1
|
Lellouche F, Blais-Lecours P, Maltais F, Sarrazin JF, Rola P, Nguyen T, Châteauvert N, Marsolais D. Ozanimod Therapy in Patients With COVID-19 Requiring Oxygen Support: A Randomized Open-Label Pilot Trial. Chest 2024; 165:810-819. [PMID: 37898184 DOI: 10.1016/j.chest.2023.10.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 09/26/2023] [Accepted: 10/20/2023] [Indexed: 10/30/2023] Open
Abstract
BACKGROUND Sphingosine-1-phosphate receptor ligands (SRLs) dampen immunopathologic damages in models of viral pneumonia. RESEARCH QUESTION Is it feasible to administer an SRL therapy, here ozanimod (OZA), to acutely ill patients infected with SARS-CoV-2? STUDY DESIGN AND METHODS The prospective randomized open-label COVID-19 Ozanimod Intervention (COZI) pilot trial was conducted in three Canadian hospitals. Patients admitted for COVID-19 requiring oxygen were eligible. Randomization was stratified for risk factors of poor outcome and oxygen needs at inclusion. Participants were allocated to standard of care or to standard of care plus OZA. OZA (oral, once daily, incremental dosage) was administered for a maximum of 14 days. Primary end point investigated for size effect and variance over time was the assessment of safety and efficacy, evaluated by the daily score on the World Health Organization-adapted six-point ordinal scale for clinical improvement analyzed under the intention-to-treat principle. RESULTS Twenty-three patients were randomized to the standard of care arm, and 20 were randomized to the OZA arm from September 2020 to February 2022. Evaluation of efficacy showed nonsignificant reductions of median (interquartile range) duration of respiratory support (6 [3-10] vs 9 [4-12] days; P = .34), median duration of hospitalization (9 [6-12] vs 10 [6-18] days; P = .20), and median time to clinical improvement (4 [3-7] vs 7 [3-11] days; P = .12) for OZA compared with standard of care, respectively. Heart rate was significantly lower with OZA (65 [ 63-67] vs 71 [69-72] beats/min; P < .0001). However, QT and PR intervals were not affected. No severe adverse drug reaction was reported. INTERPRETATION To our knowledge, SRL utility in severe pneumonia has never been tested in patients. This study shows for the first time that this new pharmacologic agent may safely be administered to patients hospitalized for viral pneumonia, with potential clinical benefits. Bradycardia was frequent but well tolerated. CLINICAL TRIAL REGISTRATION ClinicalTrials.gov; No.: NCT04405102; URL: www. CLINICALTRIALS gov.
Collapse
Affiliation(s)
- François Lellouche
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada.
| | - Pascale Blais-Lecours
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - François Maltais
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Jean-François Sarrazin
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| | - Philippe Rola
- CIUSSS EMTL, Santa Cabrini Hospital, Montreal, QC, Canada
| | - Tuyen Nguyen
- CISSS Laval, Cité-de-la-Santé Hospital, Laval, QC, Canada
| | - Nathalie Châteauvert
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada
| | - David Marsolais
- Institut universitaire de cardiologie et de pneumologie de Québec-Université Laval, Quebec City, QC, Canada; Department of Medicine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
2
|
Camp SM, Marciniak A, Chiang ET, Garcia AN, Bittman R, Polt R, Perez RG, Dudek SM, Garcia JGN. Sphingosine-1-phosphate receptor-independent lung endothelial cell barrier disruption induced by FTY720 regioisomers. Pulm Circ 2020; 10:10.1177_2045894020905521. [PMID: 32095229 PMCID: PMC7011338 DOI: 10.1177/2045894020905521] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 01/16/2020] [Indexed: 12/27/2022] Open
Abstract
RATIONALE Vascular permeability is a hallmark of acute respiratory distress syndrome (ARDS) and ventilator-induced lung injury pathobiology; however, the mechanisms underlying this vascular dysregulation remain unclear, thereby impairing the development of desperately needed effective therapeutics. We have shown that sphingosine-1-phosphate (S1P) and 2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol (FTY720) analogues are useful tools for exploring vascular barrier regulation mechanisms. OBJECTIVE To experimentally define the effects of FTY720 regioisomers on lung endothelial cell barrier regulation. METHODS Specific barrier-regulatory receptor and kinase inhibitors were utilized to probe signaling mechanisms involved in FTY720 regioisomer-mediated human lung endothelial cell barrier responses (trans-endothelial electrical resistance, TER). Docking simulations with the S1P1 receptor were performed to further evaluate FTY720 regioisomer signaling. RESULTS FTY720 regioisomers produced potent endothelial cell barrier disruption reflected by declines in TER alterations. Pharmacologic inhibition of Gi-coupled S1P receptors (S1P1, S1P2, S1P3) failed to alter FTY720 regioisomer-mediated barrier disruption; findings that were corroborated by docking simulations demonstrating FTY720 regiosomers were repelled from S1P1 docking, in contrast to strong S1P1 binding elicited by S1P. Inhibition of either the barrier-disrupting PAR-1 receptor, the VEGF receptor, Rho-kinase, MAPK, NFkB, or PI3K failed to alter FTY720 regioisomer-induced endothelial cell barrier disruption. While FTY720 regioisomers significantly increased protein phosphatase 2 (PP2A) activity, PP2A inhibitors failed to alter FTY720 regioisomer-induced endothelial cell barrier disruption. CONCLUSIONS Together, these results imply a vexing model of pulmonary vascular barrier dysregulation in response to FTY720-related compounds and highlight the need for further insights into mechanisms of vascular integrity required to promote the development of novel therapeutic tools to prevent or reverse the pulmonary vascular leak central to ARDS outcomes.
Collapse
Affiliation(s)
- Sara M. Camp
- Department of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Alexander Marciniak
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, USA
| | - Eddie T. Chiang
- Department of Medicine, The University of Arizona, Tucson, AZ, USA
| | - Alexander N. Garcia
- Department of Radiation Oncology, The University of Arizona, Tucson, AZ, USA
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, NY, USA
| | - Robin Polt
- Department of Chemistry and Biochemistry, The University of Arizona, Tucson, AZ, USA
| | - Ruth G. Perez
- Department of Molecular and Translational Medicine, Graduate School of Biomedical Sciences, Center of Emphasis in Neuroscience, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center, El Paso, TX, USA
| | - Steven M. Dudek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Joe G. N. Garcia
- Department of Medicine, The University of Arizona, Tucson, AZ, USA
| |
Collapse
|
3
|
Zhao J, Zhu M, Jiang H, Shen S, Su X, Shi Y. Combination of sphingosine-1-phosphate receptor 1 (S1PR1) agonist and antiviral drug: a potential therapy against pathogenic influenza virus. Sci Rep 2019; 9:5272. [PMID: 30918324 PMCID: PMC6437142 DOI: 10.1038/s41598-019-41760-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 03/13/2019] [Indexed: 01/13/2023] Open
Abstract
The pandemic 2009 influenza A H1N1 virus is associated with significant mortality. Targeting S1PR1, which is known to modulate the immune response, provides protection against pathogenic influenza virus. The functional role and molecular mechanism of S1PR1 were analysed by generating inducible endothelial cell-specific S1PR1 knockout mice and assessing the therapeutic efficacy of the selective S1PR1 agonist CYM5442 against acute lung injury (ALI) induced by the 2009 influenza A H1N1 virus. Immune-mediated pulmonary injury is aggravated by the absence of endothelial S1PR1 and alleviated by treatment with CYM-5442, suggesting a protective function of S1PR1 signaling during H1N1 infection. S1PR1 signaling does not affect viral clearance in mice infected with influenza. Mechanistically, the MAPK and NF-kB signaling pathways are involved in the ALI mediated by S1PR1 in infected mice. Combined administration of the S1PR1 agonist CYM-5442 and the antiviral drug oseltamivir provides maximum protection from ALI. Our current study provides insight into the molecular mechanism of S1PR1 mediating the ALI induced by H1N1 infection and indicates that the combination of S1PR1 agonist with antiviral drug could potentially be used as a therapeutic remedy for future H1N1 virus pandemics.
