1
|
Biby S, Mondal P, Xu Y, Gomm A, Kaur B, Namme JN, Wang C, Tanzi RE, Zhang S, Zhang C. Functional Characterization of an Arylsulfonamide-Based Small-Molecule Inhibitor of the NLRP3 Inflammasome. ACS Chem Neurosci 2024; 15:3576-3586. [PMID: 39297418 PMCID: PMC11450741 DOI: 10.1021/acschemneuro.4c00512] [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: 08/08/2024] [Revised: 09/04/2024] [Accepted: 09/09/2024] [Indexed: 10/03/2024] Open
Abstract
Considerable evidence indicates that the NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome plays key roles in human pathophysiology, suggesting it as a potential drug target. Currently, studies have yet to develop compounds that are promising therapeutics in the clinic by targeting the NLRP3 inflammasome. Herein, we aim to further biologically characterize a previously identified small-molecule inhibitor of the NLRP3 inflammasome from our group, YM-I-26, to confirm its functional activities. We showed that YM-I-26 is highly selective toward the NLRP3 inflammasome and binds to NLRP3 directly. A systemic analysis revealed YM-I-26 with inflammation-related and immunomodulatory activities by the Eurofins BioMAP Diversity PLUS panel. In addition, studies using the mouse microglia BV2 cell model demonstrated that YM-I-26 is not cytotoxic, improved the phagocytotic functions of BV2 cells toward beta-amyloid, and suppressed the production of cytokines of IL-1β and IL-10 upon the activation of the NLRP3 inflammasome. Collectively, our studies support the functional activities of YM-I-26 as a NLRP3 inhibitor in physiologically relevant cell models, and warrant future studies of YM-I-26 and its analogs to advance the drug development as potential therapeutics.
Collapse
Affiliation(s)
- Savannah Biby
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Prasenjit Mondal
- Genetics
and Aging Research Unit, McCance Center for Brain Health, MassGeneral
Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Yiming Xu
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Ashley Gomm
- Genetics
and Aging Research Unit, McCance Center for Brain Health, MassGeneral
Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Baljit Kaur
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Jannatun N. Namme
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Changning Wang
- Athinoula
A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Rudolph E. Tanzi
- Genetics
and Aging Research Unit, McCance Center for Brain Health, MassGeneral
Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| | - Shijun Zhang
- Department
of Medicinal Chemistry, Virginia Commonwealth
University, Richmond, Virginia 23298, United States
| | - Can Zhang
- Genetics
and Aging Research Unit, McCance Center for Brain Health, MassGeneral
Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
2
|
Truffin D, Marchand F, Chatelais M, Chêne G, Saias L, Herbst F, Lipner J, King AJ. Impact of Methylated Cyclodextrin KLEPTOSE ® CRYSMEB on Inflammatory Responses in Human In Vitro Models. Int J Mol Sci 2024; 25:9748. [PMID: 39273695 PMCID: PMC11396153 DOI: 10.3390/ijms25179748] [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: 07/19/2024] [Revised: 08/30/2024] [Accepted: 09/03/2024] [Indexed: 09/15/2024] Open
Abstract
KLEPTOSE® CRYSMEB methylated cyclodextrin derivative displays less methylated group substitution than randomly methylated cyclodextrin. It has demonstrated an impact on atherosclerosis and neurological diseases, linked in part to cholesterol complexation and immune response, however, its impact on inflammatory cascade pathways is not clear. Thus, the impact of KLEPTOSE® CRYSMEB on various pharmacological targets was assessed using human umbilical vein endothelial cells under physiological and inflammatory conditions, followed by screening against twelve human primary cell-based systems designed to model complex human tissue and disease biology of the vasculature, skin, lung, and inflammatory tissues using the BioMAP® Diversity PLUS® panel. Finally, its anti-inflammatory mechanism was investigated on peripheral blood mononuclear cells to evaluate anti-inflammatory or pro-resolving properties. The results showed that KLEPTOSE® CRYSMEB can modulate the immune system in vitro and potentially manage vascular issues by stimulating the expression of molecules involved in the crosstalk between immune cells and other cell types. It showed anti-inflammatory effects that were driven by the inhibition of pro-inflammatory cytokine secretion and could have different impacts on different tissue types. Moreover, this cyclodextrin showed no clear impact on pro-resolving lipid mediators. Additionally, it appeared that the mechanism of action of KLEPTOSE® CRYSMEB seems to not be shared by other well-known anti-inflammatory molecules. Finally, KLEPTOSE® CRYSMEB may have an anti-inflammatory impact, which could be due to its effect on receptors such as TLR or direct complexation with LPS or PGE2, and conversely, this methylated cyclodextrin could stimulate a pro-inflammatory response involving lipid mediators and on proteins involved in communication with immune cells, probably via interaction with membrane cholesterol.
Collapse
Affiliation(s)
- Damien Truffin
- Roquette Frères, Rue de la Haute Loge, 62136 Lestrem, France
| | - Flora Marchand
- ProfileHIT, 7 Rue du Buisson, 44680 Sainte-Pazanne, France
| | | | - Gérald Chêne
- Ambiotis, 3 Can Biotech 3 r Satellites, 31400 Toulouse, France
| | - Laure Saias
- Ambiotis, 3 Can Biotech 3 r Satellites, 31400 Toulouse, France
| | - Frauke Herbst
- Eurofins Discovery, 6 Research Park Drive, St. Charles, MO 63304, USA
| | - Justin Lipner
- Eurofins Discovery, 6 Research Park Drive, St. Charles, MO 63304, USA
| | - Alastair J King
- Eurofins Discovery, 6 Research Park Drive, St. Charles, MO 63304, USA
| |
Collapse
|
3
|
Xu Y, Biby S, Guo C, Liu Z, Cai J, Wang XY, Zhang S. Characterization of a small molecule inhibitor of the NLRP3 inflammasome and its potential use for acute lung injury. Bioorg Chem 2024; 150:107562. [PMID: 38901282 PMCID: PMC11270536 DOI: 10.1016/j.bioorg.2024.107562] [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: 04/24/2024] [Revised: 05/23/2024] [Accepted: 06/13/2024] [Indexed: 06/22/2024]
Abstract
Accumulating data support the key roles of the NLRP3 inflammasome, an essential component of the innate immune system, in human pathophysiology. As an emerging drug target and a potential biomarker for human diseases, small molecule inhibitors of the NLRP3 inflammasome have been actively pursued. Our recent studies identified a small molecule, MS-II-124, as a potent NLRP3 inhibitor and potential imaging probe. In this report, MS-II-124 was further characterized by an unbiased and comprehensive analysis through Eurofins BioMAP Diversity PLUS panel that contains 12 human primary cell-based systems. The analysis revealed promising activities of MS-II-124 on inflammation and immune functions, further supporting the roles of the NLRP3 inflammasome in these model systems. Further studies of MS-II-124 in mouse model of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) and NLRP3 knockout mice demonstrated its target engagement, efficacy to suppress inflammatory cytokines and infiltration of immune cells in the lung tissues. In summary, the results support the therapeutic potential of MS-II-124 as a NLRP3 inhibitor and warrant future studies of this compound and its analogs to develop therapeutics for ALI/ARDS.
