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Christian Bergsdorf
- Chemical Biology & Therapeutics, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Janet Dawson
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Nathalie Carte
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Andreas Hofmann
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Christian Markert
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Liz Hardaker
- Respiratory Research, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Andreas Billich
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Romain M Wolf
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Carlos A Penno
- Analytical Sciences & Imaging, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Birgit Bollbuck
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Wolfgang Miltz
- Global Discovery Chemistry, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland
| | - Till A Röhn
- Autoimmunity, Transplantation and Inflammation, Novartis Institutes for BioMedical Research, Novartis Pharma AG, Basel, Switzerland.
| |
Collapse
|
9
|
Enomoto H, Morikawa Y, Miyake Y, Tsuji F, Mizuchi M, Suhara H, Fujimura KI, Horiuchi M, Ban M. Synthesis and biological evaluation of N-mercaptoacylproline and N-mercaptoacylthiazolidine-4-carboxylic acid derivatives as leukotriene A4 hydrolase inhibitors. Bioorg Med Chem Lett 2008; 18:4529-32. [DOI: 10.1016/j.bmcl.2008.07.043] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 07/09/2008] [Accepted: 07/11/2008] [Indexed: 11/26/2022]
|
10
|
Morgan EL, Maskrey BH, Rowley AF. At what stage in metazoan evolution did leukotriene generation first appear?--key insights from cartilaginous fish. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2005; 29:53-59. [PMID: 15325523 DOI: 10.1016/j.dci.2004.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 04/19/2004] [Accepted: 05/06/2004] [Indexed: 05/24/2023]
Abstract
Leukotriene (LT) B4 is a key player in inflammatory responses in mammals. During the generation of this derivative of arachidonic acid, the unstable product of 5-lipoxygenase, termed LTA4, is converted to LTB4 by LTA4 hydrolase. Invertebrates do not generate LTs yet all vertebrates from bony fish onwards synthesize this compound. As cartilaginous fish are the most primitive living jawed vertebrates, we investigated if the leukocytes from such a fish, the Thornback ray (Raja clavata) could generate LTB4. Supernatants from ionophore-challenged leukocytes generated the 5-lipoxygenase products, 6-trans-LTB4 and 6-trans-12-epi-LTB4 but were unable to synthesize LTB4. To determine if these cells contained an active LTA4 hydrolase, LTA4 was incubated with lysates from ray leukocytes. Such preparations did not contain any demonstrable LTA4 hydrolase activity. Our findings imply at the stage of cartilaginous fish evolution over 350 million years ago that the evolution of an active LTA4 hydrolase had yet to occur.
Collapse
Affiliation(s)
- Elizabeth L Morgan
- School of Biological Sciences, University of Wales Swansea, Singleton Park, Swansea SA2 8PP, Wales, UK
| | | | | |
Collapse
|
11
|
Chen X, Li N, Wang S, Wu N, Hong J, Jiao X, Krasna MJ, Beer DG, Yang CS. Leukotriene A4 hydrolase in rat and human esophageal adenocarcinomas and inhibitory effects of bestatin. J Natl Cancer Inst 2003; 95:1053-61. [PMID: 12865451 DOI: 10.1093/jnci/95.14.1053] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Esophageal adenocarcinoma (EAC) is increasing at the most rapid rate of any cancer in the United States. An esophagogastroduodenal anastomosis (EGDA) surgical model in rats mimics human gastroesophageal reflux and results in EAC. Leukotriene A4 hydrolase (LTA4H), a protein overexpressed in EAC in this model, is a rate-limiting enzyme in the biosynthesis of leukotriene B4 (LTB4), a potent inflammatory mediator. We used this model and human EAC and non-tumor tissues to elucidate the expression pattern of LTA4H and to evaluate it as a target for chemoprevention. METHODS LTA4H expression was examined by western blotting and immunohistochemistry. The functional role of LTA4H in carcinogenesis was investigated by use of an LTA4H inhibitor, bestatin, in the rat EGDA model. All statistical tests were two-sided. RESULTS LTA4H was overexpressed in all 10 rat EACs examined, compared with its level in normal rat tissue; it was also overexpressed in four of six human EAC tumor samples, compared with its level in adjacent non-tumor tissue. In tissue sections from 20 EGDA rats and 92 patients (86 with EAC, one with dysplasia, and five with columnar-lined esophagus), LTA4H was expressed in infiltrating inflammatory cells and overexpressed in the columnar cells of preinvasive lesions and cancers, especially in well-differentiated EACs, as compared with the basal cells of the normal esophageal squamous epithelium. Bestatin statistically significantly inhibited LTB4 biosynthesis in the esophageal tissues of EGDA rats (without bestatin = 8.28 ng/mg of protein; with bestatin = 4.68 ng/mg of protein; difference = 3.60, 95% CI = 1.59 to 5.61; P = .002) and reduced the incidence of EAC in the EGDA rats from 57.7% (15 of 26 rats) to 26.1% (6 of 23 rats) (difference = 31.6%, 95% CI = 0.3% to 56.2%; P = .042). CONCLUSION LTA4H overexpression appears to be an early event in esophageal adenocarcinogenesis and is a potential target for the chemoprevention of EAC.
