throbber
BMC Cardiovascular Disorders
`
`Cl
`
`()
`BioMed Central
`
`Research article
`O~nAccess
`JTT-130, a microsomal triglyceride transfer protein (MTP) inhibitor
`lowers plasma triglycerides and LDL cholesterol concentrations
`without increasing hepatic triglycerides in guinea pigs
`Dimple Aggarwal1, Kristy L West1, Tosca L Zern 1, Sudeep Shrestha1,
`Marcela Vergara-Jimenez2 and Maria Luz Fernandez* 1
`
`Address: 1 Department of Nutritional Sciences, University of Connecticut, Storrs, CT, USA and 2Department of Nutritional Sciences, University of
`Sinaloa, Culiacan, Mexico
`
`Email: Dimple Aggarwal - dimple.aggarwal@uconn.edu; Kristy L West - kristy_west@hotmail.com; Tosca L Zern - toscal9@hotmail.com;
`Sudeep Shrestha - sudeep.shrestha@uconn.edu; Marcela Vergara-Jimenez - marveji@uas.uasnet.mx; Maria Luz Fernandez• - maria(cid:173)
`Iuz.fernandez@uconn.edu
`• Corresponding author
`
`Published: 27 September 2005
`
`BMC Cardiovascular Disorders 2005, 5:30 doi: I 0.118611471-2261-5-30
`
`This article is available from: http://www.biomedcentral.com/l 471-2261/5/30
`
`Received: 22 June 2005
`Accepted: 27 September 2005
`
`© 2005 Aggarwal et al; licensee BioMed Central Ltd.
`This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http'i/creatjvecommons orgllicenses/by/2 0),
`which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
`
`Abstract
`Background: Microsomal transfer protein inhibitors (MTPi} have the potential to be used as a
`drug to lower plasma lipids, mainly plasma triglycerides (TG}. However, studies with animal models
`have indicated that MTPi treatment results in the accumulation of hepatic TG. The purpose of this
`study was to evaluate whether JTT-130, a unique MTPi, targeted to the intestine, would effectively
`reduce plasma lipids without inducing a fatty liver.
`Methods: Male guinea pigs (n = I 0 per group) were used for this experiment. Initially all guinea
`pigs were fed a hypercholesterolemic diet containing 0.08 g/ I 00 g dietary cholesterol for 3 wk.
`After this period, animals were randomly assigned to diets containing 0 (control), 0.0005 or 0.0015
`g/ I 00 g of MTPi for 4 wk. A diet containing 0.05 g/ I 00 g of atorvastatin, an HMG-CoA reductase
`inhibitor was used as the positive control. At the end of the ]th week, guinea pigs were sacrificed
`to assess drug effects on plasma and hepatic lipids, composition of LDL and VLDL, hepatic
`cholesterol and lipoprotein metabolism.
`
`Results: Plasma LDL cholesterol and TG were 25 and 30% lower in guinea pigs treated with MTPi
`compared to controls (P < 0.05). Atorvastatin had the most pronounced hypolipidemic effects with
`a 35% reduction in LDL cholesterol and 40% reduction in TG. JTT-130 did not induce hepatic lipid
`accumulation compared to controls. Cholesteryl ester transfer protein (CETP) activity was
`reduced in a dose dependent manner by increasing doses of MTPi and guinea pigs treated with
`atorvastatin had the lowest CETP activity (P < 0.0 I). In addition the number of molecules of
`cholesteryl ester in LDL and LDL diameter were lower in guinea pigs treated with atorvastatin. In
`contrast, hepatic enzymes involved in maintaining cholesterol homeostasis were not affected by
`drug treatment.
`
`Conclusion: These results suggest that JTT-130 could have potential clinical applications due to
`its plasma lipid lowering effects with no alterations in hepatic lipid concentrations.
`
`Page 1of8
`(page number not for citation purposes)
`
`1 of 8
`
`PENN EX. 2264
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`BMC Cardiovascular Disorders 2005, 5:30
`
`http://www.biomedcentral.com/1471-2261/5/30
`
`Background
`Microsomal triglyceride transfer protein (MTP) is a resi(cid:173)
`dent protein in the lumen of endoplasmic reticulum and
`is primarily responsible for transfer of triglycerides (TG)
`and other lipids from their site of synthesis in the endo(cid:173)
`plasmic reticulum into the lumen during the assembly of
`very low density lipoprotein (VLDL) [l]. VLDL produced
`by the liver are the major source ofLDL in plasma and ele(cid:173)
`vated levels ofLDL are associated with the development of
`atherosclerosis and cardiovascular disease
`(CVD).
