throbber
Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on May 1, 2017
`
`0022-3565/04/3092-758–768
`THE JOURNAL OF PHARMACOLOGY AND EXPERIMENTAL THERAPEUTICS
`U.S. Government work not protected by U.S. copyright
`JPET 309:758–768, 2004
`
`Vol. 309, No. 2
`62828/1143287
`Printed in U.S.A.
`
`Immune Cell Regulation and Cardiovascular Effects of
`Sphingosine 1-Phosphate Receptor Agonists in Rodents Are
`Mediated via Distinct Receptor Subtypes
`
`M. Forrest, S.-Y. Sun, R. Hajdu, J. Bergstrom, D. Card, G. Doherty,1 J. Hale,
`C. Keohane, C. Meyers, J. Milligan, S. Mills, N. Nomura, H. Rosen,2 M. Rosenbach,
`G.-J. Shei, I. I. Singer, M. Tian, S. West, V. White, J. Xie, R. L. Proia, and S. Mandala
`Departments of Immunology and Rheumatology, Pharmacology, and Medicinal Chemistry, Merck Research Laboratories,
`Rahway, New Jersey; and National Institutes of Health, Bethesda, Maryland (R.L.P.)
`Received November 11, 2003; accepted January 26, 2004
`
`ABSTRACT
`Sphingosine 1-phosphate (S1P) is a bioactive lysolipid with
`pleiotropic functions mediated through a family of G protein-
`coupled receptors, S1P1,2,3,4,5. Physiological effects of S1P
`receptor agonists include regulation of cardiovascular function
`and immunosuppression via redistribution of lymphocytes from
`blood to secondary lymphoid organs. The phosphorylated
`metabolite of the immunosuppressant agent FTY720 (2-
`amino-2-(2-[4-octylphenyl]ethyl)-1,3-propanediol) and other
`phosphonate analogs with differential receptor selectivity
`were investigated. No significant species differences in com-
`pound potency or rank order of activity on receptors cloned
`from human, murine, and rat sources were observed. All
`synthetic analogs were high-affinity agonists on S1P1, with
`IC50 values for ligand binding between 0.3 and 14 nM. The
`correlation between S1P1 receptor activation and the ED50
`for lymphocyte reduction was highly significant (p ⬍ 0.001)
`
`In contrast to S1P1-
`and lower for the other receptors.
`mediated effects on lymphocyte recirculation, three lines of
`evidence link S1P3 receptor activity with acute toxicity and
`cardiovascular regulation: compound potency on S1P3 cor-
`related with toxicity and bradycardia; the shift in potency of
`phosphorylated-FTY720 for inducing lymphopenia versus
`bradycardia and hypertension was consistent with affinity for
`S1P1 relative to S1P3; and toxicity, bradycardia, and hyper-
`⫺/⫺ mice. Blood pressure effects
`tension were absent in S1P3
`of agonists in anesthetized rats were complex, whereas hy-
`pertension was the predominant effect in conscious rats and
`mice. Immunolocalization of S1P3 in rodent heart revealed
`abundant expression on myocytes and perivascular smooth
`muscle cells consistent with regulation of bradycardia and
`hypertension, whereas S1P1 expression was restricted to the
`vascular endothelium.
`
`Sphingosine 1-phosphate (S1P) is a bioactive lipid derived
`from metabolism of sphingomyelin (Pyne and Pyne, 2000).
`S1P has been implicated in the regulation of many cellular
`functions including proliferation, apoptosis, survival, adhe-
`sion, differentiation, and migration (Hla et al., 2001). The
`diverse signaling has been attributed, in part, to the activa-
`tion of a family of G protein-coupled receptors called S1P or
`edg receptors that are differentially expressed and coupled to
`Gi/o, Gq, and G12/13 proteins (Chun et al., 2002).
`Few pharmacological tools with in vivo activity have
`
`1 Present address: Array BioPharma, Boulder, CO.
`2 Present address: The Scripps Research Institute, La Jolla, CA.
`Article, publication date, and citation information can be found at
`http://jpet.aspetjournals.org.
`DOI: 10.1124/jpet.103.062828.