Collapse
Affiliation(s)
- Xiaoxin Chen
- Susan Lehman Cullman Laboratory for Cancer Research, Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, the State University of New Jersey, Piscataway, NJ 08854, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Hevko JM, Murphy RC. Formation of murine macrophage-derived 5-oxo-7-glutathionyl-8,11,14-eicosatrienoic acid (FOG7) is catalyzed by leukotriene C4 synthase. J Biol Chem 2002; 277:7037-43. [PMID: 11748223 DOI: 10.1074/jbc.m108942200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
5-Oxo-7-glutathionyl-8,11,14-eicosatrienoic acid (FOG(7)), a biologically active glutathione (GSH) adduct of the eicosanoid 5-oxo-eicosatrienoic acid (5-oxoETE), is the major metabolite formed within the murine peritoneal macrophage. The conjugation of GSH to electrophilic 5-oxoETE in vitro was found to be catalyzed by both soluble glutathione S-transferase and membrane-bound leukotriene C(4) (LTC(4)) synthase. The cytosolic glutathione S-transferase-catalyzed products were not biologically active; however, the adduct formed from recombinant LTC(4) synthase had identical mass spectrometric properties and biological activity to the macrophage-derived FOG(7). The biosynthesis of FOG(7) in the macrophage was inhibited by MK-886, a known inhibitor of LTC(4) synthase, suggesting that this nuclear membrane-bound enzyme might be responsible for GSH conjugation to 5-oxoETE in the intact cell. Subcellular fractionation revealed that the microsomal fraction from the murine macrophage contained the enzyme responsible for FOG(7) biosynthesis. Western blot analysis confirmed the presence of LTC(4) synthase in the microsomal fraction that did not catalyze conjugation of GSH to 1-chloro-2,4-dinitrobenzene, indicating an absence of microsomal glutathione S- transferase activity. These results suggest that LTC(4) synthase, thought to be specific for the conjugation of GSH to LTA(4), can also recognize 5-oxoETE as an electrophilic substrate.
Collapse
Affiliation(s)
- John M Hevko
- Division of Cell Biology, Department of Pediatrics, National Jewish Medical and Research Center, 1400 Jackson Street, Denver, CO 80206, USA
| | | |
Collapse
|
13
|
Aberrant expression of active leukotriene C4 synthase in CD16+ neutrophils from patients with chronic myeloid leukemia. Blood 2000. [DOI: 10.1182/blood.v95.4.1456.004k06_1456_1464] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Elevated leukotriene (LT)C4 synthase activity was observed in peripheral blood granulocyte suspensions from patients with chronic myeloid leukemia (CML). Magnetic cell sorting (MACS) with CD16 monoclonal antibodies (mAbs), which were used to fractionate granulocytes from CML patients and healthy individuals, yielded highly purified suspensions of CD16+ neutrophils. The purity of these cell fractions was verified by extensive morphologic examination. Reverse transcriptase–polymerase chain reaction (RT-PCR) analyses, demonstrating the absence of interleukin-4 messenger RNA (IL-4 mRNA), further confirmed the negligible contamination of eosinophils in these fractions. Notably, purified CML CD16+ neutrophils from all tested patients transformed exogenous LTA4 to LTC4. These cells also produced LTC4 after activation with ionophore A23187 or the chemotactic peptide fMet-LeuPhe (N-formylmethionyl-leucyl-phenylalanine). Subcellular fractionation revealed that the enzyme activity was exclusively distributed to the microsomal fraction. Expression of LTC4 synthase mRNA in CML CD16+neutrophils was confirmed by RT-PCR. Furthermore, Western blot analyses consistently demonstrated expression of LTC4 synthase at the protein level in CML CD16+ neutrophils, whereas expression of microsomal glutathione S-transferase 2 occurred occasionally. Expectedly, LTC4 synthase activity or expression of the protein could not be demonstrated in CD16+ neutrophil suspensions from any of the healthy individuals. Instead, these cells, as well as CML CD16+neutrophils, transformed LTA4 to LTB4. The results indicate that aberrant expression of LTC4 synthase is a regular feature of morphologically mature CML CD16+neutrophils. This abnormality, possibly associated with malignant transformation, can lead to increased LTC4 synthesis in vivo. Such overproduction may be of pathophysiological relevance because LTC4 has been demonstrated to stimulate proliferation of human bone marrow–derived myeloid progenitor cells.
Collapse
|