`Increased total cholesterol and LDL cholesterol (LDL-C)
`are both considered primary risk factors for atherosclero(cid:173)
`sis [2,3]. To reduce CHD risk factors improvements in diet
`and exercise are primary recommendations however
`when plasma cholesterol concentrations reach a certain
`limit drug intervention is necessary. Statins, which are tar(cid:173)
`geted to 3-hydroxy-3-methylglutaryl coenzyme A (HMG(cid:173)
`CoA) reductase and are used extensively, are effective in
`lowering LDL-C, and somewhat effective in reducing
`plasma TG (4,5]. A number of studies done in the past
`have indicated that reduction in LDL-C values by using
`statins can significantly reduce the risk of CHD however a
`large population of patients still experience a clinical
`event (2,4,5]. Therefore, pharmaceutical companies are
`continuing to research other drug options to control
`hypercholesterolemia with the goal of developing a ther(cid:173)
`apy for treating patients with dyslipidemias. Microsomal
`triglyceride transfer protein inhibitor (MTPi) is one such
`option. It is believed that blocking MTP will not only
`reduce plasma total and LDL cholesterol (LDL-C) but also
`plasma VLDL and TG by affecting the packaging and secre(cid:173)
`tion of VLDL and chylomicrons. Certain animal and
`human studies [ 6, 7] have shown that the inhibition of
`MTP blocks the hepatic secretion ofVLDL and the intesti(cid:173)
`nal secretion of chylomicrons. Consequently, this mecha(cid:173)
`nism provides a highly efficacious pharmacological target
`for the lowering of LDL-C and reduction of postprandial
`lipemia. These effects could afford unprecedented benefit
`in the treatment of atherosclerosis and consequent cardi(cid:173)
`ovascular disease. The promise of this therapeutic target
`has attracted widespread interest in the pharmaceutical
`industry.
`
`This research study had a primary goal to evaluate
`whether (fIT-130), an MTPi reduces plasma cholesterol
`and triglyceride concentrations in male Hartley guinea'
`pigs. Since fIT-130 is mainly targeted to the intestine,
`another main objective of this study was to evaluate
`whether this MPfi resulted in less hepatic lipid accumula(cid:173)
`tion compared to other inhibitors [ 6, 7]. Guinea pigs were
`used as the animal model for this study because of their
`similarities to humans in terms of hepatic cholesterol and
`lipoprotein metabolism. Previous studies done in our lab(cid:173)
`oratory report that guinea pig serve as a good model for
`evaluating cholesterol lowering drugs [8-10].
`
`Methods
`Materials
`Reagents were obtained from the following sources. fIT-
`130, the MTPi tested was provided by Akros Pharma Inc
`(Princeton, NJ). Enzymatic cholesterol and TG kits, cho(cid:173)
`lesterol oxidase, cholesterol esterase and peroxidase were
`purchased from Roche-Diagnostics (Indianapolis, IN).
`Phospholipid and free cholesterol enzymatic kits were
`obtained from Wako Pure Chemical (Osaka, Japan).
`Quick-seal ultracentrifuge tubes were from Beckman
`(Palo Alto, CA). DL-hydroxy- [3-14CJ methyl glutaryl
`coenzyme A (1.81 GBq/mmol), DL- [5-3HJ mevalonic
`acid (370 GBq/mmol), cholesteryl- [1,2,6,7-3H] oleate
`(370 GBq/mmol), Aquasol, Liquiflor (toluene concen(cid:173)
`trate) and [1 4CJ cholesterol were purchased from DuPont
`NEN (Boston, MA). Oleoyl- [1-I4CJ coenzyme A (1.8
`GBq/mmol) and DL-3-hydroxy-3-methyl glutaryl coen(cid:173)
`zyme A were obtained from Amersham (Clearbrook, IL).
`Cholesteryl oleate, glucose-6-phosphate, glucose-6-phos(cid:173)
`phate dehydrogenase, nicotinamide adenine dinucleotide
`phosphate (NADP), sodium fluoride, Triton, bovine
`serum albumin and sucrose were obtained from Sigma
`Chemical (St. Louis, MO). Aluminum and glass silica gel
`plates were purchased from EM Science (Gibbstown, NJ).
`
`Diets
`Diets were prepared and pelleted by Research Diets (New
`Brunswick, NJ). Isocaloric diets were designed to meet all
`the nutritional requirements for guinea pigs. The four
`diets had identical composition except for the type and
`dose of tested drug as indicated in Table 1. The amount of
`cholesterol in the diets was adjusted to be 0.08 g/100 g, an
`amount equivalent to 600 mg/day in the human diet [ 11].
`
`Animals
`Forty male guinea pigs (Harlan Sprague-Dawley, Hills),
`weighing 250-300 g, were randomly assigned to either a
`control, low dose of MTPi (LDI), high dose of MTPi (HDI)
`or an atorvastatin (AT) treatment (n = 10/group) for 4
`weeks. Initially, all guinea pigs were fed the control diet
`for 3 weeks to raise plasma cholesterol concentrations.
