throbber
Downloaded from
`
`www.jlr.org
`
` by guest, on May 2, 2015
`
`CP-346086: an MTP inhibitor that lowers plasma cholesterol
`and triglycerides in experimental animals and in humans
`
`Charles E. Chandler, Donald E. Wilder, Judith L. Pettini, Yvette E. Savoy, Stephen F. Petras,
`1
`George Chang, John Vincent, and H. James Harwood, Jr.
`Department of Cardiovascular and Metabolic Diseases, Pfizer Global Research and Development, Groton
`Laboratories, Pfizer, Inc., Eastern Point Road, Groton, CT 06340
`
`Abstract A microsomal triglyceride transfer protein (MTP)
`inhibitor, CP-346086, was identified that inhibited both hu-
`man and rodent MTP activity [concentration giving half-max-
`imal inhibition (IC
`) 2.0 nM]. In Hep-G2 cells, CP-346086
`50
`inhibited apolipoprotein B (apoB) and triglyceride secretion
`(IC
` 2.6 nM) without affecting apoA-I secretion or lipid syn-
`50
`thesis. When administered orally to rats or mice, CP-346086
`lowered plasma triglycerides [dose giving 30% triglyceride
`lowering (ED
`) 1.3 mg/kg] 2 h after a single dose. Coadmin-
`30
`istration with Tyloxapol demonstrated that triglyceride lower-
`ing was due to inhibition of hepatic and intestinal triglyceride
`secretion. A 2 week treatment with CP-346086 lowered total,
`VLDL, and LDL cholesterol and triglycerides dose depen-
`dently with 23%, 33%, 75%, and 62% reductions at 10 mg/
`kg/day. In these animals, MTP inhibition resulted in in-
`creased liver and intestinal triglycerides when CP-346086 was
`administered with food. When dosed away from meals, how-
`ever, only hepatic triglycerides were increased. When admin-
`istered as a single oral dose to healthy human volunteers, CP-
`346086 reduced plasma triglycerides and VLDL cholesterol
`dose dependently with ED
`s of 10 mg and 3 mg, and maxi-
`50
`mal inhibition (100 mg) of 66% and 87% when measured 4 h
`after treatment. After a 2 week treatment (30 mg/day), CP-
`346086 reduced total and LDL cholesterol and triglycerides
`by 47%, 72%, and 75%, relative to either individual baselines
`or placebo, with little change in HDL cholesterol.
` Together,
`these data support further evaluation of CP-346086 in hyper-
`lipidemia.
`—Chandler, C. E., D. E. Wilder, J. L. Pettini, Y. E.
`Savoy, S. F. Petras, G. Chang, J. Vincent, and H. J. Harwood, Jr.
`CP-346086: an MTP inhibitor that lowers plasma cholesterol
`and triglycerides in experimental animals and in humans.
`J.
`
`Lipid Res. 2003.
` 1887–1901.
`44:
`
`Supplementary key words
`microsomal triglyceride transfer protein
`•
`lipid transfer inhibition
` very low density lipoprotein
` low density li-
`•
`•
`poprotein
` apolipoprotein B
` apolipoprotein A-I
` Hep-G2 cells
`•
`•
`•
`
`Cardiovascular disease remains the leading cause of death
`in industrialized nations and accounted for 950,000, or
`
`41%, of all deaths in the United States in 1998 (1). As a
`consequence of atherosclerosis, coronary heart disease
`(CHD) is the most common cause of cardiovascular mor-
`bidity and mortality, with an estimated 12 million people
`suffering from CHD in the United States alone (1). Ele-
`vated total and LDL cholesterol are both accepted pri-
`mary risk factors for atherosclerosis (1–3). An estimated
`101 million United States adults have elevated blood cho-
`⬎
`lesterol (
`200 mg/dl) and are candidates for LDL choles-
`terol lowering through dietary intervention (1, 4, 5). Of
`these, 41 million are considered high risk, having blood
`cholesterol greater than 240 mg/dl, and drug therapy is
`recommended (1, 4, 5).
`Epidemiological studies have shown that elevated tri-
`glycerides and reduced HDL cholesterol are also contrib-
`uting factors for the development of CHD (2, 3, 6–8).
`Among the adult United States population, 19% of people
`⬍
`have low HDL cholesterol (
`40 mg/dl) (3, 9, 10) and
`⬎
`21% have hypertriglyceridemia (
`150 mg/dl) (3, 10).