Collapse
Affiliation(s)
- Yiming Xu
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Savannah Biby
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Chunqing Guo
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Zheng Liu
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Jinyang Cai
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Xiang-Yang Wang
- Department of Human and Molecular Genetics, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Shijun Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, Richmond, VA 23298, USA.
| |
Collapse
|
4
|
Mokady D, Charish J, Barretto-Burns P, Grisé KN, Coles BLK, Raab S, Ortin-Martinez A, Müller A, Fasching B, Jain P, Drukker M, van der Kooy D, Steger M. Small-Molecule-Directed Endogenous Regeneration of Visual Function in a Mammalian Retinal Degeneration Model. Int J Mol Sci 2024; 25:1521. [PMID: 38338800 PMCID: PMC10855388 DOI: 10.3390/ijms25031521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/31/2023] [Accepted: 01/20/2024] [Indexed: 02/12/2024] Open
Abstract
Degenerative retinal diseases associated with photoreceptor loss are a leading cause of visual impairment worldwide, with limited treatment options. Phenotypic profiling coupled with medicinal chemistry were used to develop a small molecule with proliferative effects on retinal stem/progenitor cells, as assessed in vitro in a neurosphere assay and in vivo by measuring Msx1-positive ciliary body cell proliferation. The compound was identified as having kinase inhibitory activity and was subjected to cellular pathway analysis in non-retinal human primary cell systems. When tested in a disease-relevant murine model of adult retinal degeneration (MNU-induced retinal degeneration), we observed that four repeat intravitreal injections of the compound improved the thickness of the outer nuclear layer along with the regeneration of the visual function, as measured with ERG, visual acuity, and contrast sensitivity tests. This serves as a proof of concept for the use of a small molecule to promote endogenous regeneration in the eye.
Collapse
Affiliation(s)
- Daphna Mokady
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Jason Charish
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | | | - Kenneth N. Grisé
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Brenda L. K. Coles
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Susanne Raab
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Arturo Ortin-Martinez
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Alex Müller
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Bernhard Fasching
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Payal Jain
- Endogena Therapeutics, Inc., 661 University Ave, Toronto, ON M5G 0B7, Canada (P.B.-B.)
| | - Micha Drukker
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| | - Derek van der Kooy
- Department of Molecular Genetics, University of Toronto, Donnelly Centre Rm 1110, 160 College Street, Toronto, ON M5S 3E1, Canada
| | - Matthias Steger
- Endogena Therapeutics, AG, Binzmuehlestrasse 170 d, CH-8050 Zuerich, Switzerland
| |
Collapse
|
5
|
Tóth KF, Ádám D, Arany J, Ramirez YA, Bíró T, Drake JI, O'Mahony A, Szöllősi AG, Póliska S, Kilić A, Soeberdt M, Abels C, Oláh A. Fluoxetine exerts anti-inflammatory effects on human epidermal keratinocytes and suppresses their endothelin release. Exp Dermatol 2024; 33:e14988. [PMID: 38284184 DOI: 10.1111/exd.14988] [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: 05/02/2023] [Revised: 11/08/2023] [Accepted: 11/22/2023] [Indexed: 01/30/2024]
Abstract
Fluoxetine is a safe antidepressant with remarkable anti-inflammatory actions; therefore, we aimed to investigate its effects on immortalized (HaCaT) as well as primary human epidermal keratinocytes in a polyinosinic-polycytidylic acid (p(I:C))-induced inflammatory model. We found that a non-cytotoxic concentration (MTT-assay, CyQUANT-assay) of fluoxetine significantly suppressed p(I:C)-induced expression and release of several pro-inflammatory cytokines (Q-PCR, cytokine array, ELISA), and it decreased the release of the itch mediator endothelins (ELISA). These effects were not mediated by the inhibition of the NF-κB or p38 MAPK pathways (western blot), or by the suppression of the p(I:C)-induced elevation of mitochondrial ROS production (MitoSOX Red labeling). Instead, unbiased activity profiling revealed that they were most likely mediated via the inhibition of the phosphoinositide 3-kinase (PI3K) pathway. Importantly, the PI3K-inhibitor GDC0941 fully mimicked the effects of fluoxetine (Q-PCR, ELISA). Although fluoxetine was able to occupy the binding site of GDC0941 (in silico molecular docking), and exerted direct inhibitory effect on PI3K (cell-free PI3K activity assay), it exhibited much lower potency and efficacy as compared to GDC0941. Finally, RNA-Seq analysis revealed that fluoxetine deeply influenced the transcriptional alterations induced by p(I:C)-treatment, and exerted an overall anti-inflammatory activity. Collectively, our findings demonstrate that fluoxetine exerts potent anti-inflammatory effects, and suppresses the release of the endogenous itch mediator endothelins in human keratinocytes, most likely via interfering with the PI3K pathway. Thus, clinical studies are encouraged to explore whether the currently reported beneficial effects translate in vivo following its topical administration in inflammatory and pruritic dermatoses.
Collapse
Affiliation(s)
- Kinga Fanni Tóth
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - Dorottya Ádám
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - József Arany
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- University of Debrecen, Doctoral School of Molecular Medicine, Debrecen, Hungary
| | - Yesid A Ramirez
- Design and Applied Sciences, School of Applied Sciences and Sustainable Industry, Department of Pharmaceutical and Chemical Sciences, Faculty of Engineering, Universidad Icesi, Cali, Valle del Cauca, Colombia
- Cannaflos-Gesellschaft für medizinisches Cannabis mbH, Köln, Germany
| | - Tamás Bíró
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | | | - Alison O'Mahony
- Eurofins Discovery, St. Charles, Missouri, USA
- Recursion, Salt Lake City, Utah, USA
| | - Attila Gábor Szöllősi
- Department of Immunology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Szilárd Póliska
- Genomic Medicine and Bioinformatics Core Facility, Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Ana Kilić
- Dr. August Wolff GmbH & Co. KG Arzneimittel, Bielefeld, Germany
| | - Michael Soeberdt
- Dr. August Wolff GmbH & Co. KG Arzneimittel, Bielefeld, Germany
- Bionorica SE, Neumarkt, Germany
| | - Christoph Abels
- Dr. August Wolff GmbH & Co. KG Arzneimittel, Bielefeld, Germany
- Bionorica SE, Neumarkt, Germany
| | - Attila Oláh
- Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| |
Collapse
|
6
|
Du Y, Gao F, Sun H, Wu C, Zhu G, Zhu M. Novel substituted 4-(Arylethynyl)-Pyrrolo[2,3-d]pyrimidines negative allosteric modulators (NAMs) of the metabotropic glutamate receptor subtype 5 (mGlu5) Treat depressive disorder in mice. Eur J Med Chem 2023; 261:115855. [PMID: 37847955 DOI: 10.1016/j.ejmech.2023.115855] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 09/13/2023] [Accepted: 10/04/2023] [Indexed: 10/19/2023]
Abstract
In view of the fact that the G-protein-coupled receptors (GPCRs) sit at the top of the signaling pathways triggering a diverse range of signaling cascades towards a cellular event, GPCRs are regarded as central drug targets. mGlu5, a type of classical GPCRs, is highly expressed in the central nervous system (CNS) and responds to the neurotransmitter glutamate. Researches show that mGlu5 is a potential drug target for the treatment of depression. Up to now, multiple mGlu5 negative allosteric modulators (NAMs) have entered clinical trials, but no small molecule mGlu5 NAM has yet to reach market. Herein, we report the structural optimization and structure-activity relationship studies of a series of novel mGlu5 NAMs. Among them, the novel compound 10b is a high-affinity mGluR5 antagonist, with an IC50 value of 11.5 nM. Besides, we evaluated the anti-depressant effect of compound 10b using the chronic unpredictable mild stress (CUMS)-induced depression model. The data showed that the mice in CUMS group were featured by decreased level of serum 5-HT and increased level of serum CORT, and the expression of synaptic proteins were reduced, including GluA1, GluA2, p-PKA, BDNF and TrkB. However, those factors for identifying sensitivity to depression-like behaviors could be improved by compound 10b treatment. The preliminary toxicology evaluations indicated that compound 10b had a good safety profile in vivo. Collectively, the compound 10b represents a promising lead compound for the treatment of depressive disorder.
Collapse
Affiliation(s)
- Yonglei Du
- Department of Chemistry and Centre for Atomic Engineering of Advanced Materials, Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Institutes of Physical Science and Information Technology and Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei, Anhui, 230601, China
| | - Feng Gao
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Hongwei Sun
- Department of Chemistry and Centre for Atomic Engineering of Advanced Materials, Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Institutes of Physical Science and Information Technology and Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei, Anhui, 230601, China
| | - Chenglin Wu
- School of Pharmacy, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Guoqi Zhu
- Key Laboratory of Xin'an Medicine, the Ministry of Education and Key Laboratory of Molecular Biology (Brain diseases), Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Manzhou Zhu
- Department of Chemistry and Centre for Atomic Engineering of Advanced Materials, Key Laboratory of Structure and Functional Regulation of Hybrid Materials of Ministry of Education, Institutes of Physical Science and Information Technology and Anhui Province Key Laboratory of Chemistry for Inorganic/Organic Hybrid Functionalized Materials, Anhui University, Hefei, Anhui, 230601, China.
| |
Collapse
|
7
|
Deken M, Niewola-Staszkowska K, Peyruchaud O, Mikulčić N, Antolić M, Shah P, Cheasty A, Tagliavini A, Nizzardo A, Pergher M, Ziviani L, Milleri S, Pickering C, Lahn M, van der Veen L, Di Conza G, Johnson Z. Characterization and translational development of IOA-289, a novel autotaxin inhibitor for the treatment of solid tumors. IMMUNO-ONCOLOGY TECHNOLOGY 2023; 18:100384. [PMID: 37234285 PMCID: PMC10205783 DOI: 10.1016/j.iotech.2023.100384] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Background Autotaxin-lysophosphatidic acid (ATX-LPA) signaling has a predominant role in immunological and fibrotic processes, including cancer. Several ATX inhibitors and LPA receptor antagonists have been clinically evaluated, but none in patients with solid tumors. Many cancers are burdened with a high degree of fibrosis and an immune desert phenotype (so-called 'cold' tumors). In these cold tumors, the fibrotic stroma provides an intrinsic cancer-supporting mechanism. Furthermore, the stroma prevents penetration and limits the effectiveness of existing therapies. IOA-289 is a novel ATX inhibitor with a unique chemical structure, excellent potency and an attractive safety profile. Materials and methods In vitro and in vivo pharmacology studies have been carried out to elucidate the pharmaceutical properties and mechanism of action of IOA-289. A phase I clinical study in healthy volunteers was carried out to determine the pharmacokinetics and pharmacodynamics of IOA-289 following a single oral dose. Results In vitro and in vivo studies showed that IOA-289 is a potent inhibitor of ATX and, as a monotherapy, is able to slow progression of lung fibrosis and tumor growth in mouse models. In a clinical study, IOA-289 showed a dose-dependent increase in plasma exposure levels and a corresponding decrease in circulating LPA. Conclusions Our data show that IOA-289 is a novel ATX inhibitor with a unique chemical structure, excellent potency and an attractive safety profile. Our data support the further development of IOA-289 as a novel therapeutic approach for the treatment of cancer, particularly those with a high fibrotic and immunologically cold phenotype.
Collapse
Affiliation(s)
| | | | - O. Peyruchaud
- INSERM, UMR 1033, Faculté de Médecine Lyon Est, Université Claude Bernard Lyon 1, Lyon, France
| | | | | | - P. Shah
- Cancer Research Horizons, Therapeutic Discovery Laboratories, Cambridge, UK
| | - A. Cheasty
- Cancer Research Horizons, Therapeutic Discovery Laboratories, Cambridge, UK
| | | | | | | | - L. Ziviani
- Centro Ricerche Cliniche di Verona srl, Verona, Italy
| | - S. Milleri
- Centro Ricerche Cliniche di Verona srl, Verona, Italy
| | | | - M. Lahn
- iOnctura, Geneva, Switzerland
| | | | | | | |
Collapse
|
8
|
Sabini E, O'Mahony A, Caturegli P. MyMD-1 Improves Health Span and Prolongs Life Span in Old Mice: A Noninferiority Study to Rapamycin. J Gerontol A Biol Sci Med Sci 2023; 78:227-235. [PMID: 35914953 DOI: 10.1093/gerona/glac142] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Aging and age-related diseases represent a compelling therapeutic goal for senolytics and drugs targeting inflammatory or metabolic pathways. We compared MyMD-1, a synthetic derivative of the alkaloid myosmine capable of suppressing TNF-α production, to rapamycin, the best characterized drug endowed with antiaging properties. In vivo, a longitudinal cohort of 54 C57BL/6 mice, 19-month-old at the start, was randomized to receive MyMD-1, high-dose (126 ppm) rapamycin, or low-dose (14 ppm) rapamycin plus metformin. Each treatment arm included 18 mice (10 females and 8 males) and was followed for 16 months or until death. Life span was significantly longer in MyMD-1 than rapamycin (p = .019 vs high-dose and .01 vs low-dose) in a Cox survival model that accounted for sex and serum levels of IL-6, TNF-α, and IL-17A. MyMD-1 also improved several health span characteristics, resulting in milder body weight loss, greater muscle strength, and slower progression to frailty. In vitro, MyMD-1 and rapamycin were compared using a panel of 12 human primary cell systems (BioMAP Diversity PLUS), where a total of 148 biomarkers were measured. MyMD-1 possessed antiproliferative, anti-inflammatory, and antifibrotic properties. Many were shared with rapamycin, but MyMD-1 was more active in the inhibition of proinflammatory and profibrotic biomarkers. Overall, MyMD-1 emerges as a new compound that, even when begun at an advanced age, induces beneficial effects on health and life span by modulating inflammation and tissue remodeling.
Collapse
Affiliation(s)
- Elena Sabini
- Johns Hopkins Hospital, School of Medicine, Department of Pathology, Baltimore, Maryland, USA
| | - Alison O'Mahony
- Eurofins Discovery, Phenotypic Services, St. Charles, Missouri, USA
| | - Patrizio Caturegli
- Johns Hopkins Hospital, School of Medicine, Department of Pathology, Baltimore, Maryland, USA
| |
Collapse
|
9
|
Vandersluis S, Reid JC, Orlando L, Bhatia M. Evidence-based support for phenotypic drug discovery in acute myeloid leukemia. Drug Discov Today 2022; 27:103407. [DOI: 10.1016/j.drudis.2022.103407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 08/01/2022] [Accepted: 10/10/2022] [Indexed: 11/03/2022]
|
10
|
Phenotypic drug discovery: recent successes, lessons learned and new directions. Nat Rev Drug Discov 2022; 21:899-914. [DOI: 10.1038/s41573-022-00472-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2022] [Indexed: 12/29/2022]
|
11
|
Chong LH, Ching T, Farm HJ, Grenci G, Chiam KH, Toh YC. Integration of a microfluidic multicellular coculture array with machine learning analysis to predict adverse cutaneous drug reactions. LAB ON A CHIP 2022; 22:1890-1904. [PMID: 35348137 DOI: 10.1039/d1lc01140e] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Adverse cutaneous reactions are potentially life-threatening skin side effects caused by drugs administered into the human body. The availability of a human-specific in vitro platform that can prospectively screen drugs and predict this risk is therefore of great importance to drug safety. However, since adverse cutaneous drug reactions are mediated by at least 2 distinct mechanisms, both involving systemic interactions between liver, immune and dermal tissues, existing in vitro skin models have not been able to comprehensively recapitulate these complex, multi-cellular interactions to predict the skin-sensitization potential of drugs. Here, we report a novel in vitro drug screening platform, which comprises a microfluidic multicellular coculture array (MCA) to model different mechanisms-of-action using a collection of simplistic cellular assays. The resultant readouts are then integrated with a machine-learning algorithm to predict the skin sensitizing potential of systemic drugs. The MCA consists of 4 cell culture compartments connected by diffusion microchannels to enable crosstalk between hepatocytes that generate drug metabolites, antigen-presenting cells (APCs) that detect the immunogenicity of the drug metabolites, and keratinocytes and dermal fibroblasts, which collectively determine drug metabolite-induced FasL-mediated apoptosis. A single drug screen using the MCA can simultaneously generate 5 readouts, which are integrated using support vector machine (SVM) and principal component analysis (PCA) to classify and visualize the drugs as skin sensitizers or non-skin sensitizers. The predictive performance of the MCA and SVM classification algorithm is then validated through a pilot screen of 11 drugs labelled by the US Food and Drug Administration (FDA), including 7 skin-sensitizing and 4 non-skin sensitizing drugs, using stratified 4-fold cross-validation (CV) on SVM. The predictive performance of our in vitro model achieves an average of 87.5% accuracy (correct prediction rate), 75% specificity (prediction rate of true negative drugs), and 100% sensitivity (prediction rate of true positive drugs). We then employ the MCA and the SVM training algorithm to prospectively identify the skin-sensitizing likelihood and mechanism-of-action for obeticholic acid (OCA), a farnesoid X receptor (FXR) agonist which has undergone clinical trials for non-alcoholic steatohepatitis (NASH) with well-documented cutaneous side effects.
Collapse
Affiliation(s)
- Lor Huai Chong
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- Bioinformatics Institute, ASTAR, 30 Biopolis St, Singapore 138671, Singapore
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, 47500, Malaysia
| | - Terry Ching
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- Pillar of Engineering Product Development, Singapore University of Technology and Design, 8 Somapah Road, Singapore 487372, Singapore
- Digital Manufacturing and Design Centre, Singapore University of Technology and Design, 8 Somapah Rd, Singapore 487372, Singapore
| | - Hui Jia Farm
- Department of Computer Science, University of Oxford, Oxford, OX1 3QD, UK
| | - Gianluca Grenci
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- Mechanobiology Institute, 5A Engineering Drive 1, Singapore 117411, Singapore
| | - Keng-Hwee Chiam
- Bioinformatics Institute, ASTAR, 30 Biopolis St, Singapore 138671, Singapore
| | - Yi-Chin Toh
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, #04-08, Singapore 117583, Singapore
- School of Mechanical Medical & Process Engineering, Queensland University of Technology, 2 George St, Brisbane, QLD 4000, Australia.
- Centre for Biomedical Technologies, Queensland University of Technology, 60 Musk Ave, Kelvin Grove, QLD 4059, Australia
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology, 60 Musk Avenue, Kelvin Grove, QLD 4059, Australia
| |
Collapse
|
12
|
Winkler A, Sun W, De S, Jiao A, Sharif MN, Symanowicz PT, Athale S, Shin JH, Wang J, Jacobson BA, Ramsey SJ, Dower K, Andreyeva T, Liu H, Hegen M, Homer BL, Brodfuehrer J, Tilley M, Gilbert SA, Danto SI, Beebe JJ, Barnes BJ, Pascual V, Lin LL, Kilty I, Fleming M, Rao VR. The Interleukin-1 Receptor-Associated Kinase 4 Inhibitor PF-06650833 Blocks Inflammation in Preclinical Models of Rheumatic Disease and in Humans Enrolled in a Randomized Clinical Trial. Arthritis Rheumatol 2021; 73:2206-2218. [PMID: 34423919 PMCID: PMC8671219 DOI: 10.1002/art.41953] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/17/2021] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To investigate the role of PF-06650833, a highly potent and selective small-molecule inhibitor of interleukin-1-associated kinase 4 (IRAK4), in autoimmune pathophysiology in vitro, in vivo, and in the clinical setting. METHODS Rheumatoid arthritis (RA) inflammatory pathophysiology was modeled in vitro through 1) stimulation of primary human macrophages with anti-citrullinated protein antibody immune complexes (ICs), 2) RA fibroblast-like synoviocyte (FLS) cultures stimulated with Toll-like receptor (TLR) ligands, as well as 3) additional human primary cell cocultures exposed to inflammatory stimuli. Systemic lupus erythematosus (SLE) pathophysiology was simulated in human neutrophils, dendritic cells, B cells, and peripheral blood mononuclear cells stimulated with TLR ligands and SLE patient ICs. PF-06650833 was evaluated in vivo in the rat collagen-induced arthritis (CIA) model and the mouse pristane-induced and MRL/lpr models of lupus. Finally, RNA sequencing data generated with whole blood samples from a phase I multiple-ascending-dose clinical trial of PF-06650833 were used to test in vivo human pharmacology. RESULTS In vitro, PF-06650833 inhibited human primary cell inflammatory responses to physiologically relevant stimuli generated with RA and SLE patient plasma. In vivo, PF-06650833 reduced circulating autoantibody levels in the pristane-induced and MRL/lpr murine models of lupus and protected against CIA in rats. In a phase I clinical trial (NCT02485769), PF-06650833 demonstrated in vivo pharmacologic action pertinent to SLE by reducing whole blood interferon gene signature expression in healthy volunteers. CONCLUSION These data demonstrate that inhibition of IRAK4 kinase activity can reduce levels of inflammation markers in humans and provide confidence in the rationale for clinical development of IRAK4 inhibitors for rheumatologic indications.
Collapse
Affiliation(s)
| | | | - Saurav De
- The Feinstein Institute, Manhasset, New York
| | | | | | | | - Shruti Athale
- Baylor Institute for Immunology Research, Dallas, Texas
| | | | - Ju Wang
- Pfizer, Cambridge, Massachusetts
| | | | | | | | | | - Heng Liu
- Pfizer, Cambridge, Massachusetts
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Simms L, Mason E, Berg EL, Yu F, Rudd K, Czekala L, Trelles Sticken E, Brinster O, Wieczorek R, Stevenson M, Walele T. Use of a rapid human primary cell-based disease screening model, to compare next generation products to combustible cigarettes. Curr Res Toxicol 2021; 2:309-321. [PMID: 34485931 PMCID: PMC8408431 DOI: 10.1016/j.crtox.2021.08.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 07/19/2021] [Accepted: 08/04/2021] [Indexed: 12/01/2022] Open
Abstract
A growing number of public health bodies, regulators and governments around the world consider electronic vapor products a lower risk alternative to conventional cigarettes. Of critical importance are rapid new approach methodologies to enable the screening of next generation products (NGPs) also known as next generation tobacco and nicotine products. In this study, the activity of conventional cigarette (3R4F) smoke and a range of NGP aerosols (heated tobacco product, hybrid product and electronic vapor product) captured in phosphate buffered saline, were screened by exposing a panel of human cell-based model systems using Biologically Multiplexed Activity Profiling (BioMAP® Diversity PLUS® Panel, Eurofins Discovery). Following exposure, the biological activity for a wide range of biomarkers in the BioMAP panel were compared to determine the presence of toxicity signatures that are associated with specific clinical findings. NGP aerosols were found to be weakly active in the BioMAP Diversity PLUS Panel (≤3/148 biomarkers) whereas significant activity was observed for 3R4F (22/148 biomarkers). Toxicity associated biomarker signatures for 3R4F included immunosuppression, skin irritation and thrombosis, with no toxicity signatures seen for the NGPs. BioMAP profiling could effectively be used to differentiate between complex mixtures of cigarette smoke or NGP aerosol extracts in a panel of human primary cell-based assays. Clinical validation of these results will be critical for confirming the utility of BioMAP for screening NGPs for potential adverse human effects.
Collapse
Key Words
- ACM, aerosol collected mass
- AhR, Aryl hydrocarbon receptor
- Alternative methods
- COPD, Chronic obstructive pulmonary disease
- EGFR, epidermal growth factor receptor
- ELISA, enzyme-linked immunosorbent assay
- EVP, Electronic vapor product
- HDFn, Human neonatal dermal fibroblasts
- HTP, Heated Tobacco Product
- HUVEC, Human umbilical vein endothelial cells
- HYB, Hybrid product containing e-liquid drawn through a tobacco plug
- IL, interleukin
- ISO, International Organization for Standardization
- In vitro assays
- MOA, Mechanism of action
- M−CSF, Macrophage colony-stimulating factor
- NGP, Next generation product
- NRC, National Research Council
- NRF2, Nuclear factor erythroid 2-related factor 2
- Next generation products
- PBMC, Peripheral blood mononuclear cells
- PBS, Phosphate buffered saline
- Panel
- Phenotypic screening
- SRB, Sulforhodamine B
- TCR, T cell receptor
- TF, Tissue factor
- TLR, toll-like receptor
- TNFα, tumor necrosis factor alpha
- TPM, Total particulate matter
- Toxicity signature
- bPBS, Bubbled phosphate buffered saline
- mTOR, mechanistic target of rapamycin
Collapse
Affiliation(s)
- Liam Simms
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL UK
| | - Elizabeth Mason
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL UK
| | - Ellen L. Berg
- Eurofins Discovery, Inc., 111 Anza Blvd, Suite 414, Burlingame, CA 94010, USA
| | - Fan Yu
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL UK
| | - Kathryn Rudd
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL UK
| | - Lukasz Czekala
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL UK
| | - Edgar Trelles Sticken
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | - Oleg Brinster
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | - Roman Wieczorek
- Reemtsma Cigarettenfabriken GmbH, An Imperial Brands PLC Company, Albert-EinsteinRing-7, D-22761 Hamburg, Germany
| | | | - Tanvir Walele
- Imperial Brands PLC, 121 Winterstoke Road, Bristol BS3 2LL UK
| |
Collapse
|
14
|
Luban J, Sattler RA, Mühlberger E, Graci JD, Cao L, Weetall M, Trotta C, Colacino JM, Bavari S, Strambio-De-Castillia C, Suder EL, Wang Y, Soloveva V, Cintron-Lue K, Naryshkin NA, Pykett M, Welch EM, O'Keefe K, Kong R, Goodwin E, Jacobson A, Paessler S, Peltz SW. The DHODH inhibitor PTC299 arrests SARS-CoV-2 replication and suppresses induction of inflammatory cytokines. Virus Res 2021; 292:198246. [PMID: 33249060 PMCID: PMC7690341 DOI: 10.1016/j.virusres.2020.198246] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 02/01/2023]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for therapeutics that inhibit the SARS-COV-2 virus and suppress the fulminant inflammation characteristic of advanced illness. Here, we describe the anti-COVID-19 potential of PTC299, an orally bioavailable compound that is a potent inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme of the de novo pyrimidine nucleotide biosynthesis pathway. In tissue culture, PTC299 manifests robust, dose-dependent, and DHODH-dependent inhibition of SARS-COV-2 replication (EC50 range, 2.0-31.6 nM) with a selectivity index >3,800. PTC299 also blocked replication of other RNA viruses, including Ebola virus. Consistent with known DHODH requirements for immunomodulatory cytokine production, PTC299 inhibited the production of interleukin (IL)-6, IL-17A (also called IL-17), IL-17 F, and vascular endothelial growth factor (VEGF) in tissue culture models. The combination of anti-SARS-CoV-2 activity, cytokine inhibitory activity, and previously established favorable pharmacokinetic and human safety profiles render PTC299 a promising therapeutic for COVID-19.
Collapse
Affiliation(s)
- Jeremy Luban
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA; Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, MA, 01605, USA; Broad Institute of Harvard and MIT, 75 Ames Street, Cambridge, MA, 02142, USA; Massachusetts Consortium on Pathogen Readiness, Boston, MA, 02115, USA
| | - Rachel A Sattler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Elke Mühlberger
- Massachusetts Consortium on Pathogen Readiness, Boston, MA, 02115, USA; Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
| | - Jason D Graci
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | - Liangxian Cao
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | - Marla Weetall
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | | | | | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, 21702, USA
| | | | - Ellen L Suder
- Department of Microbiology, Boston University School of Medicine, Boston, MA, 02118, USA; National Emerging Infectious Diseases Laboratories, Boston University, Boston, MA, 02118, USA
| | - Yetao Wang
- Program in Molecular Medicine, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Veronica Soloveva
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter Street, Fort Detrick, MD, 21702, USA
| | | | | | - Mark Pykett
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | - Ellen M Welch
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | - Kylie O'Keefe
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | - Ronald Kong
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA
| | | | - Allan Jacobson
- Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, 01605, USA
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA
| | - Stuart W Peltz
- PTC Therapeutics, Inc. South Plainfield, NJ, 07080, USA.
| |
Collapse
|
15
|
Hill LJ, Botfield HF, Begum G, Qureshi O, Vigneswara V, Masood I, Barnes NM, Bruce L, Logan A. ILB ® resolves inflammatory scarring and promotes functional tissue repair. NPJ Regen Med 2021; 6:3. [PMID: 33414477 PMCID: PMC7791102 DOI: 10.1038/s41536-020-00110-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 11/13/2020] [Indexed: 11/09/2022] Open
Abstract
Fibrotic disease is a major cause of mortality worldwide, with fibrosis arising from prolonged inflammation and aberrant extracellular matrix dynamics. Compromised cellular and tissue repair processes following injury, infection, metabolic dysfunction, autoimmune conditions and vascular diseases leave tissues susceptible to unresolved inflammation, fibrogenesis, loss of function and scarring. There has been limited clinical success with therapies for inflammatory and fibrotic diseases such that there remains a large unmet therapeutic need to restore normal tissue homoeostasis without detrimental side effects. We investigated the effects of a newly formulated low molecular weight dextran sulfate (LMW-DS), termed ILB®, to resolve inflammation and activate matrix remodelling in rodent and human disease models. We demonstrated modulation of the expression of multiple pro-inflammatory cytokines and chemokines in vitro together with scar resolution and improved matrix remodelling in vivo. Of particular relevance, we demonstrated that ILB® acts, in part, by downregulating transforming growth factor (TGF)β signalling genes and by altering gene expression relating to extracellular matrix dynamics, leading to tissue remodelling, reduced fibrosis and functional tissue regeneration. These observations indicate the potential of ILB® to alleviate fibrotic diseases.
Collapse
Affiliation(s)
- Lisa J Hill
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Hannah F Botfield
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ghazala Begum
- Neuroscience and Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Omar Qureshi
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Vasanthy Vigneswara
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Imran Masood
- School of Biomedical Sciences, Institute of Clinical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nicholas M Barnes
- School of Pharmacy, Institute of Clinical Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Lars Bruce
- TikoMed AB, P.O. Box 81, 263 03, Viken, Sweden
| | - Ann Logan
- Axolotl Consulting Ltd., Droitwich, Worcestershire, WR9 0JS, UK. .,Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
16
|
cPLA 2α Enzyme Inhibition Attenuates Inflammation and Keratinocyte Proliferation. Biomolecules 2020; 10:biom10101402. [PMID: 33023184 PMCID: PMC7600040 DOI: 10.3390/biom10101402] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2020] [Revised: 09/25/2020] [Accepted: 09/29/2020] [Indexed: 02/06/2023] Open
Abstract
As a regulator of cellular inflammation and proliferation, cytosolic phospholipase A2 α (cPLA2α) is a promising therapeutic target for psoriasis; indeed, the cPLA2α inhibitor AVX001 has shown efficacy against plaque psoriasis in a phase I/IIa clinical trial. To improve our understanding of the anti-psoriatic properties of AVX001, we sought to determine how the compound modulates inflammation and keratinocyte hyperproliferation, key characteristics of the psoriatic epidermis. We measured eicosanoid release from human peripheral blood mononuclear cells (PBMC) and immortalized keratinocytes (HaCaT) and studied proliferation in HaCaT grown as monolayers and stratified cultures. We demonstrated that inhibition of cPLA2α using AVX001 produced a balanced reduction of prostaglandins and leukotrienes; significantly limited prostaglandin E2 (PGE2) release from both PBMC and HaCaT in response to pro-inflammatory stimuli; attenuated growth factor-induced arachidonic acid and PGE2 release from HaCaT; and inhibited keratinocyte proliferation in the absence and presence of exogenous growth factors, as well as in stratified cultures. These data suggest that the anti-psoriatic properties of AVX001 could result from a combination of anti-inflammatory and anti-proliferative effects, probably due to reduced local eicosanoid availability.
Collapse
|
17
|
Pottel J, Armstrong D, Zou L, Fekete A, Huang XP, Torosyan H, Bednarczyk D, Whitebread S, Bhhatarai B, Liang G, Jin H, Ghaemi SN, Slocum S, Lukacs KV, Irwin JJ, Berg EL, Giacomini KM, Roth BL, Shoichet BK, Urban L. The activities of drug inactive ingredients on biological targets. Science 2020; 369:403-413. [PMID: 32703874 DOI: 10.1126/science.aaz9906] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Accepted: 05/18/2020] [Indexed: 12/22/2022]
Abstract
Excipients, considered "inactive ingredients," are a major component of formulated drugs and play key roles in their pharmacokinetics. Despite their pervasiveness, whether they are active on any targets has not been systematically explored. We computed the likelihood that approved excipients would bind to molecular targets. Testing in vitro revealed 25 excipient activities, ranging from low-nanomolar to high-micromolar concentration. Another 109 activities were identified by testing against clinical safety targets. In cellular models, five excipients had fingerprints predictive of system-level toxicity. Exposures of seven excipients were investigated, and in certain populations, two of these may reach levels of in vitro target potency, including brain and gut exposure of thimerosal and its major metabolite, which had dopamine D3 receptor dissociation constant K d values of 320 and 210 nM, respectively. Although most excipients deserve their status as inert, many approved excipients may directly modulate physiologically relevant targets.
Collapse
Affiliation(s)
- Joshua Pottel
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Duncan Armstrong
- Preclinical Safety, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Ling Zou
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Alexander Fekete
- Preclinical Safety, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Xi-Ping Huang
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27759, USA
| | - Hayarpi Torosyan
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Dallas Bednarczyk
- PK Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Steven Whitebread
- Preclinical Safety, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Barun Bhhatarai
- PK Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Guiqing Liang
- PK Sciences, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - Hong Jin
- Preclinical Safety, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA
| | - S Nassir Ghaemi
- Translational Medicine, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA.,Tufts University School of Medicine, Boston, MA 02111, USA.,Harvard Medical School, Boston, MA 02115, USA
| | - Samuel Slocum
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27759, USA
| | - Katalin V Lukacs
- National Heart and Lung Institute, Imperial College, London SW7 2AZ, UK
| | - John J Irwin
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA
| | - Ellen L Berg
- Eurofins, DiscoverX, South San Francisco, CA 94080, USA
| | - Kathleen M Giacomini
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94158, USA
| | - Bryan L Roth
- Department of Pharmacology, University of North Carolina, Chapel Hill, NC 27759, USA
| | - Brian K Shoichet
- Department of Pharmaceutical Chemistry, University of California, San Francisco, CA 94158, USA.
| | - Laszlo Urban
- Preclinical Safety, Novartis Institutes for Biomedical Research, Cambridge, MA 02139, USA.
| |
Collapse
|
18
|
Luban J, Sattler R, Mühlberger E, Graci JD, Cao L, Weetall M, Trotta C, Colacino JM, Bavari S, Strambio-De-Castillia C, Suder EL, Wang Y, Soloveva V, Cintron-Lue K, Naryshkin NA, Pykett M, Welch EM, O'Keefe K, Kong R, Goodwin E, Jacobson A, Paessler S, Peltz S. The DHODH Inhibitor PTC299 Arrests SARS-CoV-2 Replication and Suppresses Induction of Inflammatory Cytokines. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020. [PMID: 32793904 DOI: 10.1101/2020.08.05.238394] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has created an urgent need for therapeutics that inhibit the SARS-CoV-2 virus and suppress the fulminant inflammation characteristic of advanced illness. Here, we describe the anti-COVID-19 potential of PTC299, an orally available compound that is a potent inhibitor of dihydroorotate dehydrogenase (DHODH), the rate-limiting enzyme of the de novo pyrimidine biosynthesis pathway. In tissue culture, PTC299 manifests robust, dose-dependent, and DHODH-dependent inhibition of SARS CoV-2 replication (EC 50 range, 2.0 to 31.6 nM) with a selectivity index >3,800. PTC299 also blocked replication of other RNA viruses, including Ebola virus. Consistent with known DHODH requirements for immunomodulatory cytokine production, PTC299 inhibited the production of interleukin (IL)-6, IL-17A (also called IL-17), IL-17F, and vascular endothelial growth factor (VEGF) in tissue culture models. The combination of anti-SARS-CoV-2 activity, cytokine inhibitory activity, and previously established favorable pharmacokinetic and human safety profiles render PTC299 a promising therapeutic for COVID-19.
Collapse
|
19
|
Berg EL. Human Cell-Based in vitro Phenotypic Profiling for Drug Safety-Related Attrition. Front Big Data 2019; 2:47. [PMID: 33693370 PMCID: PMC7931891 DOI: 10.3389/fdata.2019.00047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 11/25/2019] [Indexed: 12/17/2022] Open
Abstract
Ensuring the safety of new drugs is critically important to regulators, pharmaceutical researchers and patients alike. Even so, unexpected toxicities still account for 20–30% of clinical trial failures, in part due to the persistence of animal testing as the primary approach for de-risking new drugs. Clearly, improved methods for safety attrition that incorporate human-relevant biology are needed. This recognition has spurred interest in non-animal alternatives or new approach methodologies (NAMs) including in vitro models that utilize advances in the culture of human cell types to provide greater clinical relevance for assessing risk. These phenotypic assay systems use human primary and induced pluripotent stem cell-derived cells in various formats, including co-cultures and advanced cellular systems such as organoids, bioprinted tissues, and organs-on-a-chip. Despite the promise of these human-based phenotypic approaches, adoption of these platforms into drug discovery programs for reducing safety-related attrition has been slow. Here we discuss the value of large-scale human cell-based phenotypic profiling for incorporating human-specific biology into the de-risking process. We describe learnings from our experiences with human primary cell-based assays and analysis of clinically relevant reference datasets in developing in vitro-based toxicity signatures. We also describe how Adverse Outcome Pathway (AOP) frameworks can be used to integrate results from diverse platforms congruent with weight-of-evidence approaches from risk assessment to improve safety-related decisions in early discovery.
Collapse
Affiliation(s)
- Ellen L Berg
- Eurofins Discovery, Translational Biology, Burlingame, CA, United States
| |
Collapse
|
20
|
Singer JW, Al-Fayoumi S, Taylor J, Velichko S, O'Mahony A. Comparative phenotypic profiling of the JAK2 inhibitors ruxolitinib, fedratinib, momelotinib, and pacritinib reveals distinct mechanistic signatures. PLoS One 2019; 14:e0222944. [PMID: 31560729 PMCID: PMC6764664 DOI: 10.1371/journal.pone.0222944] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/10/2019] [Indexed: 02/06/2023] Open
Abstract
Janus kinase-signal transducers and activators of transcription (JAK-STAT) signaling is critical to multiple cellular processes, including survival, differentiation, and proliferation. JAK-STAT signaling dysregulation has been noted in inflammatory disorders, and aberrant JAK2 pathway activation has been implicated in myelofibrosis and polycythemia vera. Moreover, 4 therapeutic JAK2 inhibitors (ruxolitinib, fedratinib, momelotinib, and pacritinib) have either been approved or are in advanced clinical development for myelofibrosis. Although all inhibit JAK2, reports indicate that they also inhibit other kinases. Profiling based solely on in vitro potencies is insufficient to predict the observed clinical effects. To provide further translational insights into clinical outcomes, we compared phenotypic biomarker profiles of ruxolitinib, fedratinib, momelotinib, and pacritinib in the BioMAP® Diversity PLUS panel of 12 human primary cell systems designed to recapitulate key aspects of tissue and disease states. Biomarker activity profiles that represent mechanistic signatures for each agent were compared with each other and a database of reference benchmark profiles. At clinically relevant concentrations, these agents had distinct biomarker impacts indicating diverse mechanistic signatures, suggesting divergent clinical effects for each agent. They disparately modulated inflammatory cytokine production and immune function. At clinically relevant concentrations, ruxolitinib had the broadest scope of activities across all 12 cellular systems, whereas pacritinib was more specific for the BT system (modelling T cell-dependent B cell activation) and exhibited the strongest inhibition of sIL-17A, sIL-2, and sIL-6. All 4 agents were antiproliferative to B cells, but ruxolitinib and momelotinib were also antiproliferative to T cells. These differential activities likely reflect distinct secondary pharmacology for these agents known primarily as JAK2 inhibitors. The phenotypic analysis reported herein represents key data on distinct modes-of-action that may provide insights on clinical outcomes reported for these agents. Such translational findings may also inform the development of next-generation molecules with improved efficacy and safety.
Collapse
Affiliation(s)
- Jack W Singer
- CTI BioPharma Corp., Seattle, Washington, United States of America.,Elson Floyd College of Medicine, Washington State University, Seattle, Washington, United States of America
| | | | - Jason Taylor
- CTI BioPharma Corp., Seattle, Washington, United States of America
| | - Sharlene Velichko
- Eurofins Discovery, Phenotypic Services, Burlingame, California, United States of America
| | - Alison O'Mahony
- Eurofins Discovery, Phenotypic Services, Burlingame, California, United States of America
| |
Collapse
|
21
|
Graziani D, Caligari S, Callegari E, De Toma C, Longhi M, Frigerio F, Dilernia R, Menegon S, Pinzi L, Pirona L, Tazzari V, Valsecchi AE, Vistoli G, Rastelli G, Riva C. Evaluation of Amides, Carbamates, Sulfonamides, and Ureas of 4-Prop-2-ynylidenecycloalkylamine as Potent, Selective, and Bioavailable Negative Allosteric Modulators of Metabotropic Glutamate Receptor 5. J Med Chem 2019; 62:1246-1273. [PMID: 30624919 DOI: 10.1021/acs.jmedchem.8b01226] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Negative allosteric modulators (NAMs) of the metabotropic glutamate receptor 5 (mGlu5) hold great promise for the treatment of a variety of central nervous system disorders. We have recently reported that prop-2-ynylidenecycloalkylamine derivatives are potent and selective NAMs of the mGlu5 receptor. In this work, we explored the amide, carbamate, sulfonamide, and urea derivatives of prop-2-ynylidenecycloalkylamine compounds with the aim of improving solubility and metabolic stability. In silico and experimental analyses were performed on the synthesized series of compounds to investigate structure-activity relationships. Compounds 12, 32, and 49 of the carbamate, urea, and amide classes, respectively, showed the most suitable cytochrome inhibition and metabolic stability profiles. Among them, compound 12 showed excellent selectivity, solubility, and stability profiles as well as suitable in vitro and in vivo pharmacokinetic properties. It was highly absorbed in rats and dogs and was active in anxiety, neuropathic pain, and lower urinary tract models.