`Two animals were housed per metal cage in a light cycle
`room (light from 0700-1900 h) and had free access to
`diets and water. Non-fasted guinea pigs were sacrificed by
`heart puncture after isoflurane anesthesia. Blood and liv(cid:173)
`ers were harvested for analysis and were stored at -80 ° C
`for further analysis. All animal experiments were con(cid:173)
`ducted in accordance with U.S. Public Health Service/U.S.
`Department of Agriculture guidelines. Experimental pro(cid:173)
`tocols were approved by the University of Connecticut
`Institutional Care and Use Committee.
`
`Lipoprotein isolation
`Plasma samples were collected from blood obtained by
`heart puncture from guinea pigs under anesthesia. A
`
`Page 2 of 8
`(page number not for citation purposes)
`
`2 of 8
`
`PENN EX. 2264
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`BMC Cardiovascular Disorders 2005, 5:30
`
`http://www.biomedcentral.com/14 71-2261 /5/30
`
`Table I: Composition of Control, low dose of the inhibitor (LOI), high dose of the inhibitor (HDI} and atorvastatin diets
`
`Components
`
`Control%
`
`Soybean protein
`Methionine
`Sucrose
`Corn Starch
`Fat mixl
`Cellulose
`Guar gum
`Mineral Mix2
`Vitamin Mix2
`Cholesterol
`JTT-130
`Statin
`
`22.S
`o.s
`2S
`IS
`IS. I
`10
`2.S
`8.2
`I.I
`0.08
`0
`0
`
`LOI%
`
`22.S
`o.s
`2S
`IS
`IS.I
`10
`2.S
`8.2
`I.I
`0.08
`o.ooos
`0
`
`HDI%
`
`Atorvastatin %
`
`22.S
`o.s
`2S
`IS
`IS.I
`10
`2.S
`8.2
`I.I
`0.08
`O.OOIS
`0
`
`22.S
`o.s
`2S
`IS
`IS.I
`10
`2.S
`8.2
`I.I
`0.08
`0
`o.os
`
`I Fat mix for the diet contains olive oil-palm kernel oil-safflower oil (I :2: 1.8), high in lauric and myristic acids.
`2 Mineral and vitamin mix adjusted to meet NRC requirements for guinea pigs. Detailed composition of the vitamin and mineral mix has been
`reported elsewhere (Fernandez et al. I 992b).
`
`preservation cocktail of aprotonin, phenyl methyl sulfo(cid:173)
`nyl fluoride and sodium azide was added to plasma sam(cid:173)
`ples to minimize changes in lipoprotein composition
`during isolation. Plasma was aliquoted for LCAT and
`CEfP determinations, plasma lipid analysis and lipopro;
`tein isolation.
`''
`
`Lipoproteins were isolated by sequential ultracentrifuga(cid:173)
`tion [12] in a LE-SOK ultracentrifuge (Beckman Instru(cid:173)
`ments, Palo Alto, CA). VLDL was isolated at d = 1.006 kg/
`Lat 125,000 g at 15 °C for 19 h in a Ti-50 rotor. LDL was
`isolated at d = 1.019-1.09 kg/Lin quick-seal tubes at 15 ° C
`for 3 hat 200,000 gin a vertical Ti-65 rotor [13). LDLsam(cid:173)
`ples were dialyzed in 0.9 g/L sodium chloride-0.1 g/L eth(cid:173)
`ylene diamine tetra acetic acid (EDTA), pH 7.2, for 12 h
`and stored at 4 ° C for further analysis.
`
`Plasma and hepatic lipids
`Plasma samples were analyzed for cholesterol and TG by
`enzymatic methods [14]. Hepatic total and free choles(cid:173)
`terol and TG were determined according to the method by
`Carr et al. [15] following extraction of hepatic lipids with
`chloroform-methanol 2: 1. Cholesteryl ester concentra(cid:173)
`tions were calculated by subtracting free from total
`cholesterol.
`
`Lipoprotein characterization
`VLDL and LDL composition was calculated by determin(cid:173)
`ing free and esterified cholesterol [14], protein by a modr.
`ified Lowry method [16], and TG and phospholipids by
`enzymatic kits. VLDL apo B was selectively precipitated
`with isopropanol [17). The number of constituent mole(cid:173)
`cules of LDL was calculated on the basis of one apo B per
`particle with a molecular mass of 412000 kD[l8]. The
`molecular weights were 885.4, 386.6, 645 and 734 forTG>
`free and esterified cholesterol, and phospholipids, respec-
`
`tively [19]. LDL diameters were calculated according to
`Van Heek et al (20). HDL cholesterol was also determined
`according to Warnick et al, with a modification, which
`consisted of using 2 mol/L MgC12 for precipitation of apo(cid:173)
`B containing lipoproteins [ 13).