`Thus, as important as elevated LDL cholesterol is as a risk
`factor for CHD, it is important to recognize that the most
`common spectrum of lipid abnormalities is atherogenic
`dyslipidemia, which is present in 45–50% of men with
`CHD (11, 12) and includes borderline high-risk LDL cho-
`lesterol (e.g., 130–159 mg/dl), elevated triglycerides, small
`dense LDL particles, and low HDL cholesterol.
`The HMG-CoA reductase inhibitors (statins) are very
`effective in lowering LDL cholesterol and somewhat effec-
`tive in reducing triglycerides, but they have only minimal
`effects on HDL cholesterol (2, 5, 13–15). Indeed, al-
`though numerous clinical trials have demonstrated that
`LDL cholesterol reduction can significantly reduce CHD
`risk, a great number of treated subjects who achieve sub-
`stantial LDL cholesterol reduction still experience a clini-
`cal event (2, 3, 13–18). Therefore, with the goal of develop-
`
`Manuscript received 2 February 2003 and in revised form 3 June 2003.
`Published, JLR Papers in Press, July 1, 2003.
`DOI 10.1194/jlr.M300094-JLR200
`
`Abbreviations: CETP, cholesterol ester transfer protein; CHD, cor-
`onary heart disease; ER, endoplasmic reticulum; MTP, microsomal tri-
`glyceride transfer protein.
`1
` To whom correspondence should be addressed.
`e-mail: h_james_harwood@groton.pfizer.com
`
`Copyright © 2003 by the American Society for Biochemistry and Molecular Biology, Inc.
`
`This article is available online at http://www.jlr.org
`
`Journal of Lipid Research
`
`Volume 44, 2003
`
`1887
`
`CFAD Ex. 1016 (1 of 15)
`
`

`
`Downloaded from
`
`www.jlr.org
`
` by guest, on May 2, 2015
`
`Wilder, and Moberly (34). In the second stage of the pro-
`tocol, confirmed apoB secretion inhibitors were evaluated
`for their ability to inhibit the MTP-mediated transfer of ra-
`diolabeled triolein from synthetic phospholipid donor li-
`posomes to acceptor liposomes (34). Using this two-stage
`screening protocol, we identified CP-94792, a potent in-
`hibitor of apoB, but not apoA-I, secretion (33). Inhibition
`of apoB secretion was subsequently determined to be
`through inhibition of MTP activity (33, 35). However, al-
`though CP-94792 inhibited Hep-G2 cell apoB secretion with
`an half-maximal inhibition (IC
`) of 200 nM and inhib-
`50
`ited MTP-mediated triglyceride transfer (rat MTP) with an
`IC
` of 250 nM, the compound displayed only weak triglyc-
`50
`eride lowering activity when administered orally to rats (33).
`The potent and orally efficacious MTP inhibitor, CP-
`⬘
`346086 (4
`-trifluoromethyl-biphenyl-2-carboxylic acid [2-(2H-
`[1,2,4]triazol-3-ylmethyl)-1,2,3,4-tetrahydro-isoquinolin-6-yl]
`Fig. 1
`amide);
`, inset), was ultimately identified (33, 35–
`1
`37) by
`) employing a robotics-assisted parallel synthesis
`strategy as a means of developing structure-activity rela-
`2
`tionships and improving in vitro potency, and
`) using in
`vitro hepatic microsomal clearance and in vivo triglycer-
`ide lowering as guides for improving pharmacokinetic
`properties. In this report, we describe the biochemical
`
`Fig. 1.
`Inhibition of human microsomal triglyceride transfer pro-
`tein (MTP)-mediated neutral lipid transfer by CP-346086. Aliquots
`of solubilized human liver MTP, 150 ␮l, were incubated at 37⬚C for
`45 min with 50 ␮l donor liposomes, 100 ␮l acceptor liposomes, and
`200 ␮l assay buffer containing either 5% BSA (control) or 5% BSA
`plus sufficient CP-346086 to produce the indicated final concentra-
`tions of CP-346086, as described in Experimental Procedures. After
`incubation, triglyceride transfer was terminated by addition of 300
`␮l of a 50% (w/v) DEAE cellulose suspension in assay buffer. After
`a 4 min agitation, the donor liposomes, bound to DEAE cellulose,
`were selectively sedimented by low speed centrifugation (3,000 g, 5
`min). An aliquot of the supernatant containing the acceptor lipo-
`somes was assessed for radioactivity as outlined in Experimental
`Procedures. Shown is the percentage of control [14C]triolein or
`[14C]cholesterol oleate transfer as a function of CP-346086 concen-
`tration. Control triolein and cholesterol oleate transfer during
`the 45 min assay averaged 42% and 13%, respectively. Inset: The
`structure of CP-346086 (4⬘-trifluoromethyl-biphenyl-2-carboxylic
`acid [2-(2H-[1,2,4]triazol-3-ylmethyl)-1,2,3,4-tetrahydro-isoquinolin-
`6-yl]amide).
