Abstract
The sphingosine-1-phosphate-1 receptor (S1P1) and its endogenous ligand sphingosine-1-phosphate (S1P) cooperatively regulate lymphocyte trafficking from the lymphatic system. Herein, we disclose 4-methoxy-N-[2-(trifluoromethyl)biphenyl-4-ylcarbamoyl]nicotinamide (8), an uncommon example of a synthetic S1P1 agonist lacking a polar headgroup, which is shown to effect dramatic reduction of circulating lymphocytes (POC = −78%) in rat 24 h after a single oral dose (1 mg/kg). The excellent potency that 8 exhibits toward S1P1 (EC50 = 0.035 μM, 96% efficacy) and the >100-fold selectivity that it displays against receptor subtypes S1P2–5 suggest that it may serve as a valuable tool to understand the clinical relevance of selective S1P1 agonism.
Keywords: Sphingosine-1-phosphate-1 receptor agonist, peripheral lymphocyte count, immunosuppression, multiple sclerosis
Sphingosine-1-phosphate (1, S1P) and its affiliated G-protein-coupled receptors S1P1–5 are involved in modulating myriad physiological processes including cardiovascular function, pulmonary endothelium integrity, and cell motility (Figure 1).1,2 S1P (1) is derived biosynthetically from sphingosine (2) via phosphorylation mediated by either sphingosine kinase 1 or sphingosine kinase 2 (SphK1 or SphK2).3 Several years ago, S1P (1) and S1P1 were shown to regulate lymphocyte trafficking from the lymphatic system.4,5 At high concentrations of 1 or upon exposure to synthetic agonists of S1P1, binding to S1P1 located on the cell surface of lymphocytes in lymphatic tissues induces receptor internalization (RI) and prevents lymphocyte egress to the periphery and the central nervous system (CNS).6,7 Synthetic agonists of S1P1 are under intense investigation as potential therapies for medical conditions that may benefit from immunosuppression, such as multiple sclerosis (MS).8,9
Figure 1.
Endogenous and synthetic agonists of S1P1.
Fingolimod (3, FTY720)10,11 bears a dihydroxyamino polar headgroup akin to that of 2 and is likewise phosphorylated by SphK2 to give its bioactive congener 4 (FTY720-P),12,13 the first non-natural agonist of S1P1 to be reported (Figure 1). Notably, 4 displays agonist activity toward several S1P receptor subtypes,14 and until recently, its activation of S1P3 was believed to be exclusively responsible for its bradycardia side effects in humans and preclinical animal models.15,16 A short time ago, our laboratory disclosed 1-(3-fluoro-4-(5-(1-phenylcyclopropyl)thiazolo-[5,4-b]pyridin-2-yl)benzyl)azetidine-3-carboxylic acid (5, AMG369), a potent and selective dual agonist of S1P1/S1P5 bearing a polar headgroup that does not require bioactivation.17,18 Much more rare are S1P1 agonists that do not contain a polar headgroup,8,9 such as 5-(4-phenyl-5-(trifluoromethyl)thiophen-2-yl)-3-(3-(trifluoromethyl)phenyl)-1,2,4-oxadiazole (6, SEW2871),19,20 a compound that is exquisitely selective for S1P1 versus the other S1P receptor subtypes.21 The agonism of S1P1 by 4, 5, and 6 has been shown to effect reduction of circulating lymphocytes in vivo,11,17,22 and 3 (FTY720) was recently approved by the U.S. Food and Drug Administration for the treatment of relapsing forms of MS.23,24 As part of an effort to discover structurally novel agonists of S1P1, we set out to search for atypical chemotypes like 6 that lack a polar headgroup.25,26
During a high-throughput screening (HTS) campaign, we identified N-(3-chloro-4-(piperidin-1-yl)phenylcarbamothioyl)-2-methoxybenzamide (7, Scheme 1).27 The auspicious potency of 7 toward S1P1 (EC50 = 0.43 μM, 87% efficacy),28 coupled with its excellent selectivity against S1P3 (EC50 > 25 μM)29 and reasonable molecular and physicochemical properties (MW = 404, PSA = 54 Å2, cLogP = 4.5, and cLogD7.4 = 4.7),30 prompted us to investigate analogues that might exhibit improved physicochemical and biological profiles. The highlights of this lead optimization effort and its fruition in the discovery of a potent, selective, and orally bioavailable agonist of S1P1 lacking a polar headgroup, 4-methoxy-N-(2-(trifluoromethyl)biphenyl-4-ylcarbamoyl)nicotinamide (8) are the subject of this report (Scheme 1).
Scheme 1. Evolution of HTS Hit 7 to Carbamoylnicotinamide 8.