`
`Lecithin Cholesterol Acyltransferase (LCAT) and
`Cholesterol Ester Transfer Protein (CETP) determinations
`in plasma
`LCAT and CETP activities were determined according to
`Ogawa & Fielding [21). Physiological CETP activity was
`determined without inhibiting LCAT activity by measur(cid:173)
`ing the mass transfer of cholesterol ester between HDL
`and apo B containing lipoproteins. Samples were incu(cid:173)
`bated at 3 7 ° C for 6 h in a shaking water bath and total
`and free plasma cholesterol and HDL cholesterol were
`measured. LCAT activity was determined by mass analysis
`of the decrease in plasma free cholesterol between 0 and
`6 h at 37°C. Assays were carried out concurrently with
`measurements of CETP. Both of these methods have been
`well-standardized for guinea pig plasma (22].
`
`Hepatic microsome isolation
`Hepatic microsomes were isolated as described previously
`[8]. Briefly a microsomal fraction was isolated by two 25-
`min centrifugations at 10,000 g (JA-20 rotor, J2-2 l) fol(cid:173)
`lowed by ultracentrifugation at 100,000 gin a Ti-50 rotor
`at 4 ° C for 1 hour. Microsomes were resuspended in the
`homogenization buffer and centrifuged for an additional
`hour at 100,000 g. After centrifugation, microsomal pel(cid:173)
`lets were homogenized and stored at-70°C.
`
`Hepatic HMG-CoA reductase assay
`The activity of microsomal HMG-CoA reductase (E.C.
`1.1.1.34) was measured
`in hepatic microsomes as
`described by Shapiro et al. (23]. HMG-CoA reductase
`
`Page 3 of 8
`(page number not for citation purposes)
`
`3 of 8
`
`PENN EX. 2264
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`BMC Cardiovascular Disorders 2005, 5:30
`
`http://www.biomedcentral.com/14 71-2261 /5/30
`
`activity was expressed as pmol of [1 4C] mevalonate pro(cid:173)
`duced per min per mg microsomal protein. Recoveries of
`[3H] mevalonate ranged from 60-90%.
`
`Hepatic Acy/ CoA Cholesteryl Acyltransferase (ACAT)
`activity
`Hepatic ACAT (E.C. 2.3.1.26) activity was measured by
`the incorporation of [ 14C] oleoyl CoA in cholesteryl ester
`in hepatic microsomes by preincubating 0.8-1 mg of
`microsomal protein per assay with 84 g/L albumin and
`buffer for microsomal isolation [24]. Recoveries of (3H]
`cholesteryl oleate were between 70-90%.
`
`Hepatic cholesterol 7 a-hydroxylase activity
`Cholesterol 7a-hydroxylase (E.C. 1.14.13. 7) activity was
`measured according to the method modified by Jelinik et
`al [25]. (14C] cholesterol was used as a substrate and deliv(cid:173)
`ered as cholesterol-phosphatidylcholine liposomes (1:8
`by weight) prepared by sonication. An NADPH-regenerat(cid:173)
`ing system (glucose-6-phosphate dehydrogenase, NADP,
`and glucose-6-phosphate) was included in the assay as a
`source of NADPH.
`
`700
`
`600
`
`c;
`:::- 400
`.c
`"" a; 300
`;;::
`
`100
`
`'
`'!
`
`Time (weeks)
`
`Figure I
`Weight gain of guinea pigs treated with control, low dose,
`high dose of JTT-130 and atorvastatin.
`
`Statistical analysis
`One-way analysis of variance (ANOVA) (SSPS for Win(cid:173)
`dows version 12) was used to evaluate significant differ(cid:173)
`ences among groups in regards to plasma and hepatic
`lipids, LDL composition, hepatic enzyme activities and
`LCAT and CEfP activities. The LSD post hoc test was used
`to evaluate the differences among groups. Data are pre(cid:173)
`sented as the mean± SD. Differences were considered sig(cid:173)
`nificant at P < 0.05.
`
`The changes in plasma TC values were mostly due to
`decreases in the cholesterol carried by LDL. LDL-C was
`also significantly decreased (24. 7% & 26.9%) by the MTPi
`diets tested. There were no major differences between
`these two doses for plasma lipid parameters except for
`VLDL-C, which were significantly higher when compared
`to the low dose of the drug. No significant differences
`were observed for HDL-C values with MTPi or with AT
`(Table 2).