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`ing a therapy for treating patients with dyslipidemias that
`extend beyond primary hypercholesterolemia, we targeted
`inhibition of microsomal triglyceride transfer protein
`(MTP) as a mechanism for preventing triglyceride-rich li-
`poprotein assembly in the liver and intestine.
`MTP, which is located within the lumen of the endo-
`plasmic reticulum (ER) in hepatocytes and absorptive en-
`terocytes, is a heterodimeric protein consisting of a 97
`kDa subunit, which confers all of the lipid transfer activity
`of the heterodimer, and the 58 kDa multifunctional pro-
`tein disulfide isomerase (19). MTP plays a pivotal, if not
`obligatory role, in the assembly and secretion of triglycer-
`ide-rich, apolipoprotein B (apoB)-containing lipoproteins
`(VLDL and chylomicrons) from the liver and intestine
`and also catalyzes the transport of triglycerides, choles-
`teryl esters, and phospholipids between membranes (19–
`21). Although the exact role of MTP in the assembly of
`apoB-containing lipoproteins is still under investigation
`(21–23), MTP is proposed to transport lipids from the ER
`membrane to the growing apoB polypeptide chain in the
`ER lumen, thereby allowing proper translocation and
`folding of apoB to occur (19–24). MTP has also been pro-
`posed to mediate bulk triglyceride transfer into the ER lu-
`men for incorporation into poorly lipidated apoB-con-
`taining lipoprotein particles during the process of VLDL
`and chylomicron assembly (25, 26). Recent studies have
`also suggested a role for MTP in the movement of choles-
`terol ester into the ER lumen for inclusion into nascent
`apoB-containing lipoprotein particles (27).
`The initial suggestion that MTP inhibition could be a vi-
`able lipid-lowering therapy came with the discovery that
`functional MTP is absent in individuals with abetalipopro-
`teinemia, a genetic disorder characterized by low plasma
`cholesterol and triglycerides due to a defect in the assem-
`bly and secretion of apoB-containing lipoproteins (28,
`29). A similar phenotype is observed in MTP knockout
`mice (23, 30). Abetalipoproteinemia, however, represents
`an extreme example of MTP inhibition and is not without
`its clinical sequelae, all of which presumably are related
`directly or indirectly to fat malabsorption (steatorrhea),
`vitamin malabsorption, and hepatic and intestinal steato-
`sis (29, 31). A less severe, and probably more relevant, ex-
`ample of the consequences of therapeutic MTP inhibition
`is a related genetic disease, hypobetalipoproteinemia,
`caused by mutations in apoB (32). Heterozygous individu-
`als with this disease possess half of the normal levels of
`apoB-containing lipoproteins, lack the clinical signs and
`symptoms of abetalipoproteinemia, and have a normal
`lifespan (24).
`We devised a two-stage empirical screening protocol for
`compound evaluation (33) to identify potent MTP inhibi-
`tors with the potential to inhibit hepatic and intestinal
`apoB-containing lipoprotein assembly and consequently
`lower plasma total, LDL, and VLDL cholesterol and tri-
`glycerides in experimental animals and in humans. In the
`first stage of the protocol, compounds were evaluated for
`their ability to inhibit apoB, but not apoA-I secretion,
`from Hep-G2 cells in a high-throughput, 96-well multi-
`plexed format, essentially as described by Haghpassand,
`
`1888
`
`Journal of Lipid Research
`
`Volume 44, 2003
`
`CFAD Ex. 1016 (2 of 15)
`
`

`
`Downloaded from
`
`www.jlr.org
`
` by guest, on May 2, 2015
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`mechanism of action of CP-346086 that leads to its LDL
`cholesterol-, VLDL cholesterol-, and triglyceride-lowering
`efficacy in experimental animals and in humans.
`
`EXPERIMENTAL PROCEDURES
`
`Materials
`14
`14
`Sodium [2-
`C]acetate (56 mCi/mmol), [
`C]triolein (110
`14
`3
`mCi/mmol), cholesteryl [1-
`C]oleate (55 mCi/mmol), [
`H]tri-
`3
`olein (25 Ci/mmol), [
`H]egg phosphatidylcholine (50 mCi/
`mmol), and Aquasol-2 were from New England Nuclear (Boston,
`3
`MA). [
`H]glycerol (20 Ci/mmol) was from American Radiochemi-
`cals (St. Louis, MO). Ready Safe™ liquid scintillation cocktail was
`from Beckman Instruments (Fullerton, CA). Dulbecco’s modified
`Eagle’s medium (DMEM),
`-glutamine, and gentamicin were from
`l
`GIBCO Laboratories (Grand Island, NY). Heat-inactivated fetal bo-
`vine serum was from HyClone Laboratories (Logan, UT). DEAE
`cellulose was from Whatman International (Maidstone, England).