`
`been described for the S1P receptors, but functions of the
`individual receptors are beginning to be elucidated. S1P1/
`edg1 has a widespread distribution and is highly abundant
`on endothelial cells where it works in concert with S1P3/
`edg3 to regulate cell migration, differentiation, and barrier
`function (Lee et al., 1999; Garcia et al., 2001). Although
`these receptors stimulate some pathways in common, they
`are not redundant. The S1P1 null embryos are defective in
`the migration of smooth muscle cell pericytes that are
`required to support vascular maturation and the embryos
`die at day 13.5 from hemorrhage (Liu et al., 2000). In
`contrast, the S1P3 null mouse is phenotypically normal
`(Ishii et al., 2001). S1P2/edg5 is a potent activator of the
`Rho pathway and inhibits cell migration, whereas S1P1
`and S1P3 stimulate chemotaxis of many cell types (Oka-
`
`ABBREVIATIONS: S1P, sphingosine 1-phosphate; IK.Ach, inwardly rectifying K⫹ currents; CHO, Chinese hamster ovary; MAP, mean arterial
`[35S]GTP␥S, guanosine 5⬘-O-(3-[35S]thio)triphosphate; FTY720, 2-amino-2-(2-[4-octylphenyl]
`pressure; DAPI, 4,6-diamidino-2-phenylindole;
`ethyl)-1,3-propanediol; PECAM, platelet-endothelial cell adhesion molecule.
`
`758
`
`Apotex v. Novartis
`IPR2017-00854
`NOVARTIS 2009
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on May 1, 2017
`
`Immune Cell Regulation and Cardiovascular Effects of S1P Receptor Agonists
`
`759
`
`moto et al., 2000; Graeler and Goetzl, 2002). Much less is
`known about the function of S1P4/edg6, which is restricted
`to hematopoietic and lymphoid tissues (Graeler et al.,
`1999), and S1P5/edg8, which is predominant in rodent
`brain (Im et al., 2000) but more broadly expressed in
`human tissues (Niedernberg et al., 2002).
`A novel physiological role for S1P in immune regulation
`has been discovered recently by elucidating the mechanism
`of FTY720 (Brinkmann et al., 2002; Mandala et al., 2002), an
`immunosuppressive agent with activity in many models of
`transplantation and immune-based disease (Dumont, 2000;
`Brinkmann et al., 2001). FTY720 depletes peripheral blood
`lymphocytes and sequesters them in secondary lymphoid
`organs (Chiba et al., 1998). We discovered that FTY720 is
`phosphorylated in vivo to become a high-affinity ligand
`(Compound A) for S1P1,3,4,5 but not S1P2 (Mandala et al.,
`2002). A non-hydrolyzable phosphonate analog (Compound
`B) with similar S1P receptor selectivity was also able to alter
`lymphocyte recirculation (Mandala et al., 2002). Close ana-
`logs of FTY720, such as the (S) enantiomer of 2-amino-4-(4-
`heptyloxyphenyl)-2-methylbutanol, that were not substrates
`for phosphorylation did not have immunosuppressive activ-
`ity, thus providing additional evidence that the phosphory-
`lated metabolite is the active species (Brinkmann et al., 2002;
`Mandala et al., 2002). The receptor(s) responsible for im-
`mune modulation has not been determined, although both
`S1P1 and S1P4 have been implicated based on their roles in
`regulating lymphocyte chemotaxis (Brinkmann et al., 2001;
`Dorsam et al., 2003).
`Clinical studies with FTY720 have identified dose-depen-
`dent transient asymptomatic bradycardia in stable renal
`transplant patients (Budde et al., 2002). Although there are
`no published reports on the effects of FTY720 on heart rate in
`rodents, S1P decreased heart rate in anesthetized rats (Sug-
`iyama et al., 2000a). Bradycardia is consistent with previous
`reports of S1P activation of muscarinic receptor-activated
`inwardly rectifying K⫹ currents (IK.Ach) and concomitant
`slowing of sino-atrial node pacemaker activity (Bunemann et
`al., 1995; Guo et al., 1999). In contrast to observations in rats,
`S1P administration to a canine isolated heart preparation
`evoked a sinus tachycardia (Sugiyama et al., 2000b). The
`effects of S1P receptor agonists on vascular tone and mean
`arterial pressure (MAP) are similarly complex. FTY720 has
`not been reported to have any effects on MAP in man. How-
`ever, FTY720 has been shown to either decrease or increase
`MAP in anesthetized rats (Tawadrous et al., 2002). On iso-
`lated vascular preparations, S1P induced relaxation at low
`concentrations (10–100 nM) (Dantas et al., 2003) and vaso-
`constrictor effects at higher concentrations (100 nM to 100
`␮M) (Bischoff et al., 2000; Tosaka et al., 2001). The complex
`cardiovascular effects of S1P receptor agonists are likely due
`to differential effects on S1P receptor subtypes. S1P2 contrac-
`tion of coronary smooth muscle cells was implicated using a
`selective antagonist (Ohmori et al., 2003) but antisense to
`S1P3, not S1P2, inhibited contraction of rat basilar arteries
`(Salomone et al., 2003). The availability of S1P subtype re-
`ceptor-selective agonists with in vivo activity that are de-
`scribed in this paper and S1P3 receptor knockout mice allows
`for a more detailed investigation of the cardiovascular and
`lymphocyte trafficking effects of S1P agonists.