`
`Results
`Plasma lipids and lipoproteins
`No difference in weight gain overtime was observed in
`guinea pigs fed the different test diets (Fig 1 ), indicating
`that animals consumed comparable amounts of their
`respective test diets. After feeding the test diets for a period
`of four weeks, blood was isolated and plasma was ana(cid:173)
`lyzed for cholesterol and TG concentrations. The two
`doses of MTPi evaluated, low dose (LDI) and high dose
`(HDI) decreased plasma total cholesterol values signifi(cid:173)
`cantly by 19.2% (P < 0.01) as compared to the control ani(cid:173)
`mals (Table 2). There was no significant difference
`between the two doses of MTPi tested. Atorvastatin, which
`was used as a positive control, led to a significantly robust
`decrease in plasma total cholesterol values of 46%, which
`was significantly different (P < 0.01) from the two MTPi
`doses used. Plasma TG values were also significantly lower
`in LDI (50.8%) and HDI ( 45.3%) when compared to their
`control counterparts whereas atorvastatin (AT) treatment
`resulted in the maximum decrease of plasma TG (Table
`2).
`
`LDL size and composition
`No significant effect of MTPi on the number of CE mole(cid:173)
`cules or on the size of LDL particle was found with any of
`the two doses tested as compared to their control counter(cid:173)
`parts (Table 3). ATtreatment reduced the number of ester(cid:173)
`ified cholesterol molecules ( 45%) as well as decreased the
`size ofLDL particle (30%) (Table 3).
`
`LCAT and CETP activities
`Table 3 also summarizes the activities of these two pro(cid:173)
`teins, which play a major role in the intravascular process(cid:173)
`ing of plasma cholesterol. There were no significant
`differences in LCAT activity when comparing MTPi or sta(cid:173)
`tin groups to the control group. However, HDI decreased
`the activity of CEfP, which was comparable to the atorv(cid:173)
`astatin treated group (P < 0.01).
`
`Hepatic lipids and enzymes
`No significant changes were found in hepatic total choles(cid:173)
`terol, free cholesterol, cholesteyl ester or TG values in any
`of the four treatments (Table 4). Results suggest that MTPi
`did not lead to lipid accumulation in the liver, as there
`
`Page 4 of 8
`(page number not for citation purposes)
`
`4 of 8
`
`PENN EX. 2264
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`BMC Cardiovascular Disorders 2005, 5:30
`
`http://www.biomedcentral.com/1471-2261 /5/30
`
`Table 2: Plasma total cholesterol (TC), triglycerides (TG), VLOL-C, LOL-C and HOL-C of guinea pigs fed a control diet, low dose MTPi
`(LOI), high dose MTPi (HOI) or atorvastatin
`
`Diets
`
`TC
`
`TG
`
`VLDL-C
`
`LDL-C
`
`HDL-C
`
`Control (I 0)
`LDI (10)
`HDI (9)
`Atorvastatin (9)
`
`146.9 ± 42.2•
`116.9± 29.7b
`116.1 ± 23.3b
`76.5 ± 29.8<
`
`135.8 ± I 18.9•
`66.7 ± 29.6b
`74.3 ± 31.?b
`49.4 ± 32.2<
`
`(mg/dL}
`8.1 ± 5.S•b
`3.7 ± 3.0•
`9.0 ± 8.6b
`2.9 ± 2.6•
`
`123.9 ± 43.5•
`93.3 ± 26.2b
`90.5 ± 28.8b
`59.4 ± 29.3<
`
`13.6 ± 4.5•
`18.5 ± 8.0•
`I 5.6 ± 7.1•
`14.1 ± 5.6•
`
`1 Data are presented as mean ± SD for the number of guinea pigs indicated in parenthesis. Numbers in a column with different superscripts are
`considered significantly different (P < 0.0 I) as determined by one way AN OVA and LSD as a post-hoc test
`
`Table 3: Number of molecules of cholesteryl ester (CE), LOL diameter and LCAT and CETP activities of guinea pigs fed a control diet,
`low dose MTPi (LOI), high dose MTPi (HOI) or atorvastatin.
`
`Diets
`
`CE molecules
`
`LDL diameter
`
`LCAT
`
`CETP
`
`Control (n = I 0)
`LDI (n= 10)
`HDI (n = 9)
`Atorvastatin ( n = 9)
`
`1993 ± 422•
`2072 ± 536•
`2026 ± 132.5•
`1080 ± 1092b
`
`nanometers
`16.47 ± 3.63•
`16.67 ± 3.05•
`16.58 ± 7.85•
`I 1.52 ± 2.69b
`
`(pmol/min.mg)
`
`19.6 ± 11.0•
`14.4 ± 6.4•
`14.6 ± 8.5•
`13.4 ± 10.9•
`
`36.I ± 12.5•
`31.0 ± 21.I•
`19.4 ± 13.0•b
`12.8 ± 4.2b
`
`1 Data are presented as mean ± SD for the number of guinea pigs indicated in parenthesis. Numbers in a column with different superscripts are
`considered significantly different (P < 0.01) as determined by one way ANOVA and LSD as post hoc test.