`Silica gel 60C TLC plates were from Eastman Kodak (Rochester,
`NY). BCA protein assay reagent was from Pierce (Rockford, IL).
`Cholesterol/HP reagent (Cat. 1127578), TG/GPO reagent (Cat.
`1128027), Preciset Cholesterol Calibrator Set (Cat. 125512), and
`Precitrol-N serum (Cat. 620212) were from Boehringer Mannheim
`(Indianapolis, IN). Cholesterol CII reagent kit (Cat. 276-64909),
`Triglyceride E reagent kit (Cat. 432-40201), Standard Cholesterol
`CII Solution (Cat. 277-64939), and Standard Triglyceride G Solu-
`tion (Cat. 998-69831) were from Waco Chemicals USA (Richmond,
`VA). Hep-G2 cells were from the American Type Culture Collec-
`tion (Rockville, MD). Mouse anti-human apoB monoclonal anti-
`bodies (MoAB-012), goat anti-human apoB polyclonal antibodies
`(AB-742), mouse anti-human apoA-I monoclonal antibodies
`(MAB-011), goat anti-human apoA-I polyclonal antibodies (AB-
`740), and human apoA-I purified standard (ALP10) were from
`Chemicon (Temecula, CA). B6CBAF1J mice were from Jackson
`Laboratory (Bar Harbor, ME). Transgenic huA1/CIII/cholesteryl
`ester transfer protein (CETP) mice, originally obtained from
`Charles River (Boston, MA), were bred on site. Sprague Dawley rats
`were from Charles River. RMH 3200 laboratory meal was from Ag-
`way, Inc. (Syracuse, NY). AIN76A semipurified diet was from US
`Biochemicals (Cleveland, OH). F0739 liquid diet powder was from
`Bio-Serve, Inc. (Frenchtown, NJ). Fast protein liquid chromatogra-
`phy (FPLC) instrumentation was from Gilson, Inc. (Middletown,
`WI). Superose-6 gel filtration columns were from Pharmacia (Pis-
`cataway, NJ). Postcolumn reaction (PCX) instrumentation was
`from Pickering Laboratories (Mountain View, CA). All other chem-
`icals and reagents were from previously listed sources (38, 39).
`
`Preparation of human MTP
`The procedure for isolating and solubilizing human MTP
`from frozen hepatic tissue is based on the method of Wetterau
`and Zilversmit (40) and was conducted essentially as described
`by Haghpassand, Wilder, and Moberly (34). Solubilized human
`⬚
`liver MTP was stored at 4
`C and was diluted 1:5 with assay buffer just
`before use. MTP preparations showed no notable loss of transfer
`activity with storage up to 30 days. Rat and mouse MTP were pre-
`pared similarly, except that freshly isolated liver tissue was used.
`
`Preparation of phospholipid vesicles for use in lipid
`transfer assays
`Donor and acceptor liposomes were prepared essentially as
`described by Wetterau et al. (28). Donor liposomes were pre-
`pared under nitrogen by room temperature bath sonication of a
`␮
`␮
`dispersion containing 447
`M egg phosphatidylcholine, 83
`M
`
`␮
`14
`bovine heart cardiolipin, and 0.91
`M [
`C]triolein (110 Ci/mol).
`Acceptor liposomes were prepared under nitrogen by room tem-
`perature bath sonication of a dispersion containing 1.3 mM egg
`␮
`3
`phosphatidylcholine, 2.6
`M triolein, and 0.5 nM [
`H]egg phos-
`phatidylcholine in assay buffer. The donor and acceptor lipo-
`⬚
`g
`somes were centrifuged at 160,000
` for 2 h at 7
`C. The upper
`80% of the supernatant, containing small unilamellar liposomes,
`⬚
`was carefully removed and stored at 4
`C until use.