`
`Materials and Methods
`Synthesis of S1P Receptor Agonists. The syntheses of Com-
`pounds A and B have been described previously (Mandala et al.,
`2002). Compound C was prepared in seven steps from 2-acetylamino-
`2-(2-(4-octylphenyl)ethyl)propane-1,3-dicarboxylic acid, diethyl ester
`(Durand et al., 2000). Compounds D to F were prepared by reductive
`amination of the appropriate aryl aldehyde with 3-aminopropylphos-
`phonic acid [Na(CN)BH3, MeOH, 50°C]. All compounds were char-
`acterized by 1H NMR, mass spectroscopy, and high-pressure liquid
`chromatography and were judged to be ⬎95% pure. Detailed proce-
`dures are provided in patents WO 03074008 and WO 03062252.
`Mouse Lymphocyte Reduction Assay. Mice (three per group)
`were dosed intravenously with 0.1 ml of test compound dissolved in
`vehicle [2% (w/v) hydroxypropyl-␤-cyclodextrin (Cerestar, Cedar
`Rapids, IA) and 0.12 M NaCl], and peripheral blood lymphocyte
`counts were assessed 3 h later. Mice were euthanized via CO2 inha-
`lation, the chest was opened, 0.5 ml of blood was withdrawn via
`direct cardiac puncture into EDTA, and hematology was evaluated
`using a clinical hematology AutoAnalyzer calibrated for performing
`murine differential counts (H2000; CARESIDE, Culver City, CA).
`Toxicity was observed upon administration of some of the test com-
`pounds. Severe signs included death, seizure, paralysis, or uncon-
`sciousness. Milder signs were also noted and included ataxia, la-
`bored breathing, ruffling, or reduced activity relative to normal. To
`assess lymphopenic activity with these compounds, upon noting
`symptoms in the first animal, the dosing solution was diluted in the
`same vehicle and administered to a second mouse for observation.
`The process was repeated until a dose was reached that produced
`only brief, mild symptoms. This was considered the maximum toler-
`ated dose. All procedures were performed in accordance with the
`highest standards for the humane handling, care, and treatment of
`research animals and were approved by the Merck Institutional
`Animal Care and Use Committee.
`Receptors and Cell Lines. CHO cells stably expressing human
`S1P1,2,3,4,5 were as previously described (Mandala et al., 2002). cDNA
`sequences encoding rodent S1P receptors were cloned from genomic
`DNA by polymerase chain reaction using the following primers for
`each respective receptor: 5⬘-GAACCCGGGTGTCCACTAGCATC-
`CCGG and 5⬘-CCCGAATTCTTAGGAAGAAGAATTGACGTTTCC
`(mouse S1P1), 5⬘-GAACCCGGGCGGCTTATACTCAGAGTACC and
`5⬘-GGCGAATTCTCAGACCACTGTGTTACCCTC (mouse S1P2), 5⬘-
`GAACCCGGGCAACCACGCATGCGCAGG and 5⬘-GTCGAATTCT-
`CACTTGCAGAGGACCCCG (mouse S1P3), 5⬘-GAACCCGGGAA-
`CATCAGTACCTGGTCCACGC and GCGGAATTCTAGGTGCTGC-
`GGACGCTGG (mouse S1P4), 5⬘-GAACCCGGGCTGCTGCGGCCGG
`and 5⬘-CGCGAATTCAGTCTGTAGCAGTAGGCACC (mouse S1P5),
`5⬘- GTAGGATCCGTGTCCTCCACCAGCATC and 5⬘-GGCCGAAT-
`TCTTAAGAAGAAGAATTGACGTTTC (rat S1P1), and 5⬘- GAAC-
`CCGGGCATCCACGCATGCGCAG and 5⬘-GCCGAATTCTCACTT-
`GCAGAGGACCCCATTCTG (rat S1P3). The polymerase chain
`reaction products were inserted in frame after a FLAG tag using
`vector pCMV-Tag2 (Stratagene, La Jolla, CA). Stable lines were
`established by transfecting plasmids into CHO cells using lipo-
`fectamine reagent, selecting for neomycin resistance, and screen-
`ing single cell cultures for increased [33P]S1P-specific binding.
`Membranes were prepared from positive clones and confirmed in
`[33P]S1P and [35S]GTP␥S binding assays.