`
`Table 4: Hepatic total cholesterol (TC), free cholesterol (FC) cholesteryl ester (CE) and TG of guinea pigs fed a control diet, low dose
`MTPi (LOI), high dose MTPi (HOI) or atorvastatinl,
`
`Diets
`
`TC
`
`FC
`
`CE
`
`TG
`
`Control (n = I 0)
`LDI (n = 10)
`HDI (n = 9)
`Atorvastatin (n = 9)
`
`1.20 ± 0.53
`1.47 ± 0.40
`1.42 ± 0.66
`1.71±0.76
`
`(mg/g)
`
`1.00 ± 0.44
`1.24 ± 0.35
`0.98 ± 0.39
`1.31 ± 0.56
`
`0.20 ± 0.23
`0.23 ± 0.11
`0.44 ± 0.58
`0.40 ± 0.39
`
`16.2 ± 2.8
`13.9 ± 9.4
`13.5 ± 7.8
`18.8 ± 10.8
`
`1 Data are presented as mean ± SD for n = the number of guinea pigs indicated in parenthesis.
`
`was no significant difference between the two doses of
`MTPi tested with control or with atorvastatin group. Sim(cid:173)
`ilarly no significant changes were observed in any of the
`three regulatory hepatic enzymes involved, namely CYP7, .·
`ACAT and HMG-CoA Reductase (Table 5).
`
`Discussion
`In this study we were able to demonstrate that JTI-130,
`the MTPi tested has the potential to decrease the prime
`risk factors of cardiovascular disease, namely plasma TG
`and LDL-C concentrations in guinea pigs. The novelty of
`the drug
`tested
`is
`that
`there was no significant
`accumulation of lipid in the liver as seen in some other
`studies done with other MTPi (26,27].
`
`Drug treatment and hepatic lipids
`Previous studies evaluating MTPi have shown increase in
`the lipid content of the liver (7,26,27]. Chandler et al (7]
`treated Hep-G2 cells with CP-346086, another MTPi, for a
`period of two weeks. They reported that in addition to
`decreasing plasma TC, LDL-C, VLDL-C and TG values, this
`treatment also increased hepatic and intestinal TG when
`the MTPi was administered with food and when it was
`dosed away from meals, only hepatic TG were influenced.
`In contrast, the major finding of this study is that the MTPi
`tested did not lead to any fat accumulation in the liver as
`confirmed by no significant changes found in the hepatic
`lipid content as compared to their control counterparts.
`The main reason for these differences between MTPi could
`
`Page 5 of 8
`(page number not for citation purposes)
`
`5 of 8
`
`PENN EX. 2264
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`BMC Cardiovascular Disorders 2005, 5:30
`
`http://www.biomedcentral.com/14 71-2261 /5/30
`
`Table 5: Hepatic HMG-CoA reductase, ACAT and cholesterol 7a-hydroxylase activities (CYP7} guinea pigs fed a control diet, low dose
`MTPi (LOI}, high dose MTPi (HDI} or atorvastatin'
`
`Diets
`
`HMG-CoA Reductase
`
`ACAT
`
`Control
`LDI
`HDI
`Atorvastatin
`
`1.8 ± 0.8
`2.7 ± 0.8
`2.5 ± 1.4
`3.4 ± 2.9
`
`(pmol/min.mg)
`0.8 ± 0.4
`2.2 ± 0.9
`3.5 ± 2.4
`4.4 ± 3.1
`
`I Data are presented as mean ± SD for n = 3-7 guinea pigs per group.
`
`CYP7
`
`1.7 ± 1.8
`1.2 ± 0.5
`0.6 ± 0.7
`0.7 ± 0.4
`
`be that the main target of JTI-130 was the intestine.
`Because of this, we speculate that due to MTP inhibition,
`less TG were transferred to the chylomicron particle being
`packaged in the intestine. As a result a lower concentra(cid:173)
`tion of TG was taken up by the hepatocytes through the
`chylomicron remnant. Thus the VLDL particles secreted
`from the liver had lower concentrations ofTG molecules
`due to the major inhibitory effect of the MTPi in the intes(cid:173)
`tine. Because there were no significant changes in hepatic
`cholesterol concentrations, we did not find any significant
`differences in hepatic enzyme activities. Similar to the
`study by Conde et al. [9] in atorvastatin treated guinea
`pigs with 0.015% atorvastatin, there were no significant
`differences in hepatic cholesterol concentrations when
`compared with a control group. However, significant dif(cid:173)
`ferences in hepatic esterified cholesterol were observed
`when guinea pigs were treated with a higher dose of the
`statin (0.05%) (9).