`
`Measurement of MTP inhibitory activity
`MTP inhibitory activity as outlined by Haghpassand, Wilder,
`␮
`and Moberly (34) was determined by adding 200
`l of a buffer
`containing either 5% BSA (control) or 5% BSA plus CP-346086 to
`␮
`␮
`a mixture containing 50
`l donor liposomes, 100
`l acceptor lipo-
`␮
`somes, and 150
`l of solubilized, dialyzed MTP protein diluted in
`⬚
`assay buffer as outlined above. After incubation at 37
`C for 45 min,
`␮
`triglyceride transfer was terminated by addition of 300
`l of a 50%
`(w/v) DEAE cellulose suspension in assay buffer. After 4 min of ag-
`itation, the donor liposomes, bound to DEAE cellulose, were selec-
`
`g; 5 min). An
`tively sedimented by low speed centrifugation (3,000
`aliquot of the supernatant containing the acceptor liposomes was
`14
`3
`assessed for radioactivity, and the
`C and
`H counts obtained were
`used to calculate the percent recovery of acceptor liposomes and
`the percent triglyceride transfer using first order kinetics.
`
`Measurement of Hep-G2 cell apoB and apoA-I
`secretion inhibition
`HepG2 cells were grown in DMEM containing 10% fetal bo-
`vine serum in 96-well plates in a humidified, 5% CO
` atmo-
`2
`ⵑ
`⬚
`sphere at 37
`C until
`70% confluent. CP-346086 was dissolved
`␮
`in DMSO, diluted to 1
`M in growth medium, serially diluted in
`growth medium to the desired concentration range, and added
`␮
`in 100
`l aliquots to separate wells of 96-well culture plates con-
`taining HepG2 cells. Twenty-four hours later, growth medium
`was collected and assessed for apoB and apoA-I concentrations as
`described by Haghpassand and Moberly (41).
`
`Measurement of Hep-G2 cell cholesterol and fatty
`acid synthesis
`Cholesterol and fatty acid synthesis were evaluated in Hep-G2
`14
`cells by measuring incorporation of [2-
`C]acetate into cellular
`lipids as previously described (38, 39), with the following modifi-
`cations to allow simultaneous assessment of both cholesterol and
`fatty acid synthesis. After collection and assessment of the hexane
`fraction containing cholesterol and nonsaponifiable lipids as pre-
`viously described (38, 39), the remaining aqueous phase (con-
`⬍
`taining fatty acid sodium salts) was acidified to pH
`2 by addi-
`tion of 0.5 ml of 12 M HCl. The resulting mixtures were then
`transferred to glass conical tubes and extracted three times with
`4.5 ml hexane. The pooled organic fractions were dried under ni-
`␮
`trogen, resuspended in 50
`l of chloroform-methanol (1:1; v/v),
`⫻
`and applied to 1
` 20 cm channels of Silica Gel 60C TLC plates.
`Channels containing nonradioactive fatty acids were included on
`selected TLC plates as separation markers. TLC plates were devel-
`oped in hexane-diethyl ether-acetic acid (70:30:2; v/v/v) and air
`dried, and the region of chromatograms corresponding to fatty
`acid mobility was removed and assessed for radioactivity in Aqua-
`sol-2 using a Beckmann LS6500 liquid scintillation counter. Cho-
`14
`lesterol and fatty acid synthesis are expressed as dpm [2-
`C]ace-
`tate incorporated into either cholesterol or saponifiable lipids
`⬚
`during the 6 h incubation at 37
`C per mg cellular protein.
`
`Measurement of HepG2 cell triglyceride synthesis
`and secretion
`HepG2 cells, grown in T-75 flasks as previously described (38,
`⫻
`5
`39), were seeded into 24-well plates at 4–6
`10
` cells/well and
`
`Chandler et al.
`
`Lipid-lowering efficacy of the MTP inhibitor CP-346086
`
`1889
`
`CFAD Ex. 1016 (3 of 15)
`
`

`
`Downloaded from
`
`www.jlr.org
`
` by guest, on May 2, 2015
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`model 715-system controller software running under Microsoft
`Windows. The mobile phase was 154 mM NaCl, 1 mM EDTA,
`0.02% sodium azide (pH 8.1) at a flow rate of 0.36 ml/min. The
`⫻
`cholesterol-PCX reagent, Cholesterol/HP, was prepared at 2
`concentration by adding deionized water. The triglyceride-PCX
`⫻
`reagent, TG/GPO, was prepared at 2
` concentration by adding
`two bottles of enzyme to one bottle of buffer. The sterile filtered
`PCX reagents were stored and used under nitrogen in the dark
`⬚
`at 4
`C at a flow rate of 0.06 ml/min. Preciset Cholesterol Calibra-
`tors, Standard Cholesterol CII, Standard Triglyceride G, Pre-
`citrol-N serum, fresh human plasma (EDTA), and unknown sam-
`⬚
`ples were serially diluted in mobile phase and placed in a 2
`C
`␮
`refrigerated rack for FPLC sampling. Aliquots of 50–500
`l were
`injected automatically. Plasma standards were included to com-
`pare peak areas obtained via direct PCX injection (e.g., Super-
`ose-6 column bypassed), with peak areas obtained after column
`fractionation via FPLC-PCX and as an intra/inter-assay control
`sample, which was used before, interdispersed with, and after the
`unknown samples. The split-column eluent was combined with
`the cholesterol or triglyceride reagent in the PCX module form-
`ing a reaction product that was measured spectrophotometri-
`⬚
`cally at 500 nm after an 11 min pass through the 37
`C reaction
`coil. The Gilson 715 software performed the analysis of the spec-
`trophotometer output. The peak start, peak end, and baselines
`were checked visually and adjusted as necessary for each stan-
`dard, control plasma, and sample prior to integrating the peak
`areas. In the case of unresolved peaks, a perpendicular was
`drawn from each peak valley to the horizontal baseline for deter-
`mining the peak area. A plot of peak area versus quantity of cho-
`lesterol injected was made for the cholesterol and triglyceride
`standards, and a least squares regression analysis was performed
`and then used to convert chromatographic peaks from unknown
`samples into cholesterol and triglyceride concentrations in units
`of mg/dl.