`S1P Receptor Assays. Binding assays were conducted as previ-
`ously described (Mandala et al., 2002). In brief, [33P]S1P was soni-
`cated with fatty acid-free bovine serum albumin, added to test com-
`pounds diluted in dimethyl sulfoxide, and mixed with membranes in
`200 ␮l in 96-well plates with assay concentrations of 0.1 nM [33P]S1P
`(22,000 dpm), 0.5% bovine serum albumin, 50 mM HEPES-Na (pH
`7.5), 5 mM MgCl2, 1 mM CaCl2, and 0.3 to 0.7 ␮g of membrane
`protein. Binding was performed for 60 min at room temperature and
`terminated by collecting the membranes onto GF/B filter plates with
`a Packard Filtermate Universal Harvester. Filter bound radionu-
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on May 1, 2017
`
`760
`
`Forrest et al.
`
`clide was measured on a PerkinElmer 1450 MicroBeta. Specific
`binding was calculated by subtracting radioactivity that remained in
`the presence of 1000-fold excess of unlabeled S1P.
`To measure functional activation of the S1P receptors, [35S]GTP␥S
`binding was measured. Membranes (1–4 ␮g of protein) were incu-
`bated in 96-well plates with test compounds diluted in dimethyl
`sulfoxide in 100 ␮l of buffer containing 20 mM HEPES (pH 7.4), 100
`mM NaCl, 10 mM MgCl2, and 2 to 10 ␮M GDP, depending on the
`expressed receptor. The assay was initiated with the addition of 100
`␮l of [35S]GTP␥S (1200 Ci/mmol; PerkinElmer Life and Analytical
`Sciences, Boston, MA) for an assay concentration of 125 pM. After 60
`min of incubation at room temperature, membranes were harvested,
`onto GF/B filter plates and bound radionuclides were measured as
`described for ligand binding. S1P was subject to significant dephos-
`phorylation in the [35S]GTP␥S binding assay as measured with
`[33P]S1P or [3H]S1P. EC50 values are not reported for S1P. Degra-
`dation of S1P was less than 10% in the [33P]S1P binding assay, and
`[3H]-Compound A was not metabolized under either assay condition.
`Assessment of Cardiovascular Function. Cardiovascular
`function was assessed in anesthetized and conscious rats and in
`conscious mice. Anesthetized rats were used to evaluate the dose-
`dependent profile of cardiovascular responses to S1P receptor ago-
`nists of differing S1P receptor selectivity. Conscious rats were used
`to verify data from anesthetized animals and to allow a longer period
`of observation (4 h) that included a concurrent assessment of the
`induction of lymphopenia. The use of conscious mice allowed a com-
`parison of compound effects in wild-type animals and mice with
`genetic deletion of the S1P3 receptor.
`Anesthetized Rat Cardiovascular Assay. For the assessment
`of cardiovascular function in anesthetized rats, male Sprague-Daw-
`ley rats (300–350 g. b.wt.) with surgically implanted femoral artery
`and vein catheters were obtained from Charles River Laboratories
`(Raleigh, NC). Animals were anesthetized with Nembutal (55 mg/kg,
`i.p.), and a DTX pressure transducer (TNF-R; BD Biosciences, San
`Jose, CA) was attached to the arterial catheter and subsequently to
`a Gould ACQ-7700 data acquisition system using Po-Ne-Mah soft-
`ware (PNM-P3P) (Gould Instrument Systems, Valley View, OH).
`Heart rate was derived from the arterial pulse wave. Following an
`acclimation period, baseline values were determined (approximately
`20-min duration); subsequently, compounds were administered in-
`travenously (bolus injection of approximately 10 s). Cardiovascular
`data were recorded continuously and are reported as average values
`over 1-min intervals.
`Conscious Rat Cardiovascular Assay. For the assessment of
`cardiovascular function in conscious rats, male Sprague-Dawley rats
`(300–350 g. b.wt.) with surgically implanted femoral artery and vein
`catheters were obtained from Charles River Laboratories. The cath-
`eters were connected to a tether (CIH95) and swivel (375/20; Instech
`Laboratories, Inc., Plymouth Meeting, PA), allowing the animal to
`move freely around the cage. Animals were allowed a minimum
`2-day acclimation prior to further experimentation. On the day of
`study, a BD Biosciences DTX pressure transducer (TNF-R) was
`attached to the arterial catheter and subsequently to a Gould ACQ-
`7700 data acquisition system using Po-Ne-Mah software. Heart rate
`was derived from the arterial pulse wave. Heart rate and arterial
`pressure were measured for between 30 and 60 min to establish
`baseline values. Subsequently, compound or vehicle was adminis-
`tered as a continuous intravenous infusion of 25 ␮l/min for 4 h. Blood
`samples, for the evaluation of circulating leukocytes, were obtained
`from the arterial catheter 30 min prior to dosing and 1 and 4 h post
`initiation of the infusion. Cardiovascular data were recorded contin-
`uously and are reported as average values over 1-min intervals.