`
`Drug treatment and plasma lipids and lipoproteins
`Abetalipoproteinemia, a genetic disorder characterized by
`low plasma cholesterol and TG levels, is caused by a func(cid:173)
`tional deficiency of MTP. Absence of lipid transfer activity
`in the microsomes of abetalipoproteinemia patients
`established its pivotal function in lipoprotein assem(cid:173)
`bly[ 1). This finding led to the suggestion that MTP inhibi(cid:173)
`tion could be used as a possible lipid lowering therapy.
`Further evidence was obtained from a cell culture study in
`which researchers (28] proved that MTP is limiting in the
`production of apo B containing lipoproteins. Another
`study [6) further confirmed this finding using hetero(cid:173)
`zygous MTP knockout mice which had 20% less plasma
`total cholesterol levels compared to wild type mice fed
`high fat diets; however, they did not find any significant
`differences in plasma TG concentrations. In our study, wfi
`have demonstrated that animals treated with MTPi had
`not only lower plasma TC and LDL-C but also significant
`reductions in plasma TG. It is possible that the VLDL par(cid:173)
`ticle secreted by the liver was more readily catabolized and
`therefore there was less conversion to LDL, which contrib(cid:173)
`uted to the hypocholesterolemic effects of the MTPL
`Conde et al. (9) demonstrated that there was a significant
`
`reduction in plasma TG in guinea pigs treated with atorv(cid:173)
`astatin when compared to controls. This was partly
`explained by lower secretion of VLDL particles and by
`increases in the LDL receptor [9), which could have con(cid:173)
`tributed to the faster removal ofVLDL particles. A similar
`mechanism may have taken place with the MTPi. By
`blocking MTP, JTI-130 reduced the secretion ofVLDL par(cid:173)
`ticles, and therefore, the formation ofLDL in the plasma.
`
`There was a dose response in CETP activity with ITT-130,
`and in addition, guinea pigs treated with atorvastatin
`exhibited decreased activity of this transfer protein. The
`main function of CETP is to contribute to the reverse cho(cid:173)
`lesterol pathway by transferring cholesteryl ester from
`HDL to the apo B containing lipoproteins [29]. However,
`this action prolongs the residence time of CE in LDL and
`increases the possibility of its deposition in the arterial
`wall. Thus lower CETP activity has been associated with
`decreased atherogenesis in animal studies [30). Therefore
`the lowering of CETP activity by drug treatment can be
`considered beneficial.
`
`Results from this study indicate that JTI-130 has the
`potential to reduce the primary risk factors for coronary
`heart disease. While these results are quite promising,
`more studies are needed to clarify the possibility of
`adverse effects including steatorhea, fat malabsorption
`and fat-soluble vitamin absorption. Although the lipid
`lowering effects were not as pronounced as
`those
`observed with atorvastain, the doses of MTPi used in the
`current study were lower than the doses of atorvastatin. It
`is possible that using JTI-130 in combination with statins
`could reduce the wide array of adverse effects associated
`with reductase inhibitors (31). This study also demon(cid:173)
`strates that MTP inhibitor which is mainly targeted
`towards the intestine may open a new avenue for treat(cid:173)
`ment of hyperlipidemic patients who are at high risk for
`cardiovascular diseases.
`
`List of abbreviations used
`ACAT: acyl CoA cholesteryl acyl transferase; AT: atorvasta(cid:173)
`tin; CETP: cholesterol ester transfer protein; CHD: coro-
`
`Page 6 of 8
`(page number not for citation purposes)
`
`6 of 8
`
`PENN EX. 2264
`CFAD V. UPENN
`IPR2015-01836
`
`

`
`BMC Cardiovascular Disorders 2005, 5:30
`
`http://www.biomedcentral.com/14 71-2261 /5/30
`
`nary heart disease; CYP7: cholesterol 7a-hydroxylase;
`HDi: high dose of the inhibitor; HDL-C: HDL cholesterol;
`HMG-CoA; 3-hydroxy-3methyl glutaryl Conezyme A;
`LCAT: lecithin cholesterol acyltransferase; LDi: low dose
`of the inhibitor; LDL-C: LDL cholesterol; MTP: micro(cid:173)
`somal transfer protein; MTPi: microsomal transfer protein
`inhibitor; NADP: nicotinamide adenenine dinucleotide
`phosphate; TG: triglycerides; VLDL: very low density
`lipoprotein.
`
`Competing interests
`Authors received funding from Akros Pharma Inc. (Princ(cid:173)
`eton, NJ) to carry out the studies presented in this
`manuscript.
`
`Authors' contributions
`DA did the assays, wrote the manuscript and participated
`in the interpretation of data; KLW: assisted in the assays
`for plasma lipids, CETP and LCAT; TLZ: assisted in the
`determination of ACAT activity and participated in data
`interpretation, SS: assisted in taking care of guinea pigs,
`isolation of microsomes and data interpretation; MVJ
`assisted in the determination of CYP7 and in data inter(cid:173)
`pretation and MLF designed the experiment, evaluated the
`results, interpreted the data and participated in manu(cid:173)
`script preparation.