`
`Studies using experimental animals
`The Institutional Animal Care and Use Procedures Review Board
`approved all procedures using experimental animals. B6CBAF1J
`mice, mice hemizygous for the human genes encoding apoA-I,
`apoC-III, and CETP (huA1/CIII/CETP mice), or Sprague Daw-
`ley rats were given food (RMH3200 laboratory meal or AIN76A
`semipurified diet) and water ad libitum and treated orally at a
`volume of 1.0 ml/200 g body weight (rats) or 0.25 ml/25 g body
`weight (mice) with either an aqueous solution containing 0.5%
`methyl cellulose (vehicle) or an aqueous solution containing
`0.5% methyl cellulose plus CP-346086. In experiments in which
`CP-346086 was administered in the feed, animals were fed pow-
`dered RMH3200 laboratory meal for 1 week prior to commenc-
`ing administration of CP-346086 as a dietary supplement to pow-
`dered chow. In studies in which rates of hepatic and intestinal
`triglyceride secretion were determined after Tyloxapol treat-
`ment, Tyloxapol (1.0 ml; 125 mg/ml) was administered intrave-
`nously 60 min after oral administration of CP-346086. In experi-
`ments in which rates of intestinal triglyceride absorption were
`14
`determined in the absence of Tyloxapol, [
`C]triolein mixed in
`Bioserve F0739 liquid diet [17.7% (w/w) fat] at a concentration
`␮
`of 0.5
`Ci/ml was administered orally to fasted mice as a 0.5 ml
`aliquot.
`
`Human subjects and study design
`Protocols involving human subjects were approved by the In-
`stitutional Protocol Review Board of Pfizer, Inc., and by the Pro-
`tocol Review Board of the Food and Drug Administration. Forty-
`eight healthy male adults, aged 18–45 years, participated in the
`randomized, double-blind, placebo-controlled, escalating single-
`dose study, and 30 healthy male adults, aged 18–45 years, partici-
`
`⬚
`C for
` atmosphere at 37
`maintained in a humidified, 5% CO
`2
`48 h prior to use. At the beginning of each experiment, media
`were removed and replaced with fresh media containing 0.2%
`⫾
`␮
`DMSO
` CP-346086. One hour after compound addition, 25
`l of
`␮
`3
`media containing 50
`Ci of [
`H]glycerol was added to each incuba-
`tion well. Plates were then sealed with Parafilm to avoid evapora-
`⬚
`tion, and cells were incubated at 37
`C for 6 h with gentle shak-
`ing. After incubation, the media were removed, and the cells
`were washed three times with PBS and scraped from wells into
`PBS. Lipids were extracted from the media and the cell lysate
`⫻
`with chloroform-methanol (2:1; v/v) and applied to 1
` 20 cm
`channels of Silica Gel 60C TLC plates. Channels containing non-
`radioactive triglycerides were included on selected TLC plates as
`separation markers. TLC plates were developed in petroleum
`ether-diethyl ether-acetic acid (75:25:1; v/v/v) and air dried. The
`region of chromatograms corresponding to triglyceride mobility
`was removed and assessed for radioactivity in Aquasol-2 using a
`Beckman LS6500 liquid scintillation counter. Triglyceride syn-
`3
`thesis and triglyceride secretion are expressed as dpm [
`H]glyc-
`erol incorporated into cellular triglycerides or secreted into the
`⬚
`culture medium during the 6 h incubation at 37
`C per mg cellu-
`lar protein.
`
`Measurement of plasma cholesterol and triglyceride levels
`Plasma triglyceride and total cholesterol concentrations were
`determined using Cholesterol CII and Triglyceride E reagent kits
`and employing Standard Cholesterol CII Solution and Standard
`Triglyceride G Solution as calibration standards.
`
`Measurement of hepatic and intestinal cholesterol and
`triglyceride levels
`Hepatic and intestinal cholesterol and triglyceride levels were
`measured as previously described (42) with the following modifi-
`cations. Sections, 0.5 g, of hepatic or intestinal tissue were ho-
`⬚
`mogenized at 4
`C in 3.0 ml PBS using a Polytron tissue homoge-
`␮
`nizer. Aliquots, 200
`l, of the homogenates were transferred to
`15 ml screw-capped glass tubes containing 7.5 ml of a mixture of
`CHCl
`-MeOH (2:1; v/v) and mixed vigorously for 20 s. One mil-
`3
`liliter of ddH
`O was then added, and the resulting suspension
`2
`was mixed vigorously for 15 s then centrifuged at 3,000 rpm for 5
`min at room temperature in a Sorvall RT-6000 bench-top centri-
`fuge. The chloroform-methanol layer was removed, placed in a
`⫻
`13
` 100 mm test tube, and evaporated to dryness under nitro-
`⬚
`␮
`gen at 60
`C. The lipid residue was resuspended in 200
`l 1% Tri-
`ton X-100 in absolute EtOH, and the cholesterol and triglyceride
`concentrations were determined using Cholesterol CII and Tri-
`glyceride E reagent kits, adapted for colorimetric analysis in 96-
`well plate format.
`
`Plasma lipoprotein subfractionation and visualization by
`FPLC and PCX
`Plasma lipoproteins were separated on a single Superose-6 col-
`umn by FPLC, and the effluent was split into two equal streams.
`One stream was reacted postcolumn with cholesterol enzymatic
`assay reagents for the determination of lipoprotein cholesterol.
`The second stream was reacted postcolumn with triglyceride en-
`zymatic assay reagents for the determination of lipoprotein tri-
`glyceride. Lipoprotein FPLC utilized a single Gilson autoinjector
`and dual PCX setups, each consisting of a Pickering nitrogen
`pressurized reagent delivery system, a Gilson reagent pump, a
`Pickering CRX 400 PCX with a custom 2.8 ml knitted reaction
`coil, and a Gilson spectrophotometer set at 500 nm. The stream
`splitting method used one Gilson mobile phase pump, one Phar-
`⫻
`macia Superose-6 10
` 300 mm column, and multiple switching
`valves for controlling effluent stream splitting and PCX reagent
`flow. The entire system was computer controlled by Gilson
`
`1890
`
`Journal of Lipid Research
`
`Volume 44, 2003
`
`CFAD Ex. 1016 (4 of 15)
`
`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`III concentrations by ELISA, CP-346086 inhibited Hep-G2
`cell apoB secretion with an IC50 of 2.0 nM (Fig. 2A). Un-
`der these conditions, neither apoA-I secretion (Fig. 2A)
`nor apoC-III secretion (not shown) were inhibited by CP-
`
`Downloaded from
`
`www.jlr.org
`
` by guest, on May 2, 2015
`
`Fig. 2.
`Inhibition of Hep-G2 cell apolipoprotein B (apoB) and
`triglyceride secretion by CP-346086. A: Hep-G2 cells seeded and
`maintained in culture as described in Experimental Procedures
`were incubated for 24 h at 37⬚C in supplemented DMEM plus the
`indicated concentrations of CP-346086. After incubation, the me-
`dium was removed and assessed for apoB and apoA-I concentra-
`tions by apolipoprotein-specific ELISAs as outlined in Experimen-
`tal Procedures. Data for apoB and apoA-I secretion are the mean of
`triplicate determinations ⫾ SD and are expressed as a percentage
`of control apolipoprotein secretion as a function of CP-346086 con-
`centration. B: Hep-G2 cells seeded and maintained in culture as de-
`scribed in Experimental Procedures were incubated for 6 h at 37⬚C
`in supplemented DMEM containing either 1% DMSO (control) or
`1% DMSO containing CP-346086 sufficient to produce the indi-
`cated final inhibitor concentrations. For measurement of triglycer-
`ide synthesis and secretion, cells also received 50 ␮Ci of [3H]glyc-
`erol (closed symbols). After incubation, the media was removed,
`the cells were washed three times with PBS, and the secreted (me-
`dia) and cellular triglycerides were quantitated as described in Ex-
`perimental Procedures. For measurement of cholesterol and fatty
`acid synthesis, cells also received 4 ␮Ci of [2-14C]acetate (open sym-
`bols). After incubation, newly synthesized cholesterol and fatty ac-
`ids were separated and quantitated as described in Experimental
`Procedures. Data for triglyceride secretion (closed circles), triglyc-
`eride synthesis (closed triangles), cholesterol synthesis (open dia-
`monds), and fatty acid synthesis (open triangles) are the mean of
`triplicate determinations ⫾ SD and are expressed as a percentage
`of control synthesis and secretion as a function of CP-346086 con-
`centration.