`Mouse Cardiovascular Assay. For the assessment of cardiovas-
`cular function in conscious mice, the S1P3 receptor was genetically
`deleted (R. L. Proia, manuscript submitted for publication) and bred
`⫺/⫺
`at Taconic Farms Inc. (Germantown, NY). Male (B6.129) S1P3
`⫹/⫹ mice (20–30 g b.wt.) were anesthetized with ketamine
`and S1P3
`(80–100 mg/kg, i.p.) and xylazine (10 mg/kg, i.p.), and catheters were
`
`placed in a carotid artery (PE-50, with tip modified) and Jugular vein
`(PE-10). The catheters were tunneled subcutaneously to the nape
`and exteriorized. The catheters were connected to a tether (CIH62)
`and swivel (375/25P; Instech Laboratories, Inc.), allowing the animal
`to move freely around the cage. Following surgery, animals were
`allowed an overnight recovery period prior to further experimenta-
`tion. On the day of study, a BD Biosciences DTX pressure transducer
`was attached to the arterial catheter and subsequently to a Gould
`ACQ-7700 data acquisition system using Po-Ne-Mah software. Heart
`rate was derived from the arterial pulse wave. Heart rate and arte-
`rial pressure were measured for between 30 and 60 min to establish
`baseline values. Subsequently, compound or vehicle was adminis-
`tered intravenously as a bolus of 10-s duration. Cardiovascular data
`were recorded continuously and are reported as average values over
`10-s intervals.
`Immunohistochemical Localization of S1P1 and S1P3 in Rat
`Heart. Peptides to the N terminus of mouse S1P3 (ATTHAQGHQPV-
`LGNDTLREHYDYVGKLAGRLRDPPEGGTL) and mouse S1P1 (VST-
`SIPEVKALRSSVSDYGNYDIIVRHYNYTGKLNIGAEKDHGIK) and
`the C terminus of S1P1 (EGDNPETIMSSGNVNSSS) were synthesized
`(SynPep Corp., Dublin, CA), conjugated to KLH, and used to immunize
`rabbits (Covance Research Products, Denver, PA). Specific IgG frac-
`tions were affinity purified using the immunizing peptides. The result-
`ing antisera were tested by Western blot across a panel of human,
`mouse, and rat S1P receptors. MS2031 to S1P3 was specific for the
`rodent S1P3 receptors. For the S1P1 antisera, MS2029 to the N termi-
`nus was specific for rodent S1P1, whereas the C-terminal antisera,
`MS1766, recognized human and rodent S1P1. Staining by Western and
`immunohistochemistry was blocked by incubation with the relevant but
`not the irrelevant peptide.
`For histochemical studies, blocks of atrium or ventricle from rats
`or mice treated with a lethal dose of sodium pentobarbital were
`rapidly dissected out and immediately placed in cryomolds (catalog
`no. 4557; TissueTek, Torrance, CA) filled with O.C.T. Compound
`(TissueTek catalog no. 4583), frozen in liquid nitrogen, and stored at
`⫺80°C. Frozen sections were cut (5-␮m thickness) on a Bright Model
`OTF cryotome (Hacker Instruments, Fairfield, NJ) and mounted on
`coated slides (catalog no. CFSACS; Instrumedics, Inc., Hackensack,
`NJ). To block nonspecific labeling, sections were treated with 5%
`donkey serum in PBS for 20 min, then with a clarified solution of 5%
`nonfat dry milk for 30 min, and finally with Fc blocker (Accurate
`Chemical, Westbury, NY) for 20 min. Sections were labeled for 1 h
`with affinity-purified primary antibodies or appropriate IgG controls
`(5 ␮g/ml): rabbit anti-mouse S1P1, rabbit anti-human S1P1, rabbit
`anti-mouse S1P3, and mab anti-rat PECAM (CD31 Pharmingen,
`catalog no. 555025; BD PharMingen, San Diego, CA). All nonimmune
`IgG controls were obtained from Jackson ImmunoResearch Labora-
`tories Inc. (West Grove, PA). Slides were washed and incubated with
`affinity-purified F(ab⬘)2 donkey anti-rabbit or rat Cy2 (green fluores-
`cence) or Cy3 (red fluorescence) conjugated secondary antibodies (5
`␮g/ml, 30 min) from Jackson ImmunoResearch Laboratories Inc. For
`double labeling studies, two primary antibodies raised in different
`species or corresponding species-specific fluorescent secondary anti-
`bodies were mixed together and incubated simultaneously on each
`slide. Nonimmune IgG controls also were run in this fashion. Spec-
`ificity also was demonstrated by pre-incubating each primary anti-
`body at staining concentrations with its relevant or irrelevant pep-
`tide at 5 ␮g/ml for 1 h and centrifugation for 1 h at 13,500g at 4°C
`(Beckman Microfuge 11; Beckman Coulter, Fullerton, CA). After
`staining, the slides were fixed for 30 min in 4% formaldehyde freshly
`generated from paraformaldehyde in phosphate buffer (pH 7.4; cat-
`alog no. 04042500; Fisher Scientific Co., Pittsburgh, PA). Coverslips
`were mounted on the slides with Vectashield plus DAPI nuclear
`stain (catalog no. H1200, Vector Laboratories, Burlingame, CA).