`
`Acknowledgements
`These studies were supported by Akros Pharma Inc, Princeton, NJ
`
`4.
`
`5.
`
`References
`I. Hussain MM, Iqbal J, Anwar K, Rava P, Dai K: Microsomal triglyc(cid:173)
`eride transfer protein: a multifunctional protein. Front Biosci
`2003:500-506.
`2. National Cholesterol Education Program Expert Panel. Second report
`of the National Cholesterol Education Program (NCEP) Expert panel
`on Detection: Evaluation and Treatment of High Blood Cho(cid:173)
`lesterol in Adults; Adult Treatment Panel II. Circulation 1994,
`89: 1329-1445.
`3. Association AH: Heart and Stroke Statistical Update. American
`Heart Association: 200 I 200 I .
`ShepherdJ, Cobbe SM, Ford I, Isles CG, Lorimer AR, Macfarlane PW,
`McKillop JH, Packard CJ: Prevention of coronary heart disease
`with pravastatin in men with hypercholesterolemia. N Eng j
`Med 1995, 333:1301-1307.
`Sacks FM, Pfeffer MA, Moye LA, Rouleau JL, Rutherford JD, Cole TG,
`Brown L, Warnica JW, Arnold JMO, Wun CC, Davis BR, Braunwald
`E: The effect of pravastatin on coronary events after myocar(cid:173)
`dial infarction in patients with average cholesterol levels. N
`Eng) Med 1996, 335:1001-1009.
`Raabe M, Veniant MM, Sullivan MA, Zlot CH, Bjorkegren J, Nielson
`LB, Wong JS, Hamilton RL, Young SG: Analysis of the role of
`microsomal triglyceride transfer protein in the liver of tis(cid:173)
`sue-specific knockout mice. j C/in Invest 1999, 103(9):1287-1298.
`C_handler CE, Wilder DE, Pettini JL, Savoy YE, Petras SF, Chang G,
`Vincent J, Harwood HJ: CP-346086: an MTP inhibitor that low(cid:173)
`ers plasma cholesterol and triglycerides in experimental ani(cid:173)
`mals and in humans. j Upid Res 2003, 44:1887-1901.
`8. West KL, Fernandez ML: Guinea pigs as models to study the
`hypercholesterolemic effects of drugs. Cardiavasc Drug Rev
`2004, 22( I ):55-70.
`Conde K, Pineda G, Newton RS, Fernandez ML: Hypocholestero(cid:173)
`lemic effects of 3-hydroxy-3-methylglutaryl coenzyme a
`(HMG-CoA) reductase inhibitors in the guinea pig. Biochem
`Pharmacol 1999, 58:1209-1219.

`
`6.
`
`7.
`
`9.
`
`I 0. Fernandez ML: Guinea pigs as models for cholesterol and lipo(cid:173)
`protein metabolism. J Nutr 200 I, 131 (I): I 0-20.
`I I. Lin ECK, Fernandez ML, McNamara DJ: Dietary fat type and cho(cid:173)
`lesterol quantity interact to affect cholesterol metabolism in
`guinea pigs. j Nutr 1992, 122:2019-2029.
`12. Redgrave TG, Roberts DC, West CE: Separation of plasma lipo(cid:173)
`proteins by density-gradient ultracentrifugation. Anal Biochem
`J 1975, 65:42-49.
`13. Fernandez ML, Wilson TA, Conde K, Vegara-Jimenez M, Nicolosi RJ:
`Hamsters and guinea pigs differ in their plasma lipoprotein
`cholesterol distribution when fed diets varying in animal pro(cid:173)
`tein, soluble fiber or cholesterol content. J Nutr 1999,
`129: 1323-1333.
`14. Allain CC, Poon LC, Chan CS, Richard W, Fu PC: Enzymatic deter(cid:173)
`mination of total serum cholesterol. Clin Chem 1974, 20:47-475.
`15. Carr TP, Anderssen CJ, Rudel LL: Enzymatic determination of
`triglycerides, free cholesterol and cholesterol in tissue lipid
`extracts. C/in Biochem 1993, 26:39-42.
`16. Markwell MK, Haas S, Bieber LL, Tolbert NE: A modification of the
`Lowry procedure to simplify protein determination in mem(cid:173)
`brane and lipoprotein samples. Anal Biachem J 1978, 87:206-210.
`17. Homquist L, Carlson K, Carlson LA: Comparison between the
`use of isopropanol and tetramethylurea for the solublization
`and quantitation of human serum very
`low density
`apo

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