Collapse
Affiliation(s)
- Jiangnan Zhao
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Meiying Zhu
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Hao Jiang
- Department of Emergency Medicine, the Second Affiliated Hospital, Southeast University, Nanjing, 210002, China
| | - Simen Shen
- Department of Respiratory Medicine, the First People's Hospital of Nantong, Nantong, 226000, China
| | - Xin Su
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China
| | - Yi Shi
- Department of Respiratory and Critical Medicine, Jinling Hospital, Nanjing University School of Medicine, Nanjing, 210002, China.
| |
Collapse
|
4
|
Hou X, Zhang H, Chen BC, Guo Z, Singh A, Goswami A, Gilmore JL, Sheppeck JE, Dyckman AJ, Carter PH, Mathur A. Regioselective Epoxide Ring Opening for the Stereospecific Scale-Up Synthesis of BMS-960, A Potent and Selective Isoxazole-Containing S1P1 Receptor Agonist. Org Process Res Dev 2017. [DOI: 10.1021/acs.oprd.6b00366] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Xiaoping Hou
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - Huiping Zhang
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - Bang-Chi Chen
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - Zhiwei Guo
- Chemical & Synthetic Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Amarjit Singh
- Chemical & Synthetic Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - Animesh Goswami
- Chemical & Synthetic Development, Bristol-Myers Squibb, New Brunswick, New Jersey 08903, United States
| | - John L. Gilmore
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - James E. Sheppeck
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - Alaric J. Dyckman
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - Percy H. Carter
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| | - Arvind Mathur
- Discovery
Chemistry, Bristol-Myers Squibb, Princeton, New Jersey 08540, United States
| |
Collapse
|
5
|
Novel S1P 1 receptor agonists – Part 4: Alkylaminomethyl substituted aryl head groups. Eur J Med Chem 2016; 116:222-238. [DOI: 10.1016/j.ejmech.2016.03.048] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Revised: 12/17/2015] [Accepted: 03/18/2016] [Indexed: 12/12/2022]
|
6
|
Bolli MH, Lescop C, Birker M, de Kanter R, Hess P, Kohl C, Nayler O, Rey M, Sieber P, Velker J, Weller T, Steiner B. Novel S1P1 receptor agonists – Part 5: From amino-to alkoxy-pyridines. Eur J Med Chem 2016; 115:326-41. [DOI: 10.1016/j.ejmech.2016.03.020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 03/07/2016] [Accepted: 03/09/2016] [Indexed: 12/15/2022]
|
7
|
Camp SM, Chiang ET, Sun C, Usatyuk PV, Bittman R, Natarajan V, Garcia JGN, Dudek SM. "Pulmonary Endothelial Cell Barrier Enhancement by Novel FTY720 Analogs: Methoxy-FTY720, Fluoro-FTY720, and β-Glucuronide-FTY720". Chem Phys Lipids 2015; 194:85-93. [PMID: 26496151 DOI: 10.1016/j.chemphyslip.2015.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/28/2022]
Abstract
Effective therapeutic agents are lacking for the prevention and reversal of vascular leak, a frequent pathophysiologic result of inflammatory processes such as acute respiratory distress syndrome (ARDS) and sepsis. We previously demonstrated the potent barrier-enhancing effects of related compounds sphingosine 1-phosphate (S1P), the pharmaceutical agent FTY720, and its analog (S)-FTY720 phosphonate (Tys) in models of inflammatory lung injury. In this study, we characterize additional novel FTY720 analogs for their potential to reduce vascular leak as well as utilize them as tools to better understand the mechanisms by which this class of agents modulates permeability. Transendothelial resistance (TER) and labeled dextran studies demonstrate that (R)-methoxy-FTY720 ((R)-OMe-FTY), (R)/(S)-fluoro-FTY720 (FTY-F), and β-glucuronide-FTY720 (FTY-G) compounds display in vitro barrier-enhancing properties comparable or superior to FTY720 and S1P. In contrast, the (S)-methoxy-FTY720 ((S)-OMe-FTY) analog disrupts lung endothelial cell (EC) barrier integrity in TER studies in association with actin stress fiber formation and robust intracellular calcium release, but independent of myosin light chain or ERK phosphorylation. Additional mechanistic studies with (R)-OMe-FTY, FTY-F, and FTY-G suggest that lung EC barrier enhancement is mediated through lipid raft signaling, Gi-linked receptor coupling to downstream tyrosine phosphorylation events, and S1PR1-dependent receptor ligation. These results provide important mechanistic insights into modulation of pulmonary vascular barrier function by FTY720-related compounds and highlight common signaling events that may assist the development of novel therapeutic tools in the prevention or reversal of the pulmonary vascular leak that characterizes ARDS.
Collapse
Affiliation(s)
- Sara M Camp
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - Eddie T Chiang
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - Chaode Sun
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York, NY, United States
| | - Peter V Usatyuk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York, NY, United States
| | - Viswanathan Natarajan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Joe G N Garcia
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - Steven M Dudek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
8
|
Camp SM, Chiang ET, Sun C, Usatyuk PV, Bittman R, Natarajan V, Garcia JGN, Dudek SM. Pulmonary endothelial cell barrier enhancement by novel FTY720 analogs: methoxy-FTY720, fluoro-FTY720, and β-glucuronide-FTY720. Chem Phys Lipids 2015; 191:16-24. [PMID: 26272033 DOI: 10.1016/j.chemphyslip.2015.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 08/05/2015] [Accepted: 08/06/2015] [Indexed: 12/14/2022]
Abstract
Effective therapeutic agents are lacking for the prevention and reversal of vascular leak, a frequent pathophysiologic result of inflammatory processes such as acute respiratory distress syndrome (ARDS) and sepsis. We previously demonstrated the potent barrier-enhancing effects of related compounds sphingosine 1-phosphate (S1P), the pharmaceutical agent FTY720, and its analog (S)-FTY720 phosphonate (Tys) in models of inflammatory lung injury. In this study, we characterize additional novel FTY720 analogs for their potential to reduce vascular leak as well as utilize them as tools to better understand the mechanisms by which this class of agents modulates permeability. Transendothelial resistance (TER) and labeled dextran studies demonstrate that (R)-methoxy-FTY720 ((R)-OMe-FTY), (R)/(S)-fluoro-FTY720 (FTY-F), and β-glucuronide-FTY720 (FTY-G) compounds display in vitro barrier-enhancing properties comparable or superior to FTY720 and S1P. In contrast, the (S)-methoxy-FTY720 ((S)-OMe-FTY) analog disrupts lung endothelial cell (EC) barrier integrity in TER studies in association with actin stress fiber formation and robust intracellular calcium release, but independent of myosin light chain or ERK phosphorylation. Additional mechanistic studies with (R)-OMe-FTY, FTY-F, and FTY-G suggest that lung EC barrier enhancement is mediated through lipid raft signaling, Gi-linked receptor coupling to downstream tyrosine phosphorylation events, and S1PR1-dependent receptor ligation. These results provide important mechanistic insights into modulation of pulmonary vascular barrier function by FTY720-related compounds and highlight common signaling events that may assist the development of novel therapeutic tools in the prevention or reversal of the pulmonary vascular leak that characterizes ARDS.