`Sections were photographed and analyzed with an Everest imaging
`system from Intelligent Imaging Innovations (Denver, CO) equipped
`with an Axioplan 2 microscope (Carl Zeiss, Go¨ttingen, Germany).
`This system allows the viewer to visualize two different fluoro-
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on May 1, 2017
`
`Immune Cell Regulation and Cardiovascular Effects of S1P Receptor Agonists
`
`761
`
`Fig. 1. Chemical structures of S1P and synthetic
`S1P receptor agonists.
`
`chrome conjugated secondary antibodies individually or in combina-
`tion on the same section in double labeling experiments.
`
`with an ED50 of 2.2 mg/kg. No toxicity was observed with
`Compounds E and F.
`To investigate S1P receptor selectivity of the analogs, they
`
`Results
`Lymphopenia, Toxicity, and in Vitro Activity of S1P
`Receptor Agonists. The phosphonate analog (Compound B)
`of the phosphate-ester metabolite of FTY720 (Compound A)
`was found previously to deplete peripheral blood lympho-
`cytes with a 15-fold right shift in potency relative to FTY720
`(Mandala et al., 2002). The shift in potency was consistent
`with its reduced affinity for S1P receptors. In an effort to
`identify more potent immunosuppressive compounds and ex-
`plore structure activity relationships, additional analogs
`were synthesized (Fig. 1). Using a 3-h murine assay, Com-
`pound C and several analogs in the secondary amine phos-
`phonate series were found to reduce circulating blood lym-
`phocytes (Fig. 2). Compound D was 2-fold less active than
`Compound A and was the most potent phosphonate analog
`tested. However, efficacy could only be assessed within a
`narrow concentration range due to toxicity. Doses higher
`than 0.05 mg/kg of Compound D induced symptoms of ataxia
`and paralysis, and 0.25 mg/kg was lethal. Compounds C and
`E were 5- to 10-fold less active than D in reducing peripheral
`blood lymphocytes, and compound F had the weakest activity
`
`Fig. 2. Dose response of S1P receptor agonists in peripheral blood lym-
`phocyte depletion. Compounds were administered as an intravenous bo-
`lus to mice (n ⫽ 3), and total blood lymphocytes were determined 3 h
`later.
`
`

`

`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on May 1, 2017
`
`762
`
`Forrest et al.
`
`were tested against human receptors expressed in CHO cells
`in competitive ligand binding assays using [33P]S1P as the
`ligand and functional assays of G protein coupling using
`[35S]GTP␥S binding (Table 1). As previously determined for
`Compounds A and B, none of the phosphonates had signifi-
`cant activity against S1P2. All compounds were agonists on
`the other four S1P receptors, and maximal efficacy in
`[35S]GTP␥S binding was similar to that observed with Com-
`pound A. Compound D was the most potent phosphonate on
`S1P1 with an IC50 of 0.9 nM that was only 2- to 3-fold less
`active than S1P or Compound A. Compound D was also the
`most active synthetic analog on S1P3, with an IC50 of 7.9 nM
`that was equivalent to Compound A but less potent relative
`to S1P. The 2-bromo and 5-methoxy substitutions to the
`phenyl ring found in Compounds E and F resulted in modest
`decreases in affinity for S1P1 and a substantial loss in activ-
`ity on S1P3, which was further reduced by shortening the
`alkyl chain length in Compound F. All of the phosphonates
`had similar activity on S1P4, making this receptor unlikely to
`account for the differential effects of the analogs on lym-
`phopenia or toxicity. Likewise, S1P5 did not appear to be a
`probable candidate for either in vivo effect given the equiva-
`lent potency of all three of the secondary amine compounds
`and the 20-fold reduced activity of Compound C on S1P5
`relative to Compounds D to F.
`Correlations between rodent in vivo pharmacological ef-
`fects and human in vitro receptor assays can be misleading
`unless there is a high degree of conservation in the ligand
`binding pocket of the receptors. To address this issue, we
`cloned mouse S1P1,3,4,5 and rat S1P1,3 receptors and estab-
`lished stable CHO lines and assays that were comparable to
`those used to assess the human receptors. The overall homol-
`ogy between rodent and human S1P receptors ranged from
`80% to 93%, with S1P1 being the most conserved protein.