Carbamoylnicotinamide 8 was prepared as detailed in Scheme 2. Commercially available 2-(trifluoromethyl)biphenyl-4-amine (9)31 was treated with phosgene at elevated temperature to afford isocyanate 10,32 which was used immediately in the next step of the synthesis without purification. Deprotonation of known 4-methoxynicotinamide (11)33 with NaH followed by addition of 10 gave carbamoylnicotinamide 8 in 35% overall yield for the two-step sequence.34 This expedient route provided a multigram quantity of 8 that was sufficiently pure (>99%, HPLC) for an in vivo toxicology study (vide infra).
Scheme 2. Synthesis of Carbamoylnicotinamide 8.
Table 1 summarizes the lead optimization endeavor through which carbamothioylbenzamide 7 evolved to carbamoylnicotinamide 8.35 Although initially wary of the potential chemical lability of the carbamothioylbenzamide moiety of 7,36 we speculated that its intramolecular hydrogen-bonding network37 might help impart hydrolytic stability and conformational rigidity to it and its carbamoylbenzamide analogues.38−40 However, the potential toxicity of the carbamothioylbenzamide moiety or its possible metabolites41 prompted us to exchange the sulfur atom with oxygen to give carbamoylbenzamide 12. Whereas 12 exhibits reasonable potency toward S1P1 (EC50 = 0.23 μM, 120% efficacy), its cell permeability is diminished (Papp < 1.0 × 10–6 cm/s). Postulating that the chloro and piperidinyl substituents of 12 may bind to the same region of S1P1 as do the trifluoromethyl and phenyl groups on the thiophene ring of 6,25 we replaced the Cl atom in 12 with a CF3 group to afford 13. Although the improved potency of 13 was encouraging (EC50 = 0.013 μM, 120% efficacy), we desired to improve its stability against rat liver microsomes (CLint = 26 μL/min/mg). Suspecting that the piperidine ring of 13 may be the primary site of its metabolism,42 we replaced this ring with a phenyl group to provide 14. Gratifyingly, carbamoylbenzamide 14 displays enhanced microsomal stability (CLint = 15 μL/min/mg) and exhibits good potency toward S1P1 (EC50 = 0.068 μM, 110% efficacy). In an attempt improve the potency of 14, we replaced the CF3 substituent in 14 with an isopropyl group to give 15, which does indeed exhibit potent agonism of S1P1 (EC50 = 0.0054 μM, 120% efficacy). We also sought to improve the solubility and cell permeability of 14 by inserting a polar N atom into the scaffold, exemplified by 2-pyridinyl derivative 16, which shows the desired improvements (Sol. = 8.5–28 μg/mL; Papp = 7.4 × 10–6 cm/s), in addition to excellent potency on S1P1 (EC50 = 0.013 μM, 110% efficacy). Interestingly, 2-thiazolyl congener 17 displays greater cell permeability as compared to 16 (Papp = 12 × 10–6 cm/s) but has lower solubility and microsomal stability (Sol. = < 1.0–12 μg/mL; CLint = 33 μL/min/mg). To examine the effect of replacing the left-hand heterocyclic and (hetero)aromatic rings present in 7 and 12–17 with acyclic groups, isopropyl analogue 18 was prepared. Analogue 18 exhibits both excellent potency toward S1P1 (EC50 = 0.014 μM, 110% efficacy) and microsomal stability (CLint < 14 μL/min/mg). The 2,2,2-trifluoroethoxy compound 19 also shows potent activity toward S1P1 (EC50 = 0.0068 μM, 83% efficacy) but has lower cell permeability and microsomal stability (Papp < 1.0 × 10–6 cm/s; CLint = 25 μL/min/mg). Finally, inspired by 2-pyridinyl analogue 16, we desired to probe the effect of placing a polar N atom into the right-hand ring of carbamoylbenzamide 14. Capitalizing on the ready availability of 4-methoxynicotinamide (11),33 carbamoylnicotinamide 8 was prepared (Scheme 2). Indeed, 8 displays good potency toward S1P1 (EC50 = 0.035 μM, 96% efficacy), excellent stability against both human and rat liver microsomes (CLint < 14 μL/min/mg), reasonable cell permeability (Papp = 2.3 × 10–6 cm/s), and moderate solubility (Sol. = 6.6–17 μg/mL).
Table 1. S1P1 Structure–Activity Relationship, Physicochemical and Microsomal Stability Profiles, and PLC Activitya.

| X | Y | R1 | R2 | hS1P1, (μM)b EC50 (% eff.) | sol. (μg/mL)c HCl, PBS, SIF | Papp (ER)d (× 10–6 cm/s) | CLint, RLMe (μL/min/mg) | PLC, POCf (Cplasma, ng/mL) | |
|---|---|---|---|---|---|---|---|---|---|
| 7 | S | CH | Cl | N-piperidinyl | 0.43 (87) | <1.0, < 1.0, 27 | 1.8 (1.0) | 29 | NDg |
| 12 | O | CH | Cl | N-piperidinyl | 0.23 (120) | 3.6, < 1.0, 8.5 | <1.0 (NR)h | 36 | NDg |
| 13 | O | CH | CF3 | N-piperidinyl | 0.013 (120) | 3.0, < 1.0, 4.1 | <1.0 (NR)h | 26 | NDg |
| 14 | O | CH | CF3 | phenyl | 0.068 (110) | <1.0, < 1.0, 23 | <1.0 (NR)h | 15 | –20 (4.9)i,j |
| 15 | O | CH | i-Pr | phenyl | 0.0054 (120) | <1.0, < 1.0, 24 | 1.2 (1.0) | <14 | +3.0 (7.0)i |
| 16 | O | CH | CF3 | 2-pyridinyl | 0.013 (110) | 28, 19, 8.5 | 7.4 (1.1) | 15 | –14 (13)i |
| 17 | O | CH | CF3 | 2-thiazolyl | 0.038 (130) | 1.1, < 1.0, 12 | 12 (1.4) | 33 | –18 (1.0)i |
| 18 | O | CH | CF3 | isopropyl | 0.014 (110) | <1.0, < 1.0, 6.9 | 2.1 (1.0) | <14 | –16 (BQL)i,k |
| 19 | O | CH | CF3 | CF3CH2O | 0.0068 (83) | <1.0, < 1.0, 2.2 | <1.0 (NR)h | 25 | +4.0 (1.0)i |
| 8 | O | N | CF3 | phenyl | 0.035 (96) | 17, 6.6, 16 | 2.3 (1.0) | <14 | –67 (64)l |
See the Supporting Information for experimental details.
Data represent an average of at least two determinations; see ref (28).
Solubility in 0.010 M aqueous hydrogen chloride (HCl), phosphate-buffered saline (PBS) at pH 7.4, or simulated intestinal fluid (SIF).
Apparent permeability (Papp) through porcine proximal tubule cells (LLC-PK1 cell line) and efflux ratio (ER).
Estimated intrinsic clearance (CLint) determined by incubation of test compound with rat liver microsomes (RLM) for 30 min and measurement of % turnover.
Percent-of-control (POC) reduction vs vehicle in PLC 24 h after a single oral dose (1 mg/kg; vehicle = 20% captisol, 1% HPMC, and 1% pluronic F68, pH 2.1 with MSA) administered to female Lewis rats (N = 5/group) and total compound concentration in plasma (Cplasma) at the 24 h time point.
ND, not determined.
NR, not reportable.
The measured POC reduction in PLC is not statistically significant (P > 0.05 vs vehicle by the ANOVA/Dunnett's multiple comparison test).
See ref (43).
BQL, below the quantifiable limit.
The measured POC reduction in PLC is statistically significant (P < 0.05 vs vehicle by the ANOVA/Dunnett's multiple comparison test).
We desired next to examine the effect of the S1P1 agonists in Table 1 on circulating lymphocyte levels in vivo. Female Lewis rats were administered a single oral dose of each compound (1 mg/kg); after 24 h, lymphocyte counts in blood and compound concentrations in plasma were measured. The 24 h time point was selected for our studies, as it was found to mitigate the effects of dosing on lymphocyte counts.17 Notably, carbamoylnicotinamide 8 reaches >5-fold higher concentration in plasma than the other compounds tested in Table 1 (64 ng/mL) and is also the only one that effects statistically significant (P < 0.05 vs vehicle by the ANOVA/Dunnett's multiple comparison test) reduction in circulating lymphocytes (POC = −67%). Given their comparable in vitro profiles, it is unclear why compounds 14–16 and 18 do not achieve higher plasma levels and realize statistically significant reduction of peripheral lymphocyte counts (PLCs).43
Because of its robust activity in our preliminary in vivo study, carbamoylnicotinamide 8 was selected for further examination. As mentioned previously, compound 8 exhibits potent activation of S1P1 internalization (EC50 = 0.035 μM, 96% efficacy; Table 1);28 gratifyingly, it does not display appreciable agonism of receptor subtypes S1P2–4 (EC50 > 25 μM)44 and shows only weak activation of S1P5 (EC50 = 4.3 μM, 58% efficacy; Table 2).44 As the agonism of both S1P1 and S1P5 in oligodendrocytes by 4 has been suggested to contribute to its efficacy in the treatment of MS,45 S1P1-selective agonists such as 8 may serve as valuable tool compounds to examine the clinical relevance of S1P5 agonism.
Table 2. S1P Receptor Subtype Selectivity of 8a.
| EC50 (μM) | |||
|---|---|---|---|
| hS1P2 | hS1P3 | hS1P4 | hS1P5 (% eff.) |
| >25 | >25 | >50 | 4.3 (58) |
See the Supporting Information for experimental details (data represent an average of at least two determinations; see ref (44)).
A pharmacokinetic profile of carbamoylnicotinamide 8 was undertaken in preparation for more detailed studies in vivo (Table 3). In male Sprague–Dawley rats (N = 3) given a single intravenous dose (1 mg/kg), 8 displays low clearance (CL = 0.25 L/h/kg), moderate volume of distribution (Vdss = 6.3 L/kg), and a long half-life (T1/2 = 19 h). Although the plasma protein binding of 8 was found to be moderately high (PPB = 95%), its excellent bioavailability (% F = 140) and dose-proportional plasma exposure over a wide range of doses (AUC0→48 h, 1 mg/kg iv =3400 ng h/mL; AUC0→48 h, 3 mg/kg po =13000 ng h/mL; AUC0→48 h, 100 mg/kg po = 350000 ng h/mL) enabled a subsequent in vivo toxicology study (vide infra).
Table 3. Pharmacokinetic Profile of Carbamoylnicotinamide 8a.
| 1 mg/kg iv |
|||||||
|---|---|---|---|---|---|---|---|
| PPBb (%) rat | CLc (L/h/kg) | Vdssc (L/kg) | T1/2c (h) | AUC0→48 hc (ng h/mL) | AUC0→48 hd (ng h/mL) 3 mg/kg po | Fe (%) | AUC0→48 hf (ng h/mL) 100 mg/kg po |
| 95 | 0.25 | 6.3 | 19 | 3400 | 13000 | 140 | 350000 |
In vivo experiments were conducted using male Sprague–Dawley rats (N = 3/group).
Percent rat plasma protein binding (PPB) of 8 measured in vitro following separation by ultracentrifugation.
Clearance (CL), volume of distribution (Vdss), half-life (T1/2), and area under the plasma concentration–time curve (AUC0→48 h) determined following a single intravenous dose (1 mg/kg; vehicle = DMSO).
AUC0→48 h determined following a single oral dose (3 mg/kg; vehicle = 20% HPBCD, 1% HPMC, and 1% pluronic F68, pH 2.1, with MSA).
Percent bioavailability (F) calculated using AUC0→∞ values determined from the 1 (iv) and 3 mg/kg (po) doses.
AUC0→48 h determined following a single oral dose (100 mg/kg; vehicle = 20% HPBCD, 1% HPMC, and 1% pluronic F68, pH 2.1, with MSA).
We next sought to evaluate the pharmacodynamic effects of 8 in more detail (Figure 2). Following a single oral dose in female Lewis rats (0.30, 1.0, or 3.0 mg/kg), 8 exhibits dose-proportional plasma exposure (17, 63, and 160 ng/mL, respectively) and concomitant dose-dependent reduction in circulating lymphocytes at the 24 h time point. Statistically significant lymphocyte depletion is realized for the 1.0 and 3.0 mg/kg doses (78 and 81% reduction in PLCs; P < 0.001 and P < 0.01 vs vehicle by the ANOVA/Dunnett's multiple comparison test, respectively). This dose-dependent decrease in circulating lymphocytes reaching a plateau at 81% maximal reduction is consistent with the S1P1 agonist activity of 8.19,22
Figure 2.
Female Lewis rats (vehicle, 0.30, and 1.0 mg/kg dose groups: N = 5/group; 3.0 mg/kg dose group: N = 3; vehicle = 20% HPBCD, 1% HPMC, and 1% pluronic F68, pH 2.1, with MSA) administered a single oral dose of carbamoylnicotinamide 8 (0.30, 1.0, or 3.0 mg/kg) showed dose-proportional plasma exposure (black circles represent average plasma concentration ± SE) and dose-dependent reduction in circulating lymphocytes (gray bars represent average blood lymphocyte counts ± SE) at the 24 h time point (statistical significance: ***P < 0.001 and **P < 0.01 vs vehicle by the ANOVA/Dunnett's multiple comparison test, respectively); see the Supporting Information for experimental details.
The potential safety of carbamoylnicotinamide 8 was initially examined in vitro. Encouragingly, 8 displays no appreciable activity against either hERG (IC50 > 10 μM) or hCYPs 3A4 and 2D6 (IC50 > 30 μM) and exhibits negligible induction of hPXR (POC = +3.7%, 10 μM). An in vivo toxicology study of 8 was subsequently conducted. In male Sprague–Dawley rats (N = 3/group) dosed orally at 50 or 200 mg/kg (qd) for 4 days, no mortality, adverse clinical signs, or changes in body weight were observed. However, increases in total bilirubin (both doses) and cholesterol (high dose only) suggest altered bile uptake, processing, or excretion.
In conclusion, carbamoylnicotinamide 8 is a rare example of an S1P1 agonist lacking a polar headgroup. Despite its modest solubility and cell permeability, 8 is orally bioavailable and induces dose-dependent reduction in lymphocyte count in rat 24 h after a single oral dose. The excellent potency that 8 exhibits toward S1P1 and the remarkable selectivity that it displays against receptor subtypes S1P2–5 suggest that it may serve as a valuable tool to examine the proposed clinical relevance of S1P5 agonism for treating MS and the cardiovascular safety issues associated with S1P3 agonism.
Acknowledgments
We thank Laura Scott (HTS and Molecular Pharmacology, Amgen) for executing the HTS campaign that identified carbamothioylbenzamide 7, Kevin Turney (Analytical Research and Development, Amgen) and Christopher Wilde (Molecular Structure, Amgen) for confirmation of the structure assignment of carbamoylnicotinamide 8 using HRMS and NMR methods, respectively, and Ronya Shatila and Elizabeth Tominey (Pharmacokinetics and Drug Metabolism, Amgen) for in vivo pharmacokinetic studies. All live animal studies were conducted in an AAALAC-accredited facility and husbandry procedures met all of the recommendations of the Guide for the Care and Use of Laboratory Animals. All work using research animals was conducted under Institutional Animal Care and Use Committee (IACUC) approved protocols.
Glossary
Abbreviations
- EC50
molar concentration of compound that produces half maximal response
- RI
receptor internalization
- PSA
polar surface area
- cLogP
calculated log P (logarithm of the octanol/water partition coefficient)
- cLogD7.4
calculated log D (logarithm of the octanol/water distribution coefficient at pH 7.4)
- Papp
apparent permeability
- CLint
intrinsic clearance
- PLC
peripheral lymphocyte count
- HPMC
hydroxypropyl methylcellulose
- MSA
methanesulfonic acid
- Cplasma
total compound concentration in plasma
- HPBCD
hyroxypropyl β-cyclodextrin
- CL
clearance
- Vdss
volume of distribution at steady state
- T1/2
half-life of compound in plasma
- AUC
area under the plasma concentration–time curve
- F
bioavailability
- hERG
human ether-a-go-go related gene product (Kv11.1 potassium ion channel)
- hCYP 3A4
human cytochrome P450 subtype 3A4
- hCYP 2D6
human cytochrome P450 subtype 2D6
- IC50
molar concentration of compound that produces half maximal inhibition
- hPXR
human pregnane X receptor
- qd
quaque die (once per day dosing)
Supporting Information Available
Experimental procedures and characterization data for compounds 8 and 12–19, as well as experimental details and statistical analysis for key biological assays. This material is available free of charge via the Internet at http://pubs.acs.org.
Supplementary Material
References
- For a recent review, see Rosen H.; Gonzalez-Cabrera P. J.; Sanna M. G.; Brown S. Sphingosine-1-phosphate receptor signaling. Annu. Rev. Biochem. 2009, 78, 743–768. [DOI] [PubMed] [Google Scholar]
- For a review, see Takabe K.; Paugh S. W.; Milstien S.; Spiegel S. Inside-out signaling of sphingosine-1-phosphate: Therapeutic targets. Pharmacol. Rev. 2008, 60, 181–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- For a review, see Kihara A.; Mitsutake S.; Mizutani Y.; Igarashi Y. Metabolism and biological functions of two phosphorylated sphingolipids, sphingosine 1-phosphate and ceramide 1-phosphate. Prog. Lipid Res. 2007, 46, 126–144. [DOI] [PubMed] [Google Scholar]
- For a review, see Cyster J. G. Chemokines, sphingosine-1-phosphate, and cell migration in secondary lymphoid organs. Annu. Rev. Immunol. 2005, 23, 127–159. [DOI] [PubMed] [Google Scholar]
- S1P (1) hS1P1–5 EC50, μM (% efficacy): hS1P1 = 0.018 (91%), hS1P2 = 0.022 (91%), hS1P3 = 0.0094 (79%), hS1P4 = 0.16 (57%), and hS1P5 = 0.012 (63%); see the Supporting Information for experimental details.
- For a review, see Rivera J.; Proia R. L.; Olivera A. The alliance of sphingosine-1-phosphate and its receptors in immunity. Nat. Rev. Immunol. 2008, 8, 753–763. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Another possible mechanism involves S1P1-mediated lymphatic endothelium barrier modification; for a review, see Marsolais D.; Rosen H. Chemical modulators of sphingosine-1-phosphate receptors as barrier-oriented therapeutic molecules. Nat. Rev. Drug Discovery 2009, 8, 297–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- For a recent review, see Buzard D. J.; Thatte J.; Lerner M.; Edwards J.; Jones R. M. Recent progress in the development of selective S1P1 receptor agonists for the treatment of inflammatory and autoimmune disorders. Expert Opin. Ther. Patents 2008, 18, 1141–1159. [Google Scholar]
- For a review, see Cooke N.; Zécri F. Sphingosine 1-phosphate type 1 receptor modulators: Recent advances and therapeutic potential. Annu. Rep. Med. Chem. 2007, 42, 245–263. [Google Scholar]
- Mandala S.; Hajdu R.; Bergstrom J.; Quackenbush E.; Xie J.; Milligan J.; Thornton R.; Shei G.-J.; Card D.; Keohane C.; Rosenbach M.; Hale J.; Lynch C. L.; Rupprecht K.; Parsons W.; Rosen H. Alteration of lymphocyte trafficking by sphingosine-1-phosphate receptor agonists. Science 2002, 296, 346–349. [DOI] [PubMed] [Google Scholar]
- Brinkmann V.; Davis M. D.; Heise C. E.; Albert R.; Cottens S.; Hof R.; Bruns C.; Prieschl E.; Baumruker T.; Hiestand P.; Foster C. A.; Zollinger M.; Lynch K. R. The immune modulator FTY720 targets sphingosine 1-phosphate receptors. J. Biol. Chem. 2002, 277, 21453–21457. [DOI] [PubMed] [Google Scholar]
- Kharel Y.; Lee S.; Snyder A. H.; Sheasley-O’Neill S. L.; Morris M. A.; Setiady Y.; Zhu R.; Zigler M. A.; Burcin T. L.; Ley K.; Tung K. S. K.; Engelhard V. H.; Macdonald T. L.; Pearson-White S.; Lynch K. R. Sphingosine kinase 2 is required for modulation of lymphocyte traffic by FTY720. J. Biol. Chem. 2005, 280, 36865–36872. [DOI] [PubMed] [Google Scholar]
- Albert R.; Hinterding K.; Brinkmann V.; Guerini D.; Müller-Hartwieg C.; Knecht H.; Simeon C.; Streiff M.; Wagner T.; Welzenbach K.; Zécri F.; Zollinger M.; Cooke N.; Francotte E. Novel immunomodulator FTY720 is phosphorylated in rats and humans to form a single stereoisomer. Identification, chemical proof, and biological characterization of the biologically active species and its enantiomer. J. Med. Chem. 2005, 48, 5373–5377. [DOI] [PubMed] [Google Scholar]
- rac-FTY720-P (rac-4) hS1P1–5 EC50, μM (% efficacy): hS1P1 = 0.0020 (115%), hS1P2 > 1.0, hS1P3 = 0.027 (35%), hS1P4 = 0.17 (25%), and hS1P5 = 0.022 (36%); see the Supporting Information for experimental details.
- Schmouder R.; Serra D.; Wang Y.; Kovarik J. M.; DiMarco J.; Hunt T. L.; Bastien M.-C. FTY720: Placebo-controlled study of the effect on cardiac rate and rhythm in healthy subjects. J. Clin. Pharmacol. 2006, 46, 895–904 and references cited therein.. [DOI] [PubMed] [Google Scholar]
- Hamada M.; Nakamura M.; Kiuchi M.; Marukawa K.; Tomatsu A.; Shimano K.; Sato N.; Sugahara K.; Asayama M.; Takagi K.; Adachi K. Removal of sphingosine 1-phosphate receptor-3 (S1P3) agonism is essential, but inadequate to obtain immunomodulating 2-aminopropane-1,3-diol S1P1 agonists with reduced effect on heart rate. J. Med. Chem. 2010, 53, 3154–3168. [DOI] [PubMed] [Google Scholar]
- Cee V. J.; Frohn M.; Lanman B. A.; Golden J.; Muller K.; Neira S.; Pickrell A.; Arnett H.; Buys J.; Gore A.; Fiorino M.; Horner M.; Itano A.; Lee M. R.; McElvain M.; Middleton S.; Schrag M.; Rivenzon-Segal D.; Vargas H. M.; Xu H.; Xu Y.; Zhang X.; Siu J.; Wong M.; Bürli R. W. Discovery of AMG 369, a thiazolo[5,4-b]pyridine agonist of S1P1 and S1P5. ACS Med. Chem. Lett. 2011, 2, 107–112 and references cited therein.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- AMG369 (5) hS1P1–5 EC50, μM (% efficacy): hS1P1 = 0.0020 (98%), hS1P2 > 2.5, hS1P3 = 0.89 (26%), hS1P4 > 2.5, and hS1P5 = 0.029 (50%); see the Supporting Information for experimental details.