Collapse
Affiliation(s)
- Sara M Camp
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - Eddie T Chiang
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - Chaode Sun
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York, NY, United States
| | - Peter V Usatyuk
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Robert Bittman
- Department of Chemistry and Biochemistry, Queens College of the City University of New York, Flushing, New York, NY, United States
| | - Viswanathan Natarajan
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - Joe G N Garcia
- Department of Medicine and Arizona Respiratory Center, University of Arizona, Tucson, AZ, United States
| | - Steven M Dudek
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States.
| |
Collapse
|
9
|
FTY720 (s)-phosphonate preserves sphingosine 1-phosphate receptor 1 expression and exhibits superior barrier protection to FTY720 in acute lung injury. Crit Care Med 2014; 42:e189-99. [PMID: 24335440 DOI: 10.1097/ccm.0000000000000097] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Effective therapies are needed to reverse the increased vascular permeability that characterizes acute inflammatory diseases such as acute lung injury. FTY720 is a pharmaceutical analog of the potent barrier-enhancing phospholipid, sphingosine 1-phosphate. Because both FTY720 and sphingosine 1-phosphate have properties that may limit their usefulness in patients with acute lung injury, alternative compounds are needed for therapeutic use. The objective of this study is to characterize the effects of FTY720 (S)-phosphonate, a novel analog of FTY720-phosphate, on variables of pulmonary vascular permeability in vitro and alveolar-capillary permeability in vivo. SETTING University-affiliated research institute. SUBJECTS Cultured human pulmonary endothelial cells; C57BL/6 mice. INTERVENTIONS Endothelial cells were stimulated with sphingosine 1-phosphate receptor 1 agonists to determine effects on sphingosine 1-phosphate receptor 1 expression. Acute lung injury was induced in C57BL/6 mice with bleomycin to assess effects of sphingosine 1-phosphate receptor 1 agonists. MEASUREMENTS AND MAIN RESULTS FTY720 (S)-phosphonate potently increases human pulmonary endothelial cell barrier function in vitro as measured by transendothelial electrical resistance. Reduction of sphingosine 1-phosphate receptor 1 with small interference RNA significantly attenuates this transendothelial electrical resistance elevation. FTY720 (S)-phosphonate maintains endothelial sphingosine 1-phosphate receptor 1 protein expression in contrast to greater than 50% reduction after incubation with sphingosine 1-phosphate, FTY720, or other sphingosine 1-phosphate receptor 1 agonists. FTY720 (S)-phosphonate does not induce β-arrestin recruitment, sphingosine 1-phosphate receptor 1 ubiquitination, and proteosomal degradation that occur after other agonists. Intraperitoneal administration of FTY720 (S)-phosphonate every other day for 1 week in normal or bleomycin-injured mice maintains significantly higher lung sphingosine 1-phosphate receptor 1 expression compared with FTY720. FTY720 fails to protect against bleomycin-induced acute lung injury in mice, while FTY720 (S)-phosphonate significantly decreases lung leak and inflammation. CONCLUSION FTY720 (S)-phosphonate is a promising barrier-promoting agent that effectively maintains sphingosine 1-phosphate receptor 1 levels and improves outcomes in the bleomycin model of acute lung injury.
Collapse
|
10
|
Leoni A, Locatelli A, Morigi R, Rambaldi M. Novel thiazole derivatives: a patent review (2008 – 2012. Part 2). Expert Opin Ther Pat 2014; 24:759-77. [DOI: 10.1517/13543776.2014.910196] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
11
|
Xu J, Gray F, Henderson A, Hicks K, Yang J, Thompson P, Oliver J. Safety, pharmacokinetics, pharmacodynamics, and bioavailability of GSK2018682, a sphingosine-1-phosphate receptor modulator, in healthy volunteers. Clin Pharmacol Drug Dev 2014; 3:170-8. [PMID: 27128606 DOI: 10.1002/cpdd.98] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 12/09/2013] [Indexed: 11/09/2022]
Abstract
The tolerability, pharmacokinetics, and pharmacodynamics of single (SD) and repeat (RD) doses of GSK2018682, a selective S1P1 receptor modulator, were evaluated in healthy volunteers. The bioavailability (BA) of different formulations and effects of food were also evaluated. SD of up to 24 mg and RD of up to 6 mg/day for 28 days were reasonably tolerated, despite higher incidences of gastrointestinal and cardiovascular adverse events compared to placebo. There was a linear relationship between dose and systemic exposure with a dose-independent half-life (t1/2 ) between 44.9 and 63.3 hours. GSK2018682 induced acute, transient and non-symptomatic decreases in heart rate and blood pressure. Dose-dependent reduction in absolute lymphocyte count (ALC), and all tested subsets, was observed to various degrees, up to a nadir of over 70% reduction from baseline. There was no difference in major pharmacokinetic parameters among three formulations of GSK2018682 and between fasted and fed subjects. However, there was a reduction in the extent of bradycardia following dosing in the fed state. Additionally, exercise induced robust increase in heart rate in subjects who had bradycardia following RD of GSK2018682 up to 6 mg, suggesting possible physiological methods of reducing the extent of S1P mediated bradycardia and subsequent AV-block.
Collapse
Affiliation(s)
- Jianfeng Xu
- GlaxoSmithKline Research and Development Center, Shanghai, China
| | - Frank Gray
- GlaxoSmithKline Research and Development Center, Shanghai, China
| | - Alex Henderson
- GlaxoSmithKline Research and Development Center, Shanghai, China
| | - Kirsty Hicks
- GlaxoSmithKline Research and Development Center, Shanghai, China
| | - Jiansong Yang
- GlaxoSmithKline Research and Development Center, Shanghai, China
| | - Paul Thompson
- GlaxoSmithKline Research and Development Center, Shanghai, China
| | - Johann Oliver
- GlaxoSmithKline Research and Development Center, Shanghai, China
| |
Collapse
|
12
|
Bolli MH, Abele S, Birker M, Bravo R, Bur D, de Kanter R, Kohl C, Grimont J, Hess P, Lescop C, Mathys B, Müller C, Nayler O, Rey M, Scherz M, Schmidt G, Seifert J, Steiner B, Velker J, Weller T. Novel S1P(1) receptor agonists--part 3: from thiophenes to pyridines. J Med Chem 2013; 57:110-30. [PMID: 24367923 DOI: 10.1021/jm4014696] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In preceding communications we summarized our medicinal chemistry efforts leading to the identification of potent, selective, and orally active S1P1 agonists such as the thiophene derivative 1. As a continuation of these efforts, we replaced the thiophene in 1 by a 2-, 3-, or 4-pyridine and obtained less lipophilic, potent, and selective S1P1 agonists (e.g., 2) efficiently reducing blood lymphocyte count in the rat. Structural features influencing the compounds' receptor affinity profile and pharmacokinetics are discussed. In addition, the ability to penetrate brain tissue has been studied for several compounds. As a typical example for these pyridine based S1P1 agonists, compound 53 showed EC50 values of 0.6 and 352 nM for the S1P1 and S1P3 receptor, respectively, displayed favorable PK properties, and penetrated well into brain tissue. In the rat, compound 53 maximally reduced the blood lymphocyte count for at least 24 h after oral dosing of 3 mg/kg.