`Ligand binding assays using the rodent receptors (Table 2)
`revealed that the natural ligand and the synthetic com-
`pounds maintained similar potency and the same rank order
`of activity, thus indicating that there were no significant
`differences between rodent and human structure activity
`relationships. As was deduced previously from the human
`receptors, the correlation between S1P1 receptor activation
`
`and the ED50 for lymphopenia was highly significant (p ⬍
`0.001) and much lower for the other receptors. All of the
`compounds had low or sub-nanomolar potency on S1P1, and
`the rank order of activity was Compound A ⬎ D ⬎ C ⬎ E ⬎ F.
`Intravenous administration of some of the compounds
`evoked toxicity with symptoms that ranged from transient
`ruffling and paralysis to lethality. Compound D was the least
`tolerable compound tested and the most potent synthetic
`analog on mouse S1P3 with an IC50 of 1.8 nM. The other two
`compounds in the secondary amine series (E and F) had
`considerably reduced potency at mouse S1P3 with IC50 val-
`ues of 0.6 and 6.6 ␮M, respectively, and did not induce any
`adverse symptoms in mice. Only mild, transient toxicity was
`observed with Compound C, which had intermediate activity
`on S1P3. Compound A induced transient signs of toxicity in
`mice but was not lethal up to 10 mg/kg, whereas S1P was
`highly toxic and was the most potent ligand for S1P3. To test
`whether toxicity was mediated via S1P3, as suggested by the
`correlation of in vitro receptor potency with the maximal
`tolerated dose (Table 2), the severely toxic compounds were
`⫺/⫺ mice, S1P and Com-
`studied in S1P3 null mice. In S1P3
`pound D did not induce any toxic symptoms at 25 and 5
`mg/kg, respectively, whereas they were rapidly lethal in their
`wild-type litter mate controls at doses of 5 and 0.25 mg/kg,
`⫺/⫺ mice were fully susceptible to agonist-
`respectively. S1P3
`mediated lymphocyte depletion; peripheral blood lymphocyte
`counts from Compound D treated mice were 1.6 ⫻ 106/ml
`compared with 5.0 ⫻ 106/ml for vehicle-treated mice (p ⬍ 0.01).
`Effects of S1P Receptor Agonists in Anesthetized
`Rats. Administration of either S1P (Fig. 3A) or the S1P1,3,4,5
`agonist, Compound A (Fig. 3B), to anesthetized rats evoked
`an immediate decrease in heart rate, reaching a nadir within
`1 to 2 min of compound administration and returning toward
`baseline values within 10 min. The effects on mean arterial
`pressure were more complex. Initially, a rapid hypotension
`was observed that reversed concomitantly with an increase
`in heart rate. In some instances, an overshoot to a transient
`hypertension was observed before returning to baseline. Sub-
`sequently, a gradually developing somewhat variable hyper-
`tension was seen that peaked approximately 10 min post-
`compound administration before resolving within 20 min.
`
`TABLE 1
`Activity of compounds on human S1P receptors
`Compounds were tested in 关33P兴S1P binding assays to determine IC50 values and 关35S兴GTP␥S binding assays to determine EC50 values using membranes prepared from CHO
`cells expressing human S1P receptors. Values are the mean of two to six measurements performed in duplicate. S1P is subject to extensive metabolism under standard
`关35S兴GTP␥S binding assay conditions, and EC50 values are not shown.
`
`S1P
`
`0.5
`ND
`
`0.3
`ND
`
`0.2
`ND
`
`55
`ND
`
`0.5
`ND
`
`A
`
`0.3
`0.3
`
`⬎1000
`⬎1000
`
`5.0
`3.0
`
`5.9
`4.3
`
`0.6
`1.0
`
`C
`
`3.2
`2.9
`
`⬎1000
`⬎1000
`
`125
`109
`
`160
`45
`
`108
`171
`
`nM
`
`D
`
`0.9
`1.2
`
`⬎1000
`⬎1000
`
`7.9
`7.3
`
`114
`ND
`
`5.0
`1.3
`
`E
`
`2.4
`1.9
`
`⬎1000
`⬎1000
`
`1424
`1145
`
`77
`83
`
`7.5
`4.4
`
`F
`
`12.0
`12.8
`
`⬎1000
`⬎1000
`
`9265
`⬎10,000
`
`380
`ND
`
`6.8
`5.6
`
`S1P1
`IC50
`EC50
`S1P2
`IC50
`EC50
`S1P3
`IC50
`EC50
`S1P4
`IC50
`EC50
`S1P5
`IC50
`EC50
`ND, not determined.