- Hale J. J.; Lynch C. L.; Neway W.; Mills S. G.; Hajdu R.; Keohane C. A.; Rosenbach M. J.; Milligan J. A.; Shei G.-J.; Parent S. A.; Chrebet G.; Bergstrom J.; Card D.; Ferrer M.; Hodder P.; Strulovici B.; Rosen H.; Mandala S. A rational utilization of high-throughput screening affords selective, orally bioavailable 1-benzyl-3-carboxyazetidine sphingosine-1-phosphate-1 receptor agonists. J. Med. Chem. 2004, 47, 6662–6665. [DOI] [PubMed] [Google Scholar]
- Jo E.; Sanna M. G.; Gonzalez-Cabrera P. J.; Thangada S.; Tigyi G.; Osborne D. A.; Hla T.; Parrill A. L.; Rosen H. S1P1-selective in vivo-active agonists from high-throughput screening: Off-the-shelf chemical probes of receptor interactions, signaling, and fate. Chem. Biol. 2005, 12, 703–715. [DOI] [PubMed] [Google Scholar]
- SEW2871 (6) hS1P1–5 EC50, μM (% efficacy): hS1P1 = 0.27 (86%) and hS1P3 > 25 (see the Supporting Information for experimental details); hS1P2–5 EC50 > 10 μM (see ref (22)).
- Sanna M. G.; Liao J.; Jo E.; Alfonso C.; Ahn M.-Y.; Peterson M. S.; Webb B.; Lefebvre S.; Chun J.; Gray N.; Rosen H. Sphingosine 1-phosphate (S1P) receptor subtypes S1P1 and S1P3, respectively, regulate lymphocyte recirculation and heart rate. J. Biol. Chem. 2004, 279, 13839–13848. [DOI] [PubMed] [Google Scholar]; S1P1 agonists have been shown to effect ∼70% maximal reduction in circulating lymphocytes; see ref (19).
- Kappos L.; Radue E.-W.; O'Connor P.; Polman C.; Hohlfeld R.; Calabresi P.; Selmaj K.; Agoropoulou C.; Leyk M.; Zhang-Auberson L.; Burtin P. A placebo-controlled trial of oral fingolimod in relapsing multiple sclerosis. N. Engl. J. Med. 2010, 362, 387–401. [DOI] [PubMed] [Google Scholar]
- Cohen J. A.; Barkhof F.; Comi G.; Hartung H.-P.; Khatri B. O.; Montalban X.; Pelletier J.; Capra R.; Gallo P.; Izquierdo G.; Tiel-Wilck K.; de Vera A.; Jin J.; Stites T.; Wu S.; Aradhye S.; Kappos L. Oral fingolimod or intramuscular interferon for relapsing multiple sclerosis. N. Engl. J. Med. 2010, 362, 402–415. [DOI] [PubMed] [Google Scholar]
- Fujiwara Y.; Osborne D. A.; Walker M. D.; Wang D.-a.; Bautista D. A.; Liliom K.; Van Brocklyn J. R.; Parrill A. L.; Tigyi G. Identification of the hydrophobic ligand binding pocket of the S1P1 receptor. J. Biol. Chem. 2007, 282, 2374–2385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gonzalez-Cabrera P. J.; Jo E.; Sanna M. G.; Brown S.; Leaf N.; Marsolais D.; Schaeffer M.-T.; Chapman J.; Cameron M.; Guerrero M.; Roberts E.; Rosen H. Full pharmacological efficacy of a novel S1P1 agonist that does not require S1P-like headgroup interactions. Mol. Pharmacol. 2008, 74, 1308–1318. [DOI] [PMC free article] [PubMed] [Google Scholar]
- By measurement of β-galactoside activity resulting from β-arrestin binding to hS1P1 in CHO-K1 cells.
- By measurement of RI of an hS1P1–eGFP fusion protein in U2OS cells (% efficacy is reported relative to known S1P1 agonist 1-(4-(6-benzylbenzofuran-2-yl)-3-fluorobenzyl)azetidine-3-carboxylic acid at a concentration of 1.0 μM; see ref (17)); see the Supporting Information for experimental details.
- By measurement of Ca2+ mobilization in CHO-K1 cells expressing hS1P3 and a chimeric Gq/i5 G-protein (% efficacy is reported relative to S1P at a concentration of 0.20 μM; > [highest concentration tested] is reported for compounds that do not achieve >10% of control activity); see the Supporting Information for experimental details.
- PSA and cLogP values were calculated using Daylight Chemical Information Systems software (web site: www.daylight.com); the cLogD7.4 value was calculated using Advanced Chemistry Development (ACD/Labs) software (web site: www.acdlabs.com).