`
`pated in the 2 week randomized, double-blind, placebo-con-
`trolled, parallel-group multidose study. Inclusion criteria for
`both studies were body weight within 10% of ideal based on 1983
`Metropolitan Life Insurance Height and Weight Tables (43), fast-
`ing plasma cholesterol levels in the upper 50% of the normal
`range for the American population based on age, sex, and race
`(e.g., 180–250 mg/dl) (1), fasting plasma triglyceride levels
`⬎
`⬍
`150 mg/dl and
`400 mg/dl, no evidence of clinically signifi-
`cant disease based on complete medical history, full physical ex-
`amination, 12-lead ECG and clinical laboratory testing, and no
`prescription, OTC, or other drug usage for at least 2 weeks prior
`to beginning the study. Subjects were confined to the Clinical Re-
`search Unit and fed standardized meals (30% of daily calories
`from fat).
`In the single-dose study, after randomization (six subjects/
`group; eight groups) each group was assigned a CP-346086 dose
`ranging in half-log intervals from 0.1 mg to 300 mg. At 7 AM on
`the day of study, after fasting for at least 8 h, four subjects from
`each group received their respective doses of CP-346086, and two
`subjects received placebo. Plasma samples were obtained at vari-
`ous times over the next 72 h for use in determining total and li-
`poprotein cholesterol and triglyceride levels by FPLC (see above).
`In the multidose study, subjects were randomized with eight
`receiving 30 mg CP-346086 and six receiving placebo at 10 PM
`(bedtime) for 14 days. Plasma samples were obtained daily pre-
`⫺
`⬚
`dose, frozen at
`20
`C, and forwarded to Medical Research Labs
`(39 Excelsiorlaan Zaventem, Brussels, Belgium) for determina-
`tion of total and lipoprotein cholesterol and triglyceride levels
`using standardized autoanalyzer technology.
`
`RESULTS
`
`Inhibition of MTP-mediated lipid transfer by CP-346086
`When donor and acceptor liposomes, prepared as de-
`scribed in Experimental Procedures, were incubated with
`varying concentrations of inhibitor, CP-346086 dose depen-
`dently inhibited human MTP-mediated triglyceride trans-
`fer between vesicles with a concentration giving an IC
`50
`equal to 2.0 nM (Fig. 1). Similarly, when the radiolabeled
`triolein of donor vesicles was replaced by radiolabeled
`cholesteryl oleate, CP-346086 inhibited transfer of choles-
`teryl oleate between vesicles with an IC50 of 1.9 nM (Fig.
`1), demonstrating the compound’s ability to equally in-
`hibit transfer of both neutral lipids. Similar IC50 values
`were also noted for inhibition of rat (1.7 nM) and mouse
`(6.7 nM) MTP activity.
`Inhibition of neutral lipid transfer by CP-346086 was
`specific to MTP-mediated lipid transfer, however, as in an
`identical assay in which MTP was replaced by CETP, no in-
`hibition of CETP-mediated lipid transfer was observed at
`concentrations of CP-346086 up to 10 ␮M (data not
`shown), indicating the compound’s specificity for inhibi-
`tion of MTP-mediated lipid transfer and demonstrating
`the lack of an effect of CP-346086 on the physicochemical
`properties of the donor and acceptor vesicles.
`
`Inhibition of apoB and triglyceride secretion from
`Hep-G2 cells by CP-346086
`When Hep-G2 cells were incubated with various con-
`centrations of CP-346086 for 24 h at 37⬚C, and the culture
`media was evaluated for human apoB, apoA-I, and apoC-
`
`Chandler et al.
`
`Lipid-lowering efficacy of the MTP inhibitor CP-346086
`
`1891
`
`CFAD Ex. 1016 (5 of 15)
`
`

`
`Downloaded from
`
`www.jlr.org
`
` by guest, on May 2, 2015
`
`Fig. 3.
`Inhibition of hepatic and intestinal triglyceride secretion
`by CP-346086. Sprague Dawley rats given RMH3200 laboratory meal
`and water ad libitum were fasted overnight and then

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