Collapse
Affiliation(s)
- Martin H Bolli
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd. , Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Bolli MH, Velker J, Müller C, Mathys B, Birker M, Bravo R, Bur D, de Kanter R, Hess P, Kohl C, Lehmann D, Meyer S, Nayler O, Rey M, Scherz M, Steiner B. Novel S1P1 Receptor Agonists - Part 2: From Bicyclo[3.1.0]hexane-Fused Thiophenes to Isobutyl Substituted Thiophenes. J Med Chem 2013; 57:78-97. [DOI: 10.1021/jm401456d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Martin H. Bolli
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Jörg Velker
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Claus Müller
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Boris Mathys
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Magdalena Birker
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Roberto Bravo
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Daniel Bur
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Ruben de Kanter
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Patrick Hess
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Christopher Kohl
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - David Lehmann
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Solange Meyer
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Oliver Nayler
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Markus Rey
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Michael Scherz
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| | - Beat Steiner
- Drug Discovery
Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, CH-4123 Allschwil, Switzerland
| |
Collapse
|
14
|
Bolli MH, Müller C, Mathys B, Abele S, Birker M, Bravo R, Bur D, Hess P, Kohl C, Lehmann D, Nayler O, Rey M, Meyer S, Scherz M, Schmidt G, Steiner B, Treiber A, Velker J, Weller T. Novel S1P1 Receptor Agonists – Part 1: From Pyrazoles to Thiophenes. J Med Chem 2013; 56:9737-55. [DOI: 10.1021/jm4014373] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Martin H. Bolli
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Claus Müller
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Boris Mathys
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Stefan Abele
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Magdalena Birker
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Roberto Bravo
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Daniel Bur
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Patrick Hess
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Christopher Kohl
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - David Lehmann
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Oliver Nayler
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Markus Rey
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Solange Meyer
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Michael Scherz
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Gunther Schmidt
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Beat Steiner
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Alexander Treiber
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Jörg Velker
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| | - Thomas Weller
- Drug Discovery Chemistry, Actelion Pharmaceuticals Ltd., Gewerbestrasse 16, Allschwil CH-4123, Switzerland
| |
Collapse
|
15
|
Bigaud M, Guerini D, Billich A, Bassilana F, Brinkmann V. Second generation S1P pathway modulators: research strategies and clinical developments. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1841:745-58. [PMID: 24239768 DOI: 10.1016/j.bbalip.2013.11.001] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2013] [Revised: 10/30/2013] [Accepted: 11/04/2013] [Indexed: 11/17/2022]
Abstract
Multiple Sclerosis (MS) is a chronic autoimmune disorder affecting the central nervous system (CNS) through demyelination and neurodegeneration. Until recently, major therapeutic treatments have relied on agents requiring injection delivery. In September 2010, fingolimod/FTY720 (Gilenya, Novartis) was approved as the first oral treatment for relapsing forms of MS. Fingolimod causes down-modulation of S1P1 receptors on lymphocytes which prevents the invasion of autoaggressive T cells into the CNS. In astrocytes, down-modulation of S1P1 by the drug reduces astrogliosis, a hallmark of MS, thereby allowing restoration of productive astrocyte communication with other neural cells and the blood brain barrier. Animal data further suggest that the drug directly supports the recovery of nerve conduction and remyelination. In human MS, such mechanisms may explain the significant decrease in the number of inflammatory markers on brain magnetic resonance imaging in recent clinical trials, and the reduction of brain atrophy by the drug. Fingolimod binds to 4 of the 5 known S1P receptor subtypes, and significant efforts were made over the past 5 years to develop next generation S1P receptor modulators and determine the minimal receptor selectivity needed for maximal therapeutic efficacy in MS patients. Other approaches considered were competitive antagonists of the S1P1 receptor, inhibitors of the S1P lyase to prevent S1P degradation, and anti-S1P antibodies. Below we discuss the current status of the field, and the functional properties of the most advanced compounds. This article is part of a Special Issue entitled New Frontiers in Sphingolipid Biology.
Collapse
Affiliation(s)
- Marc Bigaud
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| | - Danilo Guerini
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | - Andreas Billich
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland
| | | | - Volker Brinkmann
- Novartis Institutes for Biomedical Research, CH-4056 Basel, Switzerland.
| |
Collapse
|
16
|
Harris RM, Andrews BI, Clark S, Cooke JWB, Gray JCS, Ng SQQ. The Fit For Purpose Development of S1P1 Receptor Agonist GSK2263167 Using a Robinson Annulation and Saegusa Oxidation to Access an Advanced Phenol Intermediate. Org Process Res Dev 2013. [DOI: 10.1021/op400162p] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Robert M. Harris
- Chemical Development, GlaxoSmithKline Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Benjamin I. Andrews
- Chemical Development, GlaxoSmithKline Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Stacy Clark
- Chemical Development, GlaxoSmithKline Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Jason W. B. Cooke
- Chemical Development, GlaxoSmithKline Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - John C. S. Gray
- Chemical Development, GlaxoSmithKline Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| | - Stephanie Q. Q. Ng
- Chemical Development, GlaxoSmithKline Research and Development Ltd., Gunnels Wood Road, Stevenage, Hertfordshire SG1 2NY, U.K
| |
Collapse
|
17
|
Fryer RM, Muthukumarana A, Harrison PC, Nodop Mazurek S, Chen RR, Harrington KE, Dinallo RM, Horan JC, Patnaude L, Modis LK, Reinhart GA. The clinically-tested S1P receptor agonists, FTY720 and BAF312, demonstrate subtype-specific bradycardia (S1P₁) and hypertension (S1P₃) in rat. PLoS One 2012; 7:e52985. [PMID: 23285242 PMCID: PMC3532212 DOI: 10.1371/journal.pone.0052985] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 11/22/2012] [Indexed: 12/21/2022] Open
Abstract
Sphingosine-1-phospate (S1P) and S1P receptor agonists elicit mechanism-based effects on cardiovascular function in vivo. Indeed, FTY720 (non-selective S1P(X) receptor agonist) produces modest hypertension in patients (2-3 mmHg in 1-yr trial) as well as acute bradycardia independent of changes in blood pressure. However, the precise receptor subtypes responsible is controversial, likely dependent upon the cardiovascular response in question (e.g. bradycardia, hypertension), and perhaps even species-dependent since functional differences in rodent, rabbit, and human have been suggested. Thus, we characterized the S1P receptor subtype specificity for each compound in vitro and, in vivo, the cardiovascular effects of FTY720 and the more selective S1P₁,₅ agonist, BAF312, were tested during acute i.v. infusion in anesthetized rats and after oral administration for 10 days in telemetry-instrumented conscious rats. Acute i.v. infusion of FTY720 (0.1, 0.3, 1.0 mg/kg/20 min) or BAF312 (0.5, 1.5, 5.0 mg/kg/20 min) elicited acute bradycardia in anesthetized rats demonstrating an S1P₁ mediated mechanism-of-action. However, while FTY720 (0.5, 1.5, 5.0 mg/kg/d) elicited dose-dependent hypertension after multiple days of oral administration in rat at clinically relevant plasma concentrations (24-hr mean blood pressure = 8.4, 12.8, 16.2 mmHg above baseline vs. 3 mmHg in vehicle controls), BAF312 (0.3, 3.0, 30.0 mg/kg/d) had no significant effect on blood pressure at any dose tested suggesting that hypertension produced by FTY720 is mediated S1P₃ receptors. In summary, in vitro selectivity results in combination with studies performed in anesthetized and conscious rats administered two clinically tested S1P agonists, FTY720 or BAF312, suggest that S1P₁ receptors mediate bradycardia while hypertension is mediated by S1P₃ receptor activation.
Collapse
Affiliation(s)
- Ryan M Fryer
- Department of Cardiometabolic Disease Research, Boehringer-Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut, United States of America.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Integrating the puzzle pieces: the current atomistic picture of phospholipid-G protein coupled receptor interactions. Biochim Biophys Acta Mol Cell Biol Lipids 2012; 1831:2-12. [PMID: 22982815 DOI: 10.1016/j.bbalip.2012.09.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2012] [Revised: 08/31/2012] [Accepted: 09/03/2012] [Indexed: 01/09/2023]
Abstract
A compelling question of how phospholipids interact with their target receptors has been of interest since the first receptor-mediated effects were reported. The recent report of a crystal structure for the S1P(1) receptor in complex with an antagonist phospholipid provides interesting perspective on the insights that had previously been gained through structure-activity studies of the phospholipids, as well as modeling and mutagenesis studies of the receptors. This review integrates these varied lines of investigation in the context of their various contributions to our current understanding of phospholipid-receptor interactions. This article is part of a Special Issue entitled Advances in Lysophospholipid Research.
Collapse
|
19
|
Deng G, Meng Q, Liu Q, Xu X, Xu Q, Ren F, Guo TB, Lu H, Xiang JN, Elliott JD, Lin X. Identification of benzoxazole analogs as novel, S1P(3) sparing S1P(1) agonists. Bioorg Med Chem Lett 2012; 22:3973-7. [PMID: 22583616 DOI: 10.1016/j.bmcl.2012.04.095] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2012] [Revised: 04/19/2012] [Accepted: 04/20/2012] [Indexed: 10/28/2022]
Abstract
A novel series of benzoxazole-derived S1P(1) agonists were designed based on scaffold hopping molecular design strategy combined with computational approaches. Extensive SAR studies led to the discovery of compound 17d as a selective S1P(1) agonist (over S1P(3)) with high CNS penetration and favorable DMPK properties. 17d also demonstrated in vivo pharmacological efficacy to reduce blood lymphocyte in mice after oral administration.
Collapse
Affiliation(s)
- Guanghui Deng
- Research and Development, GlaxoSmithKline Pharmaceuticals, 898 Halei Road, Zhangjiang Hi-tech Park, Pudong, Shanghai 201023, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Recent Advances in the Discovery and Development of Sphingosine-1-Phosphate-1 Receptor Agonists. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2012. [DOI: 10.1016/b978-0-12-396492-2.00013-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
21
|
Mathew B, Jacobson JR, Berdyshev E, Huang Y, Sun X, Zhao Y, Gerhold LM, Siegler J, Evenoski C, Wang T, Zhou T, Zaidi R, Moreno-Vinasco L, Bittman R, Chen CT, LaRiviere PJ, Sammani S, Lussier YA, Dudek SM, Natarajan V, Weichselbaum RR, Garcia JGN. Role of sphingolipids in murine radiation-induced lung injury: protection by sphingosine 1-phosphate analogs. FASEB J 2011; 25:3388-400. [PMID: 21712494 DOI: 10.1096/fj.11-183970] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Clinically significant radiation-induced lung injury (RILI) is a common toxicity in patients administered thoracic radiotherapy. Although the molecular etiology is poorly understood, we previously characterized a murine model of RILI in which alterations in lung barrier integrity surfaced as a potentially important pathobiological event and genome-wide lung gene mRNA levels identified dysregulation of sphingolipid metabolic pathway genes. We hypothesized that sphingolipid signaling components serve as modulators and novel therapeutic targets of RILI. Sphingolipid involvement in murine RILI was confirmed by radiation-induced increases in lung expression of sphingosine kinase (SphK) isoforms 1 and 2 and increases in the ratio of ceramide to sphingosine 1-phosphate (S1P) and dihydro-S1P (DHS1P) levels in plasma, bronchoalveolar lavage fluid, and lung tissue. Mice with a targeted deletion of SphK1 (SphK1(-/-)) or with reduced expression of S1P receptors (S1PR1(+/-), S1PR2(-/-), and S1PR3(-/-)) exhibited marked RILI susceptibility. Finally, studies of 3 potent vascular barrier-protective S1P analogs, FTY720, (S)-FTY720-phosphonate (fTyS), and SEW-2871, identified significant RILI attenuation and radiation-induced gene dysregulation by the phosphonate analog, fTyS (0.1 and 1 mg/kg i.p., 2×/wk) and to a lesser degree by SEW-2871 (1 mg/kg i.p., 2×/wk), compared with those in controls. These results support the targeting of S1P signaling as a novel therapeutic strategy in RILI.
Collapse
Affiliation(s)
- Biji Mathew
- Institute for Personalized Respiratory Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, University of Illinois at Chicago, Chicago, Illinois 60612, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Demont EH, Andrews BI, Bit RA, Campbell CA, Cooke JWB, Deeks N, Desai S, Dowell SJ, Gaskin P, Gray JRJ, Haynes A, Holmes DS, Kumar U, Morse MA, Osborne GJ, Panchal T, Patel B, Perboni A, Taylor S, Watson R, Witherington J, Willis R. Discovery of a Selective S1P1 Receptor Agonist Efficacious at Low Oral Dose and Devoid of Effects on Heart Rate. ACS Med Chem Lett 2011; 2:444-9. [PMID: 24900328 DOI: 10.1021/ml2000214] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Accepted: 03/24/2011] [Indexed: 11/29/2022] Open
Abstract
Gilenya (fingolimod, FTY720) was recently approved by the U.S. FDA for the treatment of patients with remitting relapsing multiple sclerosis (RRMS). It is a potent agonist of four of the five sphingosine 1-phosphate (S1P) G-protein-coupled receptors (S1P1 and S1P3-5). It has been postulated that fingolimod's efficacy is due to S1P1 agonism, while its cardiovascular side effects (transient bradycardia and hypertension) are due to S1P3 agonism. We have discovered a series of selective S1P1 agonists, which includes 3-[6-(5-{3-cyano-4-[(1-methylethyl)oxy]phenyl}-1,2,4-oxadiazol-3-yl)-5-methyl-3,4-dihydro-2(1H)-isoquinolinyl]propanoate, 20, a potent, S1P3-sparing, orally active S1P1 agonist. Compound 20 is as efficacious as fingolimod in a collagen-induced arthritis model and shows excellent pharmacokinetic properties preclinically. Importantly, the selectivity of 20 against S1P3 is responsible for an absence of cardiovascular signal in telemetered rats, even at high dose levels.
Collapse
Affiliation(s)
- Emmanuel H. Demont
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Benjamin I. Andrews
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Rino A. Bit
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Colin A. Campbell
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Jason W. B. Cooke
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Nigel Deeks
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Sapna Desai
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Simon J. Dowell
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Pam Gaskin
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - James R. J. Gray
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Andrea Haynes
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Duncan S. Holmes
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Umesh Kumar
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Mary A. Morse
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Greg J. Osborne
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Terry Panchal
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Bela Patel
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Alcide Perboni
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Simon Taylor
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Robert Watson
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Jason Witherington
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| | - Robert Willis
- Immuno Inflammation Center of Excellence for Drug Discovery and ‡Platform Technology and Science, GlaxoSmithKline, Gunnels Wood Road, Stevenage, SG1 2NY, United Kingdom
| |
Collapse
|
23
|
S1P receptor mediated activity of FTY720 phosphate mimics. Bioorg Med Chem Lett 2010; 20:1485-7. [DOI: 10.1016/j.bmcl.2010.01.118] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Revised: 01/20/2010] [Accepted: 01/21/2010] [Indexed: 11/19/2022]
|
24
|
Camp SM, Bittman R, Chiang ET, Moreno-Vinasco L, Mirzapoiazova T, Sammani S, Lu X, Sun C, Harbeck M, Roe M, Natarajan V, Garcia JGN, Dudek SM. Synthetic analogs of FTY720 [2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol] differentially regulate pulmonary vascular permeability in vivo and in vitro. J Pharmacol Exp Ther 2009; 331:54-64. [PMID: 19592667 PMCID: PMC2766218 DOI: 10.1124/jpet.109.153544] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2009] [Accepted: 07/09/2009] [Indexed: 12/31/2022] Open
Abstract
Novel therapies are needed to address the vascular endothelial cell (EC) barrier disruption that occurs in inflammatory diseases such as acute lung injury (ALI). We previously demonstrated the potent barrier-enhancing effects of both sphingosine 1-phosphate (S1P) and the structurally similar compound FTY720 [2-amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol] in inflammatory lung injury. In this study, we examined the therapeutic potential of several novel FTY720 analogs to reduce vascular leak. Similar to S1P and FTY720, the (R)- and (S)-enantiomers of FTY720 phosphonate and enephosphonate analogs produce sustained EC barrier enhancement in vitro, as seen by increases in transendothelial electrical resistance (TER). In contrast, the (R)- and (S)-enantiomers of FTY720-regioisomeric analogs disrupt EC barrier integrity in a dose-dependent manner. Barrier-enhancing FTY720 analogs demonstrate a wider protective concentration range in vitro (1-50 microM) and greater potency than either S1P or FTY720. In contrast to FTY720-induced EC barrier enhancement, S1P and the FTY720 analogs dramatically increase TER within minutes in association with cortical actin ring formation. Unlike S1P, these FTY720 analogs exhibit differential phosphorylation effects without altering the intracellular calcium level. Inhibitor studies indicate that barrier enhancement by these analogs involves signaling via G(i)-coupled receptors, tyrosine kinases, and lipid rafts. Consistent with these in vitro responses, the (S)-phosphonate analog of FTY720 significantly reduces multiple indices of alveolar and vascular permeability in a lipopolysaccharide-mediated murine model of ALI (without significant alterations in leukocyte counts). These results demonstrate the capacity for FTY720 analogs to significantly decrease pulmonary vascular leakage and inflammation in vitro and in vivo.
Collapse
Affiliation(s)
- S M Camp
- Department of Medicine, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Rosen H, Gonzalez-Cabrera PJ, Sanna MG, Brown S. Sphingosine 1-phosphate receptor signaling. Annu Rev Biochem 2009; 78:743-68. [PMID: 19231986 DOI: 10.1146/annurev.biochem.78.072407.103733] [Citation(s) in RCA: 327] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The sphingosine 1-phosphate (S1P) receptor signaling system is a productive model system. A hydrophobic zwitterionic lysophospholipid ligand with difficult physical properties interacts with five high-affinity G protein-coupled receptors to generate multiple downstream signals. These signals modulate homeostasis and pathology on a steep agonist concentration-response curve. Ligand presence is essential for vascular development and endothelial integrity, while acute increases in ligand concentrations result in cardiac death. Understanding this integrated biochemical system has exemplified the impact of both genetics and chemistry. Developing specific tools with defined biochemical properties for the reversible modulation of signals in real time has been essential to complement insights gained from genetic approaches that may be irreversible and compensated. Despite its knife-edge between life and death, this system, based in part on receptor subtype-selectivity and in part on differential attenuation of deleterious signals, now appears to be on the cusp of meaningful therapy for multiple sclerosis.
Collapse
Affiliation(s)
- Hugh Rosen
- Departments of Chemical Physiology and Immunology and The Scripps Research Institute Molecular Screening Center, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
26
|
Hosomizu K, Oodoi M, Umeyama T, Matano Y, Yoshida K, Isoda S, Isosomppi M, Tkachenko NV, Lemmetyinen H, Imahori H. Substituent effects of porphyrins on structures and photophysical properties of amphiphilic porphyrin aggregates. J Phys Chem B 2009; 112:16517-24. [PMID: 19053673 DOI: 10.1021/jp807991k] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Substituent effects of porphyrin on the structures and photophysical properties of the J-aggregates of protonated 5-(4-alkoxyphenyl)-10,15,20-tris(4-sulfonatophenyl)porphyrin have been examined for the first time. Selective formation of the porphyrin J-aggregate was attained when suitable length of the alkoxy group was employed for the amphiphilic porphyrin. Namely, a regular leaflike structure was observed for the J-aggregates of protonated 5-(4-octyloxyphenyl)-10,15,20-tris(4-sulfonatophenyl)porphyrin, which was consistent with the results obtained by using the UV-visible absorption and dynamic light-scattering measurements. A bilayer structure in which the hydrophobic alkoxyl groups facing inside the bilayer are interdigitated to each other, whereas the hydrophilic porphyrin moieties are exposed outside, was proposed to explain the unique porphyrin J-aggregate. Fast energy migration and efficient quenching by defect site in the J-aggregates were suggested to rationalize the short lifetimes of the excited J-aggregates.
Collapse
Affiliation(s)
- Kohei Hosomizu
- Department of Molecular Engineering, Graduate School of Engineering, Kyoto University, Nishikyo-ku, Kyoto 615-8510, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Biological Activity of Aminophosphonic Acids and Their Short Peptides. TOPICS IN HETEROCYCLIC CHEMISTRY 2009. [DOI: 10.1007/7081_2008_14] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
28
|
Delgado A, Casas J, Llebaria A, Abad JL, Fabriás G. Chemical tools to investigate sphingolipid metabolism and functions. ChemMedChem 2008; 2:580-606. [PMID: 17252619 DOI: 10.1002/cmdc.200600195] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sphingolipids comprise an important group of biomolecules, some of which have been shown to play important roles in the regulation of many cell functions. From a structural standpoint, they all share a long 2-amino-1,3-diol chain, which can be either saturated (sphinganine), hydroxylated at C4 (phytosphingosine), or unsaturated at C4 (sphingosine) as in most mammalian cells. N-acylation of sphingosine leads to ceramide, a key intermediate in sphingolipid metabolism that can be enzymatically modified at the C1-OH position to other biologically important sphingolipids, such as sphingomyelin or glycosphingolipids. In addition, both ceramide and sphingosine can be phosphorylated at C1-OH to give ceramide-1-phosphate and sphingosine-1-phosphate, respectively. To better understand the biological and biophysical roles of sphingolipids, many efforts have been made to design synthetic analogues as chemical tools able to unravel their structure-activity relationships, and to alter their cellular levels. This last approach has been thoroughly studied by the development of specific inhibitors of some key enzymes that play an important role in biosynthesis or metabolism of these intriguing lipids. With the above premises in mind, the aim of this review is to collect, in a systematic way, the recent efforts described in the literature leading to the development of new chemical entities specifically designed to achieve the above goals.
Collapse
Affiliation(s)
- Antonio Delgado
- Research Unit on Bioactive Molecules, Departament de Química Orgànica Biològica, Institut d'Investigacions Químiques i Ambientals de Barcelona (C.S.I.C); Jordi Girona 18-26, 08034 Barcelona, Spain.
| | | | | | | | | |
Collapse
|
29
|
Gamliel A, Afri M, Frimer AA. Determining radical penetration of lipid bilayers with new lipophilic spin traps. Free Radic Biol Med 2008; 44:1394-405. [PMID: 18226602 DOI: 10.1016/j.freeradbiomed.2007.12.028] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2007] [Revised: 12/16/2007] [Accepted: 12/17/2007] [Indexed: 11/20/2022]
Abstract
Predicting the susceptibility of lipid moieties to radical attack requires a determination of the depth of radical penetration into a lipid membrane. We thus synthesized three homologous series of lipophilic spin traps--DMPO analogs 2-alkanoyl-2-methyl-1-pyrroline N-oxides (11) and PBN derivatives 4-alkoxyphenyl N-tert-butylnitrones (18) and 4-alkoxyphenyl N-admantylnitrones (20). The intercalation depth of these spin traps within the liposomal bilayer was determined via the previously reported NMR technique, which correlates the chemical shift and the micropolarity (measured in ET(30) units) experienced by the pivotal nitronyl carbon. Hydroxyl and alpha-hydroxyalkyl radicals were generated in the extraliposomal aqueous phase and the lowest depth at which a radical could be spin trapped was determined. The ESR data indicate that these radicals can exit the aqueous phase, penetrate the lipid bilayer past the head groups (ET(30)=63 kcal/mol) and the glycerol ester (ET(30)=52 kcal/mol), and pass down to an ET(30) polarity of at least 44 kcal/mol. The latter depth presumably corresponds to the upper portion of the lipid slab. It is likely, if not probable, that having come this far they can abstract the allylic/diallylic hydrogens resident in the midslab at ET(30) values of >31 kcal/mol.
Collapse
Affiliation(s)
- Ayelet Gamliel
- The Ethel and David Resnick Chair in Active Oxygen Chemistry, Department of Chemistry, Bar Ilan University, Ramat Gan 52900, Israel
| | | | | |
Collapse
|
30
|
Adachi K, Chiba K. FTY720 Story. Its Discovery and the following Accelerated Development of Sphingosine 1-Phosphate Receptor Agonists as Immunomodulators Based on Reverse Pharmacology. PERSPECTIVES IN MEDICINAL CHEMISTRY 2007. [DOI: 10.1177/1177391x0700100002] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fingolimod (FTY720) is the first of a novel class: sphingosine 1-phosphate (S1P) receptor modulator and is currently in phase 3 clinical trials for multiple sclerosis (MS). FTY720 was first synthesized in 1992 by chemical modification of an immunosuppressive natural product, ISP-I (myriocin). ISP-I was isolated from the culture broth of Isaria sinclairii, a type of vegetative wasp that was an ‘eternal youth’ nostrum in traditional Chinese medicine. ISP-I is an amino acid having three successive asymmetric centers and some functionalities. We simplified the structure drastically to find a nonchiral symmetric 2-substitued-2-aminopropane-1,3-diol framework for an in vivo immunosuppressive activity (inhibition of rat skin allograft rejection test or prolonging effect on rat skin allograft survival) and finally discovered FTY720. During the course of the lead optimization process, we encountered an unexpected dramatic change of the mechanism of action with an in vivo output unchanged. Since it proved that FTY720 did not inhibit serine palmitoyltransferase that is the target enzyme of ISP-I, reverse pharmacological approaches have been preformed to elucidate that FTY720 is mainly phosphorylated by sphingosine kinease 2 in vivo and the phosphorylated drug acts as a potent agonist of four of the five G protein coupled receptors for S1P: S1P1, S1P3, S1P4 and S1P5. Evidence has accumulated that immunomodulation by FTY720-P is based on agonism at the S1P1 receptor. Medicinal chemistry targeting S1P1 receptor agonists is currently in progress. The FTY720 story provides a methodology where in vivo screens rather than in vitro screens play important roles in the lead optimization. Unlike recent drug discovery methodologies, such a strategy as adopted by the FTY720 program would more likely meet serendipity.
Collapse
Affiliation(s)
- Kunitomo Adachi
- Chemistry Laboratory, Pharmaceuticals Research Division, Mitsubishi Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, 227-0033, Japan
| | - Kenji Chiba
- Research Laboratory III (Immunology), Pharmaceuticals Research Division, Mitsubishi Pharma Corporation, 1000, Kamoshida-cho, Aoba-ku, Yokohama, 227-0033, Japan
| |
Collapse
|
31
|
Naor MM, Walker MD, Van Brocklyn JR, Tigyi G, Parrill AL. Sphingosine 1-phosphate pKa and binding constants: intramolecular and intermolecular influences. J Mol Graph Model 2007; 26:519-28. [PMID: 17467317 PMCID: PMC2040500 DOI: 10.1016/j.jmgm.2007.03.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2006] [Revised: 03/10/2007] [Accepted: 03/12/2007] [Indexed: 11/25/2022]
Abstract
The dissociation constant for an ionizable ligand binding to a receptor is dependent on its charge and therefore on its environmentally-influenced pKa value. The pKa values of sphingosine 1-phosphate (S1P) were studied computationally in the context of the wild type S1P1 receptor and the following mutants: E3.29Q, E3.29A, and K5.38A. Calculated pKa values indicate that S1P binds to S1P1 and its site mutants with a total charge of -1, including a +1 charge on the ammonium group and a -2 charge on the phosphate group. The dissociation constant of S1P binding to these receptors was studied as well. The models of wild type and mutant proteins originated from an active receptor model that was developed previously. We used ab initio RHF/6-31+G(d) to optimize our models in aqueous solution, where the solvation energy derivatives are represented by conductor-like polarizable continuum model (C-PCM) and integral equation formalism polarizable continuum model (IEF-PCM). Calculation of the dissociation constant for each mutant was determined by reference to the experimental dissociation constant of the wild type receptor. The computed dissociation constants of the E3.29Q and E3.29A mutants are three to five orders of magnitude higher than those for the wild type receptor and K5.38A mutant, indicating vital contacts between the S1P phosphate group and the carboxylate group of E3.29. Computational dissociation constants for K5.38A, E3.29A, and E3.29Q mutants were compared with experimentally determined binding and activation data. No measurable binding of S1P to the E3.29A and E3.29Q mutants was observed, supporting the critical contacts observed computationally. These results validate the quantitative accuracy of the model.
Collapse
Affiliation(s)
- Mor M. Naor
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| | - Michelle D. Walker
- Department of Physiology and University of Tennessee Cancer Institute, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - James R. Van Brocklyn
- Division of Neuropathology, Department of Pathology, The Ohio State University, Columbus, Ohio 43210
| | - Gabor Tigyi
- Department of Physiology and University of Tennessee Cancer Institute, University of Tennessee Health Science Center, Memphis, Tennessee 38163
| | - Abby L. Parrill
- Department of Chemistry and Computational Research on Materials Institute, The University of Memphis, Memphis, Tennessee 38152
| |
Collapse
|
32
|
Dudek SM, Camp SM, Chiang ET, Singleton PA, Usatyuk PV, Zhao Y, Natarajan V, Garcia JGN. Pulmonary endothelial cell barrier enhancement by FTY720 does not require the S1P1 receptor. Cell Signal 2007; 19:1754-64. [PMID: 17475445 PMCID: PMC2682440 DOI: 10.1016/j.cellsig.2007.03.011] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 03/22/2007] [Accepted: 03/22/2007] [Indexed: 10/23/2022]
Abstract
Novel therapeutic strategies are needed to reverse the loss of endothelial cell (EC) barrier integrity that occurs during inflammatory disease states such as acute lung injury. We previously demonstrated potent EC barrier augmentation in vivo and in vitro by the platelet-derived phospholipid, sphingosine 1-phosphate (S1P) via ligation of the S1P1 receptor. The S1P analogue, FTY720, similarly exerts barrier-protective vascular effects via presumed S1P1 receptor ligation. We examined the role of the S1P1 receptor in sphingolipid-mediated human lung EC barrier enhancement. Both S1P and FTY-induced sustained, dose-dependent barrier enhancement, reflected by increases in transendothelial electrical resistance (TER), which was abolished by pertussis toxin indicating Gi-coupled receptor activation. FTY-mediated increases in TER exhibited significantly delayed onset and intensity relative to the S1P response. Reduction of S1P1R expression (via siRNA) attenuated S1P-induced TER elevations whereas the TER response to FTY was unaffected. Both S1P and FTY rapidly (within 5 min) induced S1P1R accumulation in membrane lipid rafts, but only S1P stimulated S1P1R phosphorylation on threonine residues. Inhibition of PI3 kinase activity attenuated S1P-mediated TER increases but failed to alter FTY-induced TER elevation. Finally, S1P, but not FTY, induced significant myosin light chain phosphorylation and dramatic actin cytoskeletal rearrangement whereas reduced expression of the cytoskeletal effectors, Rac1 and cortactin (via siRNA), attenuated S1P-, but not FTY-induced TER elevations. These results mechanistically characterize pulmonary vascular barrier regulation by FTY720, suggesting a novel barrier-enhancing pathway for modulating vascular permeability.