- Pan S.; Mi Y.; Pally C.; Beerli C.; Chen A.; Guerini D.; Hinterding K.; Nuesslein-Hildesheim B.; Tuntland T.; Lefebvre S.; Liu Y.; Gao W.; Chu A.; Brinkmann V.; Bruns C.; Streiff M.; Cannet C.; Cooke N.; Gray N. A monoselective sphingosine-1-phosphate receptor-1 agonist prevents allograft rejection in a stringent rat heart transplantation model. Chem. Biol. 2006, 13, 1227–1234. [DOI] [PubMed] [Google Scholar]
- For a review, see Twitchett H. J. Chemistry of the production of organic isocyanates. Chem. Soc. Rev. 1974, 209–230. [Google Scholar]
- Ross W. C. J. The preparation of some 4-substituted nicotinic acids and nicotinamides. J. Chem. Soc. C 1966, 1816–1821. [DOI] [PubMed] [Google Scholar]
- The standard thermal conditions for coupling amides and isocyanates (toluene, 110 °C; Wiley P. F. The reaction of amides with isocyanates. J. Am. Chem. Soc. 1949, 71, 1310–1311) resulted in migration of the methyl group to the pyridyl nitrogen atom in 8; for the synthesis of compounds 12–19, see the Supporting Information.. [DOI] [PubMed] [Google Scholar]
- None of the compounds in Table 1 displays appreciable agonism of hS1P3 (EC50 > 25 μM); see ref (29).
- Douglass I. B.; Dains F. B. The preparation and hydrolysis of mono- and disubstituted benzoylthioureas. J. Am. Chem. Soc. 1934, 56, 1408–1409. [Google Scholar]
- For a review, see Aly A. A.; Ahmed E. K.; El-Mokadem K. M.; Hegazy M. E.-A. F. Update survey on aroyl substituted thioureas and their applications. J. Sulfur Chem. 2007, 28, 73–93. [Google Scholar]
- Gasteiger J.; Hondelmann U.; Röse P.; Witzenbichler W. Computer-assisted prediction of the degradation of chemicals: Hydrolysis of amides and benzoylphenylureas. J. Chem. Soc. Perkin Trans. 2 1995, 193–204. [Google Scholar]
- Sluiter C.; Kettenes-van den Bosch J. J.; Hop E.; van der Houwen O. A. G. J.; Underberg W. J. M.; Bult A. Degradation study of the investigational anticancer drug clanfenur. Int. J. Pham. 1999, 185, 227–235. [DOI] [PubMed] [Google Scholar]
- Quantum mechanics calculations using density functional theory at the B3LYP/6-31G* level (gas phase) indicate that 2-methoxy-N-(phenylcarbamoyl)benzamide prefers the bis-keto tautomeric form having the exchangeable protons attached to the nitrogen atoms; seeGaussian 03, Revision D.01; Gaussian, Inc.: Wallingford, CT, 2004. [Google Scholar]
- For an investigation of the metabolism and toxicity of arylthioureas and their metabolites, seeScheline R. R.; Smith R. L.; Williams R. T. The metabolism of arylthioureas—II. The metabolism of 14C- and 35S-labelled 1-phenyl-2-thiourea and its derivatives. J. Med. Pharm. Chem. 1961, 4, 109–135. [DOI] [PubMed] [Google Scholar]
- A preliminary metabolite identification study of a closely related scaffold using rat liver microsomes revealed the metabolic instability of the piperidine ring.
- In an initial study, carbamoylbenzamide 14 achieved a 9.6 ng/mL plasma concentration and effected a statistically significant (P < 0.05 vs vehicle by the ANOVA/Dunnett's multiple comparison test) 48% reduction in circulating lymphocytes (1.0 mg/kg, po, 24 h). A subsequent dose–response study (0.30, 1.0, and 3.0 mg/kg, po, 24 h) indicated that higher doses were necessary to achieve greater plasma levels and statistically significant lymphocyte count reduction; see the Supporting Information for experimental details.
- By measurement of Ca2+ mobilization in CHO-K1 cells expressing the target hS1P receptor and a chimeric Gq/i5 G-protein (for hS1P2, hS1P3, and hS1P5, % efficacy is reported relative to S1P at a concentration of 0.20 μM and > [highest concentration tested] is reported for compounds that do not achieve >10% of control activity; for hS1P4, % efficacy is reported relative to known S1P4 agonist (2R)-2-amino-3-(4-(2-benzylphenyl)-1H-indole-2-carboxamido)propanoic acid at a concentration of 0.40 μM and > [highest concentration tested] is reported for compounds that do not achieve >10% of control activity, seeAzzaoui K.; Bouhelal R.; Buehlmayer P.; Guerini D.; Koller M.. Indol-alanine derivatives as selective S1P4-agonists. World Intellectual Property Organization Patent Application WO 2005070886, 2005); see the Supporting Information for experimental details.
- For a review, see Brinkmann V. FTY720 (fingolimod) in multiple sclerosis: Therapeutic effects in the immune and the central nervous system. Br. J. Pharmacol. 2009, 158, 1173–1182. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.