Collapse
Affiliation(s)
- Davide Graziani
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Silvia Caligari
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Elisa Callegari
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Carlo De Toma
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Matteo Longhi
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Fabio Frigerio
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Roberto Dilernia
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Sergio Menegon
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Luca Pinzi
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
| | - Lorenza Pirona
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Valerio Tazzari
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Anna Elisa Valsecchi
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| | - Giulio Vistoli
- Department of Pharmaceutical Sciences , Università degli Studi di Milano , Via Mangiagalli 25 , 20133 Milan , Italy
| | - Giulio Rastelli
- Department of Life Sciences , University of Modena and Reggio Emilia , Via Campi 103 , 41125 Modena , Italy
| | - Carlo Riva
- Drug Discovery Department , Recordati S.p.A. , Via M. Civitali 1 , 20148 Milan , Italy
| |
Collapse
|
22
|
Orellana A, García-González V, López R, Pascual-Guiral S, Lozoya E, Díaz J, Casals D, Barrena A, Paris S, Andrés M, Segarra V, Vilella D, Malhotra R, Eastwood P, Planagumà A, Miralpeix M, Nueda A. Application of a phenotypic drug discovery strategy to identify biological and chemical starting points for inhibition of TSLP production in lung epithelial cells. PLoS One 2018; 13:e0189247. [PMID: 29320511 PMCID: PMC5761851 DOI: 10.1371/journal.pone.0189247] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2017] [Accepted: 11/24/2017] [Indexed: 11/18/2022] Open
Abstract
Thymic stromal lymphopoietin (TSLP) is a cytokine released by human lung epithelium in response to external insult. Considered as a master switch in T helper 2 lymphocyte (Th2) mediated responses, TSLP is believed to play a key role in allergic diseases including asthma. The aim of this study was to use a phenotypic approach to identify new biological and chemical starting points for inhibition of TSLP production in human bronchial epithelial cells (NHBE), with the objective of reducing Th2-mediated airway inflammation. To this end, a phenotypic screen was performed using poly I:C / IL-4 stimulated NHBE cells interrogated with a 44,974 compound library. As a result, 85 hits which downregulated TSLP protein and mRNA levels were identified and a representative subset of 7 hits was selected for further characterization. These molecules inhibited the activity of several members of the MAPK, PI3K and tyrosine kinase families and some of them have been reported as modulators of cellular phenotypic endpoints like cell-cell contacts, microtubule polymerization and caspase activation. Characterization of the biological profile of the hits suggested that mTOR could be a key activity involved in the regulation of TSLP production in NHBE cells. Among other targeted kinases, inhibition of p38 MAPK and JAK kinases showed different degrees of correlation with TSLP downregulation, while Syk kinase did not seem to be related. Overall, inhibition of TSLP production by the selected hits, rather than resulting from inhibition of single isolated targets, appeared to be due to a combination of activities with different levels of relevance. Finally, a hit expansion exercise yielded additional active compounds that could be amenable to further optimization, providing an opportunity to dissociate TSLP inhibition from other non-desired activities. This study illustrates the potential of phenotypic drug discovery to complement target based approaches by providing new chemistry and biology leads.
Collapse
Affiliation(s)
- Adelina Orellana
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | | | - Rosa López
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | | | - Estrella Lozoya
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Julia Díaz
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Daniel Casals
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Antolín Barrena
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Stephane Paris
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Miriam Andrés
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Victor Segarra
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Dolors Vilella
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Rajneesh Malhotra
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Paul Eastwood
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | - Anna Planagumà
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
| | | | - Arsenio Nueda
- Almirall R&D Center, Almirall S.A., Sant Feliu de Llobregat, Barcelona, Spain
- * E-mail:
| |
Collapse
|
23
|
Bone AJ, Houck KA. Primary Cell Phenotypic Screening Illuminates ADRs and AOPs. Cell Chem Biol 2017; 24:781-782. [PMID: 28732198 DOI: 10.1016/j.chembiol.2017.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Preclinical, in vitro screening for adverse drug reactions continues to present challenges in the field of drug development. In this issue of Cell Chemical Biology, Shah et al. (2017) employ a phenotypic screening strategy using a panel of human primary cells to define a signature response and an adverse outcome pathway for delayed type IV skin hypersensitivity.
Collapse
Affiliation(s)
- Audrey J Bone
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA
| | - Keith A Houck
- National Center for Computational Toxicology, U.S. Environmental Protection Agency, Research Triangle Park, NC 27711, USA.
| |
Collapse
|
24
|
Stepan AF, Claffey MM, Reese MR, Balan G, Barreiro G, Barricklow J, Bohanon MJ, Boscoe BP, Cappon GD, Chenard LK, Cianfrogna J, Chen L, Coffman KJ, Drozda SE, Dunetz JR, Ghosh S, Hou X, Houle C, Karki K, Lazzaro JT, Mancuso JY, Marcek JM, Miller EL, Moen MA, O'Neil S, Sakurada I, Skaddan M, Parikh V, Smith DL, Trapa P, Tuttle JB, Verhoest PR, Walker DP, Won A, Wright AS, Whritenour J, Zasadny K, Zaleska MM, Zhang L, Shaffer CL. Discovery and Characterization of (R)-6-Neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894), an Alkyne-Lacking Metabotropic Glutamate Receptor 5 Negative Allosteric Modulator Profiled in both Rat and Nonhuman Primates. J Med Chem 2017; 60:7764-7780. [PMID: 28817277 DOI: 10.1021/acs.jmedchem.7b00604] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
We previously observed a cutaneous type IV immune response in nonhuman primates (NHP) with the mGlu5 negative allosteric modulator (NAM) 7. To determine if this adverse event was chemotype- or mechanism-based, we evaluated a distinct series of mGlu5 NAMs. Increasing the sp3 character of high-throughput screening hit 40 afforded a novel morpholinopyrimidone mGlu5 NAM series. Its prototype, (R)-6-neopentyl-2-(pyridin-2-ylmethoxy)-6,7-dihydropyrimido[2,1-c][1,4]oxazin-4(9H)-one (PF-06462894, 8), possessed favorable properties and a predicted low clinical dose (2 mg twice daily). Compound 8 did not show any evidence of immune activation in a mouse drug allergy model. Additionally, plasma samples from toxicology studies confirmed that 8 did not form any reactive metabolites. However, 8 caused the identical microscopic skin lesions in NHPs found with 7, albeit with lower severity. Holistically, this work supports the hypothesis that this unique toxicity may be mechanism-based although additional work is required to confirm this and determine clinical relevance.
Collapse
Affiliation(s)
- Antonia F Stepan
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Michelle M Claffey
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Matthew R Reese
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Gayatri Balan
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Gabriela Barreiro
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Jason Barricklow
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Michael J Bohanon
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Brian P Boscoe
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Gregg D Cappon
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Lois K Chenard
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Julie Cianfrogna
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Laigao Chen
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Karen J Coffman
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Susan E Drozda
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Joshua R Dunetz
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Somraj Ghosh
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Xinjun Hou
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Christopher Houle
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Kapil Karki
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - John T Lazzaro
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Jessica Y Mancuso
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - John M Marcek
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Emily L Miller
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Mark A Moen
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Steven O'Neil
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Isao Sakurada
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Marc Skaddan
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Vinod Parikh
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Deborah L Smith
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Patrick Trapa
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Jamison B Tuttle
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Patrick R Verhoest
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Daniel P Walker
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Annie Won
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Ann S Wright
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Jessica Whritenour
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Kenneth Zasadny
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Margaret M Zaleska
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Lei Zhang
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| | - Christopher L Shaffer
- Neuroscience and Pain Medicinal Chemistry, ‡Pharmacokinetics, Dynamics, and Metabolism, and §Neuroscience and Pain Research Unit, Pfizer Inc. , Cambridge, Massachusetts 02139, United States.,Pharmaceutical Sciences, ⊥Pharmacokinetics, Dynamics, and Metabolism, #Biostatistics, Early Clinical Development, ∇Drug Safety Research and Development, and ○BioImaging Center, Pfizer Inc. , Groton, Connecticut 06340, United States
| |
Collapse
|