Collapse
Affiliation(s)
- S M Dudek
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, 5841 South Maryland Ave. Chicago, IL 60637, United States.
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Chapter 16 Sphingosine 1-Phosphate Type 1 Receptor Modulators: Recent Advances and Therapeutic Potential. ANNUAL REPORTS IN MEDICINAL CHEMISTRY 2007. [DOI: 10.1016/s0065-7743(07)42016-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2023]
|
34
|
Colandrea VJ, Legiec IE, Huo P, Yan L, Hale JJ, Mills SG, Bergstrom J, Card D, Chebret G, Hajdu R, Keohane CA, Milligan JA, Rosenbach MJ, Shei GJ, Mandala SM. 2,5-Disubstituted pyrrolidine carboxylates as potent, orally active sphingosine-1-phosphate (S1P) receptor agonists. Bioorg Med Chem Lett 2006; 16:2905-8. [PMID: 16580205 DOI: 10.1016/j.bmcl.2006.03.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2006] [Revised: 02/28/2006] [Accepted: 03/01/2006] [Indexed: 10/24/2022]
Abstract
A series of 2,5-cis-disubstituted pyrrolidines were synthesized and evaluated as S1P receptor agonists. Compounds 15-21 were identified with good selectivity over S1P3 which lowered circulating lymphocytes after oral administration in mice.
Collapse
Affiliation(s)
- Vincent J Colandrea
- Department of Medical Chemistry, Merck Research Laboratories, Rahway, NJ 07065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Rosen H. Chemical approaches to the lysophospholipid receptors. Prostaglandins Other Lipid Mediat 2005; 77:179-84. [PMID: 16099402 DOI: 10.1016/j.prostaglandins.2004.09.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2004] [Accepted: 09/14/2004] [Indexed: 10/26/2022]
Abstract
Both ligand-based and GPCR privileged scaffold chemical tools have recently emerged to provide new insights into the function and physiology of the GPCR lysophospholipid receptors both in vitro and in vivo. Both rational, design-based approaches as well as hybrid approaches where high throughput screening has been coupled to an understanding of critical molecular interactions have been productive in advancing understanding of physiology and potential therapeutics in this field. It is now feasible to identify reasonably potent and selective small molecules that provide chemical proof-of-concept in vivo directly from high throughput screening. These developments, coupled with the availability of receptor knock-out mice, presage rapid progress in the field.
Collapse
Affiliation(s)
- Hugh Rosen
- Department of Immunology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
36
|
Kiuchi M, Adachi K, Tomatsu A, Chino M, Takeda S, Tanaka Y, Maeda Y, Sato N, Mitsutomi N, Sugahara K, Chiba K. Asymmetric synthesis and biological evaluation of the enantiomeric isomers of the immunosuppressive FTY720-phosphate. Bioorg Med Chem 2005; 13:425-32. [PMID: 15598563 DOI: 10.1016/j.bmc.2004.10.008] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2004] [Revised: 10/05/2004] [Accepted: 10/06/2004] [Indexed: 01/23/2023]
Abstract
A practical asymmetric synthesis of both enantiomers of the immunosuppressive FTY720-phosphate (2) was accomplished, and the enantiomers were pharmacologically evaluated. Several lipases showed considerable activity and enantioselectivity for O-acylation of N-acetyl FTY720 (3) or N-benzyloxycarbonyl FTY720 (7) in combination with vinyl acetate or benzyl vinyl carbonate as the acyl donors. The synthesis using the lipase-catalyzed acylation as the key step produced the enantiomerically pure (>99.5% ee) enantiomers of 2 in multigram quantities. (S)-Isomer of 2 had more potent binding affinities to S1P(1,3,4,5) and inhibitory activity on lymphocyte migration toward S1P than (R)-2, suggesting that (S)-isomer of 2 is responsible for the immunosuppressive activity after administration of 1. Severe bradycardia was observed in anesthetized rats when (S)-2 was administered intravenously, while (R)-2 had no clear effect on heart rate up to 0.3 mg/kg.
Collapse
Affiliation(s)
- Masatoshi Kiuchi
- Research Laboratory III (Immunology), Mitsubishi Pharma Corporation, Kamoshida-cho 1000, Aoba-ku, Yokohama 227-0033, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Davis MD, Clemens JJ, Macdonald TL, Lynch KR. Sphingosine 1-phosphate analogs as receptor antagonists. J Biol Chem 2004; 280:9833-41. [PMID: 15590668 DOI: 10.1074/jbc.m412356200] [Citation(s) in RCA: 228] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Sphingosine 1-phosphate (S1P) is a lysophospholipid mediator that evokes a variety of cell and tissue responses via a set of cell surface receptors. The recent development of S1P receptor agonists, led by the immunomodulatory pro-drug FTY720, has revealed that S1P signaling is an important regulator of lymphocyte trafficking. With the twin goals of understanding structure-activity relationships of S1P ligands and developing tool compounds to explore S1P biology, we synthesized and tested numerous S1P analogs. We report herein that a subset of our aryl amide-containing compounds are antagonists at the S1P(1) and S1P(3) receptors. The lead compound in series, VPC23019, was found in broken cell and whole cell assays to behave as a competitive antagonist at the S1P(1) and S1P(3) receptors. The structure-activity relationship of this series is steep; for example, a slight modification of the lead compound resulted in VPC25239, which was one log order more potent at the S1P(3) receptor. These new chemical entities will enable further understanding of S1P signaling and provide leads for further S1P receptor antagonist development.
Collapse
Affiliation(s)
- Michael D Davis
- Biochemistry and Molecular Genetics, Pharmacology, and Chemistry, University of Virginia, Charlottesville, Virginia 22908, USA
| | | | | | | |
Collapse
|
38
|
Goetzl EJ, Rosen H. Regulation of immunity by lysosphingolipids and their G protein–coupled receptors. J Clin Invest 2004. [DOI: 10.1172/jci200423704] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
39
|
Goetzl EJ, Rosen H. Regulation of immunity by lysosphingolipids and their G protein-coupled receptors. J Clin Invest 2004; 114:1531-7. [PMID: 15578083 PMCID: PMC529289 DOI: 10.1172/jci23704] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
T and B lymphocytes, as well as endothelial cells, express distinctive profiles of G protein-coupled receptors for sphingosine 1-phosphate, which is a major regulator of T cell development, B and T cell recirculation, tissue homing patterns, and chemotactic responses to chemokines. The capacity of drugs that act on type 1 sphingosine 1-phosphate receptors to suppress organ graft rejection in humans and autoimmunity in animal models without apparent impairment of host defenses against infections suggests that this system is a promising target for new forms of immunotherapy.
Collapse
Affiliation(s)
- Edward J Goetzl
- Department of Medicine, UCSF, San Francisco, California, USA.
| | | |
Collapse
|