`
`

`

`Immune Cell Regulation and Cardiovascular Effects of S1P Receptor Agonists
`
`763
`
`TABLE 2
`Lymphopenic and toxic effects of compounds in mice compared with binding activity on rodent S1P receptors
`ED50 values (⫾S.E.) are the dose (in milligrams per kilogram) that produces 50% of the maximum reduction in peripheral blood lymphocytes at 3 h. Toxicity is the maximum
`tolerated dose (milligrams per kilogram) as defined under Materials and Methods. Compounds were tested in vitro in 关33P兴S1P binding assays to determine IC50 values
`(nanomolar) using membranes prepared from CHO cells expressing mouse and rat S1P receptors.
`
`PBL ED50
`Toxicity
`mS1P1 IC50
`rS1P1 IC50
`mS1P3 IC50
`rS1P3 IC50
`mS1P4 IC50
`mS1P5 IC50
`ND, not determined.
`
`S1P
`
`ND
`0.4
`0.3
`0.3
`0.3
`0.3
`40
`0.3
`
`A
`
`0.03 (0.01)
`1.0
`0.3
`0.3
`2.7
`3.1
`28
`0.5
`
`C
`
`0.32 (0.08)
`4.0
`2.5
`2.1
`71
`74
`99
`29
`
`D
`
`0.063 (0.007)
`0.05
`0.9
`0.8
`1.8
`2.6
`130
`2.8
`
`E
`
`0.7 (0.4)
`⬎10
`4.1
`4.2
`605
`825
`109
`7.1
`
`F
`
`2.2 (0.8)
`⬎30
`14.6
`6.4
`6576
`7172
`200
`4.2
`
`Downloaded from
`
`jpet.aspetjournals.org
`
` at ASPET Journals on May 1, 2017
`
`ated for their ability to evoke bradycardia in the anesthetized
`rat. The extent of bradycardia, expressed as the peak decrease
`in heart rate as a percentage change from the average baseline
`value recorded for 20 min prior to compound administration, is
`shown in Fig. 4. The rank order of potency of S1P agonists for
`producing bradycardia in the anesthetized rat was Compound
`A ⱖ D ⬎ E ⬃ S1P ⬎⬎ Compound F. The reduced effects of
`Compounds E and F on heart rate are consistent with their
`reduced IC50 values against S1P3 in vitro.
`Effects of S1P Receptor Agonists in Conscious Rats.
`Administration of Compound A by continuous intravenous
`infusion to conscious rats produced time- and dose-dependent
`lymphopenia, bradycardia, and hypertension. At a dose of 10
`␮g/kg/min, bradycardia (Fig. 5A) and hypertension (Fig. 5B)
`were evident within 15 to 30 min postinitiation of infusion,
`with slower onset of effects at lower doses of compound (data
`not shown). Similarly, upon cessation of infusion, cardiovas-
`cular parameters returned to baseline values within 15 to 30
`min. The decrease in circulating leukocytes was also dose
`dependent (Fig. 6). The dose dependence for the pharmaco-
`logical effects of Compound A in conscious rats (Fig. 6) indi-
`cates that maximal lymphopenia was obtained at an infusion
`dose of less than 0.1 ␮g/kg/min, whereas significant changes
`in heart rate and mean arterial pressure were not evident
`until doses of 1 ␮g/kg/min or greater were administered.
`Effects of S1P Receptor Agonists in Conscious Mice.
`Toxicity of S1P and Compound D were abrogated in mice
`with a genetic deletion of the S1P3 receptor. In accordance,
`the cardiovascular effects of S1P receptor agonists were eval-
`
`Fig. 3. The effects of S1P (A) and Compound A (B) on mean arterial
`pressure (MAP) and heart rate (HR) were evaluated in barbiturate-
`anesthetized rats (three per group). HR and MAP were recorded contin-
`uously and are expressed as average values at 1-min intervals. Error bars
`have been omitted for the sake of clarity. Compounds were administered
`(arrow) as an intravenous bolus of approximately 10-s duration at 0.2 (A)
`or 0.05 (B) mg/kg. Significant differences from baseline control values are
`shown as: ⴱ, p ⬍ 0.05 for HR; and c, p ⬍ 0.05 for MAP.
`
`This late hypertension attained statistical significance in
`animals administered S1P but not in those administered
`Compound A.
`The effects of S1P receptor agonists with varying in vitro
`potency for activation of S1P1 and S1P3 receptors were evalu-
`
`Fig. 4. The effects of S1P receptor agonists on heart rate (HR) were
`evaluated in barbiturate-anesthetized rats. HR was determined for

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket