New Peroxisome Proliferator Activated Receptor Agonists - Potential Treatments for Atherogenic...

11
1. Introduction 2. PPAR family 3. ABT-335 4. K-877 5. Dual PPAR-a/g agonists 6. GW501516 7. MBX-8025 8. GFT-505 9. INT131 10. Conclusion 11. Expert opinion Review New peroxisome proliferator- activated receptor agonists: potential treatments for atherogenic dyslipidemia and non-alcoholic fatty liver disease Amirhossein Sahebkar, Gerard T Chew & Gerald F Watts Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital and Metabolic Research Centre, School of Medicine and Pharmacology, University of Western Australia Perth, Australia Introduction: Novel peroxisome proliferator-activated receptor (PPAR) modu- lators (selective PPAR modulators [SPPARMs]) and dual PPAR agonists may have an important role in the treatment of cardiometabolic disorders owing to lipid-modifying, insulin-sensitizing and anti-inflammatory effects. Areas covered: This review summarizes the efficacy of new PPAR agonists and SPPARMs that are under development for the treatment of atherogenic dyslipidemia and non-alcoholic fatty liver disease (NAFLD). Expert opinion: ABT-335 is a new formulation of fenofibrate that has been approved for concomitant use with statins. K-877, a SPPARM-a with encour- aging preliminary results in modulating atherogenic dyslipidemia, and INT131, a SPPARM-g with predominantly insulin-sensitizing actions, may also have favorable lipid-modifying effects. Although the development of dual PPAR-a/g agonists (glitazars) and the SPPARM-d GW501516 has been aban- doned because of safety issues, another SPPARM-d (MBX-8025) and a dual PPAR-a/d agonist (GFT-505) have shown promising efficacy in decreasing plasma triglyceride and increasing high-density lipoprotein cholesterol con- centrations, as well as improving insulin sensitivity and liver function. The beneficial effects of GFT-505 are complemented by preclinical findings that indicate reduction of hepatic fat accumulation, inflammation and fibrosis, making it a promising candidate for the treatment of NAFLD/nonalcoholic steatohepatitis (NASH). Long-term trials are required to test the efficacy and safety of these new PPAR agonists in reducing cardiovascular outcomes and treating NAFLD/NASH. Keywords: dyslipidemia, fatty liver, high-density lipoprotein, peroxisome proliferator-activated receptor, triglyceride Expert Opin. Pharmacother. [Early Online] 1. Introduction Atherogenic dyslipidemia is characterized by a triad of hypertriglyceridemia, low high-density lipoprotein cholesterol (HDL-C) and increased small dense low- density lipoprotein (LDL) particles [1]. It is the most common form of dyslipidemia in subjects with type 2 diabetes, metabolic syndrome and obesity, and is associated with an increased risk of atherosclerotic cardiovascular (CV) disease and coronary events beyond the effect of each individual component [2,3]. Atherogenic dyslipide- mia is also a constant feature of nonalcoholic fatty liver disease (NAFLD), being causally related to the formation of triglyceride (TG)-laden hepatocytes, known as hepatic steatosis [4]. 10.1517/14656566.2014.876992 © 2014 Informa UK, Ltd. ISSN 1465-6566, e-ISSN 1744-7666 1 All rights reserved: reproduction in whole or in part not permitted Expert Opin. Pharmacother. Downloaded from informahealthcare.com by University New South Wales on 01/22/14 For personal use only.

description

41

Transcript of New Peroxisome Proliferator Activated Receptor Agonists - Potential Treatments for Atherogenic...

  • 1. Introduction

    2. PPAR family

    3. ABT-335

    4. K-877

    5. Dual PPAR-a/g agonists

    6. GW501516

    7. MBX-8025

    8. GFT-505

    9. INT131

    10. Conclusion

    11. Expert opinion

    Review

    New peroxisome proliferator-activated receptor agonists:potential treatments foratherogenic dyslipidemia andnon-alcoholic fatty liver diseaseAmirhossein Sahebkar, Gerard T Chew & Gerald F Watts

    Lipid Disorders Clinic, Cardiovascular Medicine, Royal Perth Hospital and Metabolic Research

    Centre, School of Medicine and Pharmacology, University of Western Australia Perth, Australia

    Introduction: Novel peroxisome proliferator-activated receptor (PPAR) modu-

    lators (selective PPAR modulators [SPPARMs]) and dual PPAR agonists may

    have an important role in the treatment of cardiometabolic disorders owing

    to lipid-modifying, insulin-sensitizing and anti-inflammatory effects.

    Areas covered: This review summarizes the efficacy of new PPAR agonists and

    SPPARMs that are under development for the treatment of atherogenic

    dyslipidemia and non-alcoholic fatty liver disease (NAFLD).

    Expert opinion: ABT-335 is a new formulation of fenofibrate that has been

    approved for concomitant use with statins. K-877, a SPPARM-a with encour-aging preliminary results in modulating atherogenic dyslipidemia, and

    INT131, a SPPARM-g with predominantly insulin-sensitizing actions, may alsohave favorable lipid-modifying effects. Although the development of dual

    PPAR-a/g agonists (glitazars) and the SPPARM-d GW501516 has been aban-doned because of safety issues, another SPPARM-d (MBX-8025) and a dualPPAR-a/d agonist (GFT-505) have shown promising efficacy in decreasingplasma triglyceride and increasing high-density lipoprotein cholesterol con-

    centrations, as well as improving insulin sensitivity and liver function. The

    beneficial effects of GFT-505 are complemented by preclinical findings that

    indicate reduction of hepatic fat accumulation, inflammation and fibrosis,

    making it a promising candidate for the treatment of NAFLD/nonalcoholic

    steatohepatitis (NASH). Long-term trials are required to test the efficacy and

    safety of these new PPAR agonists in reducing cardiovascular outcomes and

    treating NAFLD/NASH.

    Keywords: dyslipidemia, fatty liver, high-density lipoprotein,

    peroxisome proliferator-activated receptor, triglyceride

    Expert Opin. Pharmacother. [Early Online]

    1. Introduction

    Atherogenic dyslipidemia is characterized by a triad of hypertriglyceridemia, lowhigh-density lipoprotein cholesterol (HDL-C) and increased small dense low-density lipoprotein (LDL) particles [1]. It is the most common form of dyslipidemiain subjects with type 2 diabetes, metabolic syndrome and obesity, and is associatedwith an increased risk of atherosclerotic cardiovascular (CV) disease and coronaryevents beyond the effect of each individual component [2,3]. Atherogenic dyslipide-mia is also a constant feature of nonalcoholic fatty liver disease (NAFLD), beingcausally related to the formation of triglyceride (TG)-laden hepatocytes, known ashepatic steatosis [4].

    10.1517/14656566.2014.876992 2014 Informa UK, Ltd. ISSN 1465-6566, e-ISSN 1744-7666 1All rights reserved: reproduction in whole or in part not permitted

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • Pharmacotherapy for atherogenic dyslipidemia ischallenging, since its individual components, such as elevatedTG and low HDL-C, are not readily correctable by statins.However, agonists of peroxisome proliferator-activated recep-tors (PPARs) have shown promise in the management ofatherogenic dyslipidemia and its associated comorbidities, inparticular NAFLD -- a common condition for which there isno approved specific drug treatment [5,6]. Therapies forNAFLD should ideally not only reverse the accumulation ofTG in hepatocytes but should also effectively suppress hepaticinflammation, thereby preventing progression of simple steato-sis to nonalcoholic steatohepatitis (NASH), fibrosis andcirrhosis. Current treatment modalities, such as statins, fenofi-brate and ezetimibe, can target the risk factors of NASH, suchas dyslipidemia, insulin resistance, oxidative stress and inflam-mation [7,8]. The adverse effects of NAFLD are not confined tothe hepatic tissue, since this condition is also strongly associ-ated with increased CV risk. Whereas the underlying mecha-nism for this association is not fully understood, it appearsthat the chronic inflammation which accompanies NAFLDcan further contribute to atherogenesis [9]. There is also evi-dence from a post-hoc analysis of the Greek Atorvastatin andCoronary Heart Disease Evaluation Study (GREACE), indi-cating a greater reduction of CV event rate by statin therapyin patients with abnormal liver enzymes due to NAFLD com-pared with patients with normal liver enzymes. These closeassociations between NAFLD and CV risk, along with the pre-dominance of CV disease as the most important cause of mor-tality among NAFLD patients, further highlight the necessityfor proper management of NAFLD [10].With respect to vascular risk, findings from large-scale Feno-

    fibrate Intervention and Event Lowering in Diabetes (FIELD)and Action to Control Cardiovascular Risk in Diabetes(ACCORD) trials have shown that treatment with PPAR-a

    agonists (fibrates) reduces the risk of CV events in subgroupsof diabetic subjects with atherogenic dyslipidemia, as bothmonotherapy and combination therapy with statins [11,12].Regarding CV effects of PPAR-g agonists, thiazolidinediones(TZDs), the major concern has been an increased risk ofcongestive heart failure [13], although this is not associatedwith an increased mortality [14,15]. Findings of the large-scaleProspective Pioglitazone Clinical Trial In MacrovascularEvents (PROactive) trial in diabetic patients with a history ofCV disease indicated that pioglitazone significantly reducesthe risk of major adverse CV events, as well as the risk of secondmyocardial infarction and stroke in respective subgroups ofpatients [16-18].

    The purpose of this review is to provide a summary ofclinical findings on the efficacy of new PPAR agonists andselective PPAR modulators (SPPARMs) that are under devel-opment for the treatment of atherogenic dyslipidemia andNAFLD (Table 1).

    2. PPAR family

    PPARs are a superfamily of ligand-activated nuclear receptorsthat regulate energy balance by influencing the metabolism oflipids and glucose. Three PPAR isotypes have been identified,including PPAR-a (NR1C1), PPAR-g (NR1C3) andPPAR-b/d (NR1C2). PPARs are transcription factors thatpromote their biological effects by altering the gene expres-sion. The activated PPAR binds to the retinoid X receptorto form a heterodimeric complex that then binds to conservedsequences of DNA, known as PPAR response elements,located in the promoter regions. This interaction results inthe upregulation or downregulation of a cascade of down-stream genes (Figure 1) [19-21].

    PPAR-a is most frequently expressed in metabolicallyactive tissues such as liver, kidney, muscle, myocardium andbrown fat. Synthetic ligands of PPAR-a, such as fibrates, arewell-established lipid-modifying agents that act throughenhancing TG-rich lipoprotein (TRL) lipolysis, hepatic fattyacid uptake and subsequent b-oxidation, reducing TRLbiosynthesis and increasing HDL production and reverse cho-lesterol transport [22,23]. Moreover, PPAR-a agonists downre-gulate the NF-kB pathway and reduce hepatic inflammation,and also upregulate the expression of both adiponectin andadiponectin receptors [24,25], which might be especially impor-tant for preventing progression of simple steatosis to NASHthrough enhancement of insulin sensitivity and reduction ofhepatic fat [26].

    PPAR-g is the most abundant PPAR subtype in whiteadipose tissue, and its main function is the regulation of fattyacid uptake, insulin sensitivity and glucose homeostasis.TZDs, also referred to as glitazones, are agents that activatePPAR-g and promote their effects through improving periph-eral tissue insulin sensitivity and reducing fatty acid flux to theliver. Further, PPAR-g activation enhances adiponectinexpression, which further improves insulin sensitivity and

    Article highlights.

    . Atherogenic dyslipidemia is characterized byhypertriglyceridemia, low high-density lipoproteincholesterol (HDL-C), and increased small denselow-density lipoprotein particles.

    . The efficacy of statins in correcting hypertriglyceridemiaand low HDL-C is limited, thereby rendering manytreated patients at a high residual cardiovascular risk.

    . Selective peroxisome proliferator-activated receptormodulators, K-877 (a), MBX-8025 (d), INT131 (g), andthe peroxisome proliferator-activated receptor-a/dagonist GFT-505 have demonstrated efficacy inimproving several components of atherogenicdyslipidemia.

    . MBX-8025 and GFT-505 have also been shown toimprove liver function and glucose homeostasis, andGFT-505 has been reported to reduce hepatic fataccumulation in rodent models.

    This box summarizes key points contained in the article.

    A. Sahebkar et al.

    2 Expert Opin. Pharmacother. (2014) 15 (4)

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • hepatic fatty acid b-oxidation and also attenuates hepaticinflammation, stellate cell proliferation and adipogenesis[27-29]. Although the PPAR-g agonists pioglitazone and rosigli-tazone have been shown to exert beneficial effects on bothmetabolic (improvement in insulin sensitivity and glucose tol-erance) and histological parameters (reduction of hepatic fatand inflammation) in patients with biopsy-provenNASH [30-36], clinical use of these agents has been limitedby off-target adverse effects, such as fluid retention and weightgain, worsening cardiac failure and an increased risk of boneloss and fractures [37-39]. Although the use of rosiglitazone ishighly restricted in Europe and United States, current Guide-line of the American Association for the Study of Liver Dis-eases still recommends the use of pioglitazone in patientswith biopsy-proven NASH [40]. These treatments have mixedbeneficial as well as less favorable effects on fasting and post-prandial lipid concentrations and subfractions, with alsoagent-specific differences in lipid effects between pioglitazoneand rosiglitazone [41].

    PPAR-b/d (or PPAR-d) is ubiquitously distributed in dif-ferent tissues, and its physiological and pharmacologicalactions are less clear. Although there are no synthetic ligandsof PPAR-d currently in clinical use, preliminary data fromanimal and clinical studies indicate that PPAR-d activationhas several favorable metabolic effects, including enhance-ment of fatty acid b-oxidation and lipid catabolism anddecrease in hepatic insulin resistance and inflammation [42].

    The development of SPPARMs is a useful strategy fordecoupling the beneficial metabolic effects of PPAR agonistsfrom their unwanted adverse effects. PPARs undergo differentstructural conformations on interaction with different ligands,and each ligand-receptor conformation has a specific bindingaffinity for coactivators, corepressors and transcription factors,leading to different patterns of gene expression modulation(Figure 2) [43,44]. Therefore, several SPPARMs a and d, anddual SPPARMs a/d and a/g have been developed with theaim of potentiating the metabolic effects associated with therespective PPAR subtypes, while minimizing their off-targetadverse effects. However, preliminary studies of these newagents have shown variable clinical effects as detailed in thefollowing sections.

    3. ABT-335

    ABT-335, the first fibrate approved by the FDA for combineduse with statins, was developed as a delayed-release formula-tion with minimal pharmacokinetic interaction with statins.ABT-335 is the choline salt of fenofibric acid and does notrequire hepatic first-pass metabolism for activation, therebyreducing its potential for interaction with the hepatic glucur-onidation enzymes involved in statin metabolism [45].

    The efficacy and safety of ABT-335 has been confirmed inthree Phase III studies in subjects with mixed dyslipidemia(TG 1.69 mmol/l, HDL-C < 1.03 mmol/l and LDL-C 3.36 mmol/l), where ABT-335 was given as monotherapyTa

    ble

    1.Asummary

    ofnew

    PPARagonists

    andSPPARMsin

    development.

    Drugclass

    Target

    PPAR

    subtype

    %changein

    lipids

    %change

    inHOMA-IR

    Reductionof

    hepaticinjury

    biomarkers

    Severe

    adverseevents

    Testedin

    Phase

    IIItrials

    ab/d

    gTG

    HDL-C

    LDL-C

    TC

    Non-HDL-C

    ABT-335

    32

    --33%#

    15

    --20%"

    4--6%

    #12

    --13%#

    15

    --18%#

    NR

    --

    Yes

    K-877

    4

    --14%

    #NR

    NR

    NR

    NR

    NR

    --

    No

    Aleglitazar

    29

    --54%#

    20

    --56%"

    16

    --47%

    #NR

    NR

    64

    --96%

    -Bonefractures,

    gastrointestinalbleeding

    andheartfailure

    Yes

    MBX-8025

    28%#

    9--12%

    "18

    --22%

    #14

    --16%#

    18

    --22%#

    1.4

    --21%

    GGT/ALP

    -No

    GW501516

    9

    --31%

    #7

    --17%

    "23%

    #--4%

    "20%#-

    -5%

    "NR

    22%

    -Increasedrisk

    ofcancer

    No

    GFT-505

    17

    --25%#

    8--9%

    "0

    --13%#

    0--9%

    #1

    --13%#

    4--31%

    AST/ALT/GGT/ALP

    -No

    INT131

    NR

    NR

    NR

    NR

    NR

    NR

    --

    No

    ALP:Alkalinephosphatase;ALT:Alanineaminotransferase;AST:Aspartate

    aminotransferase;GGT:g-glutamyltransferase;HDL-C:High-density

    lipoprotein

    cholesterol;HOMA-IR:Homeostaticmodelassessment-insulin

    resistance;LD

    L-C:Low-density

    lipoprotein

    cholesterol;NR:Notreportedin

    percentageofchange;PPAR:Peroxisomeproliferator-activatedreceptor;SPPARM:Selectiveperoxisomeproliferator-activatedreceptor

    modulator;TC:Totalcholesterol;TG:Triglyceride.

    New PPAR agonists: potential treatments for atherogenic dyslipidemia and NAFLD

    Expert Opin. Pharmacother. (2014) 15(4) 3

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • or in combination with statin [46-48]. The combination ofABT-335 with statin was associated with a greater decreasein TG and increase in HDL-C compared with statin mono-therapy (Figures 3 and 4) and greater LDL-C reduction com-pared with ABT-335 monotherapy. In addition, subjectsreceiving combination therapy had significantly greater reduc-tions in non-HDL-C and very low-density lipoprotein choles-terol (VLDL-C). A 2-year extension study in subjects whocompleted either of the abovementioned trials showed thatthe lipid-modifying effects of the ABT-335/statin combina-tion were sustained (46% decrease in TG, 17% increase inHDL-C, 40% decrease in LDL-C). Moreover, long-termmonitoring did not indicate that ABT-335 treatment wasassociated with any increase in mortality or drug-related seri-ous adverse events, including rhabdomyolysis, and drug dis-continuation was low (2.9%). A post-hoc pooled analysis ofpatients with mixed dyslipidemia and type 2 diabetes in theabovementioned trials also confirmed the efficacy and safetyof the ABT-335/statin combination in this group. Interest-ingly, the percentage of subjects achieving optimal levels ofTG, HDL-C, LDL-C, non-HDL-C and apolipoprotein B(apoB) simultaneously was fivefold higher with combinationtherapy than with statin monotherapy [46-48].

    4. K-877

    K-877 is a SPPARM-a that can activate PPAR-a with muchgreater potency than fenofibrate (EC50 = 1 vs 14,000 --22,400 nM for fenofibrate) [49]. In a Phase II study in subjects

    with atherogenic dyslipidemia, K-877 improved both fasting(decrease in TG; increase in HDL-C) and post-prandial(decrease in TG, remnant lipoprotein cholesterol andapoB-48) lipid concentrations in a dose-dependent manner,with greater lipid changes than those achieved with fenofibrate.K-877 also improved the quality of lipoprotein subfractions bydecreasing the proportion of large VLDL and small LDLparticles and by increasing small HDL particles [50]. Thesefindings confirm the preclinical findings of K-877slipid-lowering and anti-atherosclerotic effects in LDL-receptorknockout mice [49]. Both animal and clinical studies have alsoshown that K-877 increases fibroblast growth factor-21 -- aregulator of glucose and lipid metabolism [49].

    5. Dual PPAR-a/g agonists

    Dual PPAR-a/g agonists, also referred to as glitazars, weredeveloped with the anticipation of simultaneously achieving,with a single drug, the TG-lowering and HDL-raising effectsof fibrates, as well as the insulin-sensitizing and anti-hyperglycemic effects of TZDs. Such a combination of effectswould be ideal for the treatment of type 2 diabetes, metabolicsyndrome and NAFLD, which share atherogenic dyslipidemiaand insulin resistance as common features. Increased lipidcatabolism due to PPAR-a activation was also expected tooffset the weight gain associated with TZD use.

    Muraglitazar and tesaglitazar were the first dual PPAR-a/gagonists tested in clinical trials. Although early trial findingswere promising and indicated improvements in both lipidprofile and insulin sensitivity [51,52], safety evaluations inPhase III trials revealed an increased risk of heart failurewith muraglitazar [53], and elevation of serum creatininewith tesaglitazar [54]. This led to withdrawal of these drugsfrom ongoing studies and discontinuation of their furtherresearch and development.

    Further attempts focused on developing a dual PPAR-a/gagonist with balanced affinity for both receptors, and aleglita-zar emerged as a balanced PPAR-a/g agonist with promisingeffects in preclinical and Phase I/II studies [55,56]. However, arecent Phase III trial (AleCardio; NCT01042769) investigat-ing its safety and efficacy in patients with type 2 diabetes andacute coronary syndrome was halted due to an increase inbone fractures, heart failure and gastrointestinal bleeding inthose receiving aleglitazar. This led to other simultaneousPhase III aleglitazar trials, including the large-scale AlePreventstudy, also being stopped.

    6. GW501516

    GW501516 is a potent SPPARM-d (EC50 = 1 nM). Two earlyPhase I trials in healthy subjects demonstrated its efficacy inreducing TG, LDL-C, apoB and insulin levels and improvingHDL-C and insulin sensitivity [57,58], with one of the trials alsofinding that GW501516 significantly reduced hepatic fatcontent by 20% [58]. A later study confirmed GW501516s

    RA

    Protein

    mRNA

    Transcription

    DNA

    PPAR/RXRheterodimer

    Effects on lipidmetabolism and

    glycaemic control

    PPAR activator

    Figure 1. PPAR activation and its regulatory effect on gene

    transcription, protein synthesis and cellular function are

    shown.Reproduced with permission from [8] (Rightslink license code:

    3246371243629).

    PPAR: Peroxisome proliferator-activated receptor; RA: Retinoic acid;

    RXR: Retinoid X receptor.

    A. Sahebkar et al.

    4 Expert Opin. Pharmacother. (2014) 15 (4)

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • lipid-modifying effects in dyslipidemic subjects with abdominalobesity and also provided evidence of its effects in modulatinglipoprotein kinetics (increase in catabolism of VLDL and pro-duction of apoA-I and apoA-II; decrease in production ofapoC-III and LDL-apoB). GW501516 was also associatedwith a decrease in cholesteryl ester transfer protein activity buthad no effect on insulin resistance [59]. Finally, Olson et al.reported that GW501516 treatment was associated with signif-icant improvements in multiple lipid profile components (TG,LDL-C,HDL-C, free fatty acids and apoB, apoA-I and apoA-II)and a shift in LDL particle size from small dense to larger, lessdense particles [60]. Despite these promising early results, the fur-ther investigation and development of GW501516 was discon-tinued after observations in animal studies of its associationwith the rapid induction of cancers in several organs (liver,stomach, tongue, skin, bladder, ovaries, womb and testes).

    7. MBX-8025

    The efficacy of MBX-8025, a potent SPPARM-d(EC50 = 2 nM), was demonstrated in a proof-of-conceptstudy in overweight subjects with mixed dyslipidemia, whereMBX-8025, given asmonotherapy or in combination with ator-vastatin, resulted in significant reductions in plasma LDL-C(18 -- 43%), apoB-100 (20 -- 38%), non-HDL-C (18 -- 41%),TG (26 -- 30%) and free fatty acids (16 -- 28%). MBX-8025seffects in lowering LDL-C and apoB-100 were enhanced whencombined with atorvastatin but did not exceed that of ator-vastatin monotherapy. However, the MBX-8025/atorvastatin

    combination was more efficacious in lowering TG and raisingHDL-C than atorvastatin alone, but not MBX-8025 alone [61].Further, LDL particle size distribution analysis revealed thatMBX-8025 treatment results in a reduction in small and verysmall LDL particles and an increase in large LDL particles. Itseffect on HDL elevation was also confined to the small HDLparticles. These changes were accompanied by a concomitantreduction in large VLDL particles and increase in LDL peakdiameter, and reversal of LDL pattern B (characterized by a pre-ponderance of small LDL particles) in > 90% of subjects. Theseimprovements in the lipoprotein subfractions were greaterthan those seen with atorvastatin treatment [62]. Finally,MBX-8025 also showed beneficial effects in improving insulinresistance and plasma markers of liver function [61].

    8. GFT-505

    GFT-505 is a dual PPAR-a/d agonist, with preferential a(EC50 = 6 nM) and complementary d (EC50 = 47 nM) recep-tor agonistic activity. It is a compound targeted at the liver,where it is converted to its main active metabolite,GFT-1007, in a dose-dependent manner. GFT-505 is themost widely studied SPPARM, and it is the agent with themost promising results in clinical trials to support its furtherdevelopment [63-65].

    The lipid-modifying efficacy of GFT-505 has beenconfirmed in healthy subjects [63], as well as in patients withtype 2 diabetes (NCT01261494), combined abdominalobesity and mixed dyslipidemia [64], combined abdominalobesity and pre-diabetes [64], atherogenic dyslipidemia(NCT01271751) and insulin resistance [65]. The lipid effectsof GFT-505 include decreases in TG (34% in patients withtype 2 diabetes) (NCT01261494), non-HDL-C and total cho-lesterol (9% in patients with insulin resistance, and patientswith abdominal obesity and pre-diabetes), LDL-C (13% inpatients with insulin resistance) [65], apoB (14% in patientswith insulin resistance) [65] and apoC-III (20% in patientswith abdominal obesity and pre-diabetes) [64], as well asincreases in HDL-C (15% in patients with type 2 diabetes)(NCT01261494), apoA-I (6% in patients with abdominalobesity and mixed dyslipidemia and in patients with athero-genic dyslipidemia) ([64], NCT01271751) and apoA-II (18%in patients with abdominal obesity and pre-diabetes) [64].A Phase I study in healthy subjects also showed thatGFT-505 decreases post-prandial free fatty acid levels [63].

    GFT-505 has also been reported to have favorable effects onglucose homeostasis. In patients with abdominal obesity andpre-diabetes, GFT-505 increased insulin sensitivity anddecreased fasting plasma glucose, C-peptide, insulin andfructosamine levels, but this was not seen in patients withabdominal obesity and mixed dyslipidemia [64]. The insulin-sensitizing effects of GFT-505 were demonstrated in two latertrials in treatment-nave subjects with newly diagnosedtype 2 diabetes (decrease in fasting glucose and HbA1c levels;decrease in 2-h post-challenge glucose level and glycemic area

    SPPARMs

    PPAR

    Ligandreceptordistinct and specificconformations

    Selective cofactorrecruitment

    Common and selective biological responses

    Figure 2. Selective modulation of peroxisome proliferator-

    activated receptor activity is shown.PPAR: Peroxisome proliferator-activated receptor; SPPARMs: Selective PPAR

    modulators.

    New PPAR agonists: potential treatments for atherogenic dyslipidemia and NAFLD

    Expert Opin. Pharmacother. (2014) 15(4) 5

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • under the curve following oral glucose tolerance test)(NCT01261494) and in insulin-resistant subjects (decrease inhepatic glucose production; improvements in both hepaticand peripheral insulin sensitivity, measured by hyperinsulinemic-euglycemic clamp) [65].In addition, GFT-505 may have potential to prevent the

    development and progression of NAFLD/NASH by reducinghepatic fat content [66] and by decreasing biomarkers ofhepatic inflammation and injury ([64,65], NCT01271751,NCT01261494). Preclinical studies in rodent models ofNAFLD/NASH (Western diet-fed human apo-E2 transgenicmice; methionine- and choline-deficient diet-fed db/db miceand CCl4-induced fibrotic rats) consistently demonstratedthat GFT-505 treatment decreases hepatic steatosis, inflam-mation and fibrosis, decreases liver dysfunction biomarkersand suppresses proinflammatory and profibrotic gene expres-sion, being effective in both the prevention and treatment ofhepatic fibrosis [66]. Interestingly, these same effects werereplicated in apo-E2-KI/PPAR-a-KO mice, suggesting aPPAR-a-independent hepatoprotective mechanism [66].In clinical studies, GFT-505 treatment has been reported

    to decrease a range of biomarkers of hepatic dysfunction,including alanine aminotransferase (20% in insulin-resistantsubjects) [65], alkaline phosphatase (24% in patients withabdominal obesity and pre-diabetes) [64] and g-glutamyl

    transferase (30% in insulin-resistant subjects) [65], withsimilar findings seen in dyslipidemic (NCT01271751),pre-diabetic [64], insulin-resistant [65] and diabetic patients(NCT01261494). Together with its unique pharmacokineticprofile (hepatic accumulation and extensive enterohepaticrecycling), these beneficial effects of GFT-505 support itspotential as a hepatoprotective agent. These promising find-ings, along with positive data from other SPPARM-d studies,have prompted a Phase IIb trial (NCT01694849) ofGFT-505 in patients with biopsy-proven NASH, the resultsof which are awaited with interest.

    9. INT131

    In trying to decouple the beneficial from the adverse effects offull PPAR-g agonists, there has been interest in developingSPPARMs that target alternate regions of the PPAR-g receptor,resulting in limited recruitment of coregulators and restrictedbut more favorable metabolic effects [67]. Of the SPPARMs-gstudied, only INIT131 is currently under clinical developmentfor the treatment of insulin resistance. Preclinical and earlyPhase II clinical studies showed that INIT131 improves insulinsensitivity and glycemia without some of the side effects of gli-tazones, such as fluid retention, but it did increase body weightwhen given in higher, more efficacious dose [68]. Importantly,

    Statin alone

    TG re

    duct

    ion

    (%)

    50

    45

    40

    35

    3025

    05

    10

    15

    20

    Simvastatin (20 mg) ABT-335 (135 mg)Atorvastatin (20 mg) ABT-335 (135 mg)

    Atorvastatin (40 mg) ABT-335 (135 mg)

    Rosuvastatin (10 mg) ABT-335 (135 mg)

    Rosuvastatin (20 mg) ABT-335 (135 mg)

    Simvastatin (40 mg) ABT-335 (135 mg)

    Statin + ABT-335

    Figure 3. Incremental reductions in triglycerides (TGs) following addition of ABT-335 (135 mg) to different doses of statins

    are shown. Red bars represent percentage reduction in TGs or percentage elevation in high-density lipoprotein cholesterol

    incremental to the changes achieved by statin monotherapy. All differences between statin monotherapy and

    statin + ABT-335 were statistically significant [32-34].

    A. Sahebkar et al.

    6 Expert Opin. Pharmacother. (2014) 15 (4)

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • potentially beneficial dose-dependent lipid effects (decrease infree fatty acids, increase in HDL-C, trend to decrease in TG)were reported [69].

    10. Conclusion

    Atherogenic dyslipidemia contributes significantly to theresidual CV risk in statin-treated patients, even those at opti-mal LDL-C levels, and it is causally related to NAFLD. Theefficacy of fibrates in decreasing TG and raising HDL-C levelsis established, but progress continues to be made in the devel-opment of PPAR agonists with greater receptor specificity,higher efficacy, and improved safety, particularly when usedin combination with statins.

    ABT-335 is an improved formulation of fenofibrate that hasbeen approved for combination therapy with statins.K-877 and MBX-8025 are SPPARMs (a and d, respectively)with enhanced lipid-modifying effects. INT131 (SPPARM-g)improves insulin sensitivity and glycemia but might also havefavorable lipid effects relevant to the treatment of atherogenicdyslipidemia. Despite considerable lipid-modifying efficacy,the development of dual PPAR-a/g agonists (muraglitazar,tesaglitazar and aleglitazar) and the SPPARM-d GW501516has been discontinued due to the emergence of associatedsignificant adverse events. Finally, animal and clinical studies

    of the dual PPAR-a/d agonist GFT-505 show great promiseof its efficacy in correcting dyslipidemia, enhancing insulinsensitivity and reducing hepatic fat accumulation andinflammation.

    11. Expert opinion

    Atherogenic dyslipidemia is the predominant lipid disorder inpatients with metabolic syndrome and type 2 diabetes, whoare at high risk of CV disease. It is characterized by increasedplasma levels of TG and TRL remnants, predominance ofsmall dense LDL and decreased levels of HDL-C [1]. Statinsare the mainstay of pharmacotherapy for dyslipidemia, andtheir efficacy in reducing CV outcomes has been establishedin both primary and secondary prevention trials [70]. How-ever, a significant proportion of statin-treated subjects, eventhose at target LDL-C levels on high-dose therapy, have ahigh residual risk of CV disease [71]. Further reductions inLDL-C may be achieved by adding ezetimibe to statin, butclinical trial evidence for the specific CV benefits of this com-bination is scarce. The Study of Heart and Renal Protectionshowed a reduction in the incidence of major atheroscleroticevents following combination therapy with simvastatin andezetimibe (vs placebo) in patients with advanced chronic kid-ney disease [72]; however, further evidence from outcome trials

    Statin aloneStatin + ABT-335

    HD

    L-C

    elev

    atio

    n (%

    )

    25

    20

    15

    10

    5

    0

    Simvastatin (20 mg) ABT-335 (135 mg)Atorvastatin (20 mg) ABT-335 (135 mg)

    Atorvastatin (40 mg) ABT-335 (135 mg)

    Rosuvastatin (10 mg) ABT-335 (135 mg)

    Rosuvastatin (20 mg) ABT-335 (135 mg)

    Simvastatin (40 mg) ABT-335 (135 mg)

    Figure 4. Incremental elevations in high-density lipoprotein cholesterol (HDL-C) following addition of ABT-335 (135 mg) to

    different doses of statins are shown. Red bars represent percentage reduction in triglycerides or percentage elevation in

    HDL-C incremental to the changes achieved by statin monotherapy. All differences between statin monotherapy and

    statin + ABT-335 were statistically significant [32-34].

    New PPAR agonists: potential treatments for atherogenic dyslipidemia and NAFLD

    Expert Opin. Pharmacother. (2014) 15(4) 7

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • are required to assess the impact of such a combination versusstatin monotherapy and also in other patient populations.Although according to the recent ACC/AHA guidelines thereis still inadequate clinical trial outcome evidence to supportthe use of non-statin drugs in the prevention of atheroscleroticCV disease [73], there are many agents currently in variousstages of development for the treatment of dyslipidemia asoutlined above. These non-statin agents may be particularlyuseful for patients intolerant or refractory to statins and canbe used as an add-on to statins to enhance TG reductionand HDL-C elevation. Since residual CV risk in statin-treatedsubjects may be related to coexistent atherogenic dyslipide-mia, adjunctive use of lipid-modifying agents with specificeffects on TG and HDL-C may also be indicated. Fibrateshave been shown to be especially efficacious in reducing CVoutcomes in patients with atherogenic dyslipidemia. Hence,newer potent PPAR agonists and SPPARMs are expected tohave comparable promising effects in reducing CV risk. How-ever, this needs to be demonstrated in clinical outcome trials.The value of adding niacin to statins in patients with

    optimal LDL-C lowering but high residual CV risk is notsupported by recent clinical trials (AIM-HIGH andHPS2-THRIVE) [74,75], and the use of high dose n-3 polyun-saturated fatty acids (given as pure eicosapentanoic acid) iscurrently being investigated in the REDUCE-IT trial(NCT01492361). Clinical outcome studies have shown thatfenofibrate, an agonist of PPAR-a, can significantly reduceCV events in patients with atherogenic dyslipidemia [11,12,76].However, to improve the beneficial lipid-modifying effects ofPPAR activation, while minimizing off-target adverse effects,newer agents are being developed that have less potential foradverse pharmacokinetic interactions with statins (e.g.,ABT-335), greater specificity for PPAR subtypes (SPPARMs)and balanced activation of different PPAR subtypes (dualagonists).

    Early studies of the SPPARMs K-877 (a) [36],MBX-8025 (d) [47], INT131 (g) [54,55] and the PPAR-a/d ago-nist GFT-505 ([49-51], NCT01271751) have demonstratedefficacy in improving several components of atherogenicdyslipidemia. In addition, MBX-8025 and GFT-505 havealso been shown to improve liver function ([47,50,51],NCT01271751), with the latter demonstrating beneficialeffects in reducing hepatic steatosis, inflammation and fibrosisin animal studies [52]. These findings suggest a possible role forthese agents in the treatment of NAFLD/NASH -- a conditioncausally related to atherogenic dyslipidemia, for which there isno specific pharmacotherapy at present. The results of anongoing Phase IIb trial of long-term GFT-505 treatment inpatients with NASH (NCT01694849) are keenly awaited.

    However, despite the aforementioned promising data andresults, the development of other similar agents with poten-tially beneficial lipid-modifying and metabolic effects, suchas GW501516 (SPPARM-d) and the dual PPAR-a/g agonists(glitazars), has been abandoned because of associated seriousoff-target adverse effects. This emphasizes the importance oflonger-term studies to evaluate drug safety and also therequirement for randomized controlled clinical trial data todemonstrate not only the efficacy of these new agents inimproving lipid and metabolic end points but also in improv-ing meaningful clinical outcomes.

    Declaration of interest

    GF Watts has received honoraria for lectures and commentar-ies from Genfit, Pfizer, AstraZeneca, Merck Sharp & Dohme,Novartis, Amgen, Abbott and Sanofi-Aventis. With respect tothe content of the article, GT Chew has previously receivedresearch grant funding from Pfizer and GlaxoSmithKlineand has participated in research sponsored by LaboratoiresFournier. A Sahebkar has no conflict of interest.

    BibliographyPapers of special note have been highlighted as

    either of interest () or of considerable interest() to readers.1. Rizzo M, Berneis K. Lipid triad or

    atherogenic lipoprotein phenotype: a role

    in cardiovascular prevention?

    J Atheroscler Thromb 2005;12:237-9

    2. Tenenbaum A, Fisman EZ, Motro M,

    Adler Y. Atherogenic dyslipidemia in

    metabolic syndrome and type 2 diabetes:

    therapeutic options beyond statins.

    Cardiovasc Diabetol 2006;5:20

    3. Ghandehari H, Kamal-Bahl S,

    Wong ND. Prevalence and extent of

    dyslipidemia and recommended lipid

    levels in US adults with and without

    cardiovascular comorbidities: the

    National Health and Nutrition

    Examination Survey 2003--2004.

    Am Heart J 2008;156(1):112-19

    4. Makadia SS, Blaha M, Keenan T, et al.

    Relation of hepatic steatosis to

    atherogenic dyslipidemia. Am J Cardiol

    2013;112(10):1599-604

    5. Wilding JP. PPAR agonists for the

    treatment of cardiovascular disease in

    patients with diabetes.

    Diabetes Obes Metab 2012;14:973-82

    6. Kallwitz ER, McLachlan A, Cotler SJ. Role

    of peroxisome proliferators-activated

    receptors in the pathogenesis and treatment

    of nonalcoholic fatty liver disease.

    World J Gastroenterol 2008;14:22-8

    7. Filippatos TD, Elisaf MS. Role of

    ezetimibe in non-alcoholic fatty liver

    disease. World J Hepatol 2011;3:265-7

    8. Kostapanos MS, Kei A, Elisaf MS.

    Current role of fenofibrate in the

    prevention and management of non-

    alcoholic fatty liver disease.

    World J Hepatol 2013;5:470-8

    9. Katsiki N, Athyros VG, Karagiannis A,

    Mikhailidis DP. Hyperuricaemia and

    non-alcoholic fatty liver disease

    (NAFLD): a relationship with

    implications for vascular risk?

    Curr Vasc Pharmacol 2011;9:698-705

    10. Athyros VG, Tziomalos K, Gossios TD,

    et al.; GREACE Study Collaborative

    Group. Safety and efficacy of long-term

    statin treatment for cardiovascular events

    in patients with coronary heart disease

    and abnormal liver tests in the Greek

    Atorvastatin and Coronary Heart Disease

    A. Sahebkar et al.

    8 Expert Opin. Pharmacother. (2014) 15 (4)

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • Evaluation (GREACE) Study: a post-hoc

    analysis. Lancet 2010;376:1916-22

    11. Bruckert E, Labreuche J, Deplanque D,

    et al. Fibrates effect on cardiovascular

    risk is greater in patients with high

    triglyceride levels or atherogenic

    dyslipidemia profile: a systematic review

    and meta-analysis.

    J Cardiovasc Pharmacol 2011;57:267-72

    12. Lee M, Saver JL, Towfighi A, et al.

    Efficacy of fibrates for cardiovascular risk

    reduction in persons with atherogenic

    dyslipidemia: a meta-analysis.

    Atherosclerosis 2011;217:492-8

    13. Lago RM, Singh PP, Nesto RW.

    Congestive heart failure and

    cardiovascular death in patients with

    prediabetes and type 2 diabetes given

    thiazolidinediones: a meta-analysis of

    randomised clinical trials. Lancet

    2007;370:1129-36

    14. Erdmann E, Charbonnel B, Wilcox RG,

    et al. Pioglitazone use and heart failure in

    patients with type 2 diabetes and

    preexisting cardiovascular disease: data

    from the PROactive study (PROactive 08).

    Diabetes Care 2007;30:2773-8

    15. Eurich DT, McAlister FA,

    Blackburn DF, et al. Benefits and harms

    of antidiabetic agents in patients with

    diabetes and heart failure: systematic

    review. BMJ 2007;335:497

    16. Dormandy JA, Charbonnel B,

    Eckland DJ, et al. Secondary prevention

    of macrovascular events in patients with

    type 2 diabetes in the PROactive Study

    (PROspective pioglitAzone Clinical Trial

    In macroVascular Events): a randomized

    controlled trial. Lancet

    2005;366:1279-89

    17. Erdmann E, Dormandy JA,

    Charbonnel B, et al. The effect of

    pioglitazone on recurrent myocardial

    infarction in 2,445 patients with

    type 2 diabetes and previous myocardial

    infarction: results from the PROactive

    (PROactive 05) Study. J Am

    Coll Cardiol 2007;49:1772-80

    18. Wilcox R, Bousser MG, Betteridge DJ,

    et al. Effects of pioglitazone in patients

    with type 2 diabetes with or without

    previous stroke: results from PROactive

    (PROspective pioglitAzone Clinical Trial

    In macroVascular Events 04). Stroke

    2007;38:865-73

    19. Bardot O, Aldridge TC, Latruffe N,

    Green S. PPAR-RXR heterodimer

    activates a peroxisome proliferator

    response element upstream of the

    bifunctional enzyme gene.

    Biochem Biophys Res Commun

    1993;192:37-45

    20. Gearing KL, Gottlicher M, Teboul M,

    et al. Interaction of the

    peroxisome-proliferator-activated receptor

    and retinoid X receptor. Proc Natl Acad

    Sci USA 1993;90:1440-4

    21. Wilding JP. PPAR agonists for the

    treatment of cardiovascular disease in

    patients with diabetes.

    Diabetes Obes Metab

    2012;14(11):973-82

    22. Schoonjans K, Staels B, Auwerx J. The

    peroxisome proliferator activated

    receptors (PPARS) and their effects on

    lipid metabolism and adipocyte

    differentiation. Biochim Biophys Acta

    1996;1302:93-109

    23. Schoonjans K, Staels B, Auwerx J. Role

    of the peroxisome proliferator activated

    receptor (PPAR) in mediating the effects

    of fibrates and fatty acids on gene

    expression. J Lipid Res 1996;37:907-25

    24. Nagasawa T, Inada Y, Nakano S, et al.

    Effects of bezafibrate, PPAR pan-agonist,

    and GW501516, PPARdelta agonist, on

    development of steatohepatitis in mice

    fed a methionine- and choline-deficient

    diet. Eur J Pharmacol 2006;536:182-91

    25. Sahebkar A, Watts GF. Fibrate therapy

    and circulating adiponectin

    concentrations: a systematic review and

    meta-analysis of randomized placebo-

    controlled trials. Atherosclerosis

    2013;230:110-20

    26. Masaki T, Chiba S, Tatsukawa H, et al.

    Adiponectin protects LPS-induced liver

    injury through modulation of TNF-alpha

    in KK-Ay obese mice. Hepatology

    2004;40:177-84

    27. Tontonoz P, Spiegelman BM. Fat and

    beyond: the diverse biology of

    PPARgamma. Annu Rev Biochem

    2008;77:289-312

    28. Feige JN, Gelman L, Michalik L, et al.

    From molecular action to physiological

    outputs: peroxisome proliferator activated

    receptors are nuclear receptors at the

    crossroads of key cellular functions.

    Prog Lipid Res 2006;45:120-59

    29. Spiegelman BM. PPAR-gamma:

    adipogenic regulator and

    thiazolidinedione receptor. Diabetes

    1998;47:507-14

    30. Van Wagner LB, Rinella ME. The role

    of insulin-sensitizing agents in the

    treatment of nonalcoholic steatohepatitis.

    Thera Adv Gastroenterol 2011;4:249-63

    31. Lutchman G, Promrat K, Kleiner DE,

    et al. Changes in serum adipokine levels

    during pioglitazone treatment for

    nonalcoholic steatohepatitis: relationship

    to histological improvement.

    Clin Gastroenterol Hepatol

    2006;4:1048-52

    32. Neuschwander-Tetri BA, Brunt EM,

    Wehmeier KR, et al. Improved

    nonalcoholic steatohepatitis after

    48 weeks of treatment with the PPAR-

    gamma ligand rosiglitazone. Hepatology

    2003;38:1008-17

    33. Ratziu V, Giral P, Jacqueminet S, et al.

    Rosiglitazone for nonalcoholic

    steatohepatitis: one-year results of the

    randomized placebo-controlled fatty liver

    improvement with rosiglitazone therapy

    (FLIRT) trial. Gastroenterology

    2008;135:100-10

    34. Ratziu V, Charlotte F, Bernhardt C,

    et al. Long-term efficacy of rosiglitazone

    in nonalcoholic steatohepatitis: results of

    the fatty liver improvement by

    rosiglitazone therapy (FLIRT 2)

    extension trial. Hepatology

    2010;51:445-53

    35. Ratziu V, Caldwell S,

    Neuschwander-Tetri BA. Therapeutic

    trials in nonalcoholic steatohepatitis:

    insulin sensitizers and related

    methodological issues. Hepatology

    2010;52:2206-15

    36. Belfort R, Harrison SA, Brown K, et al.

    A placebo-controlled trial of pioglitazone

    in subjects with nonalcoholic

    steatohepatitis. N Engl J Med

    2006;355:2297-307

    37. Tolman KG. The safety of

    thiazolidinediones. Expert Opin Drug Saf

    2011;10:419-28

    38. Rubenstrunk A, Hanf R, Hum DW,

    et al. Safety issues and prospects for

    future generations of PPAR modulators.

    Biochim Biophys Acta

    2007;1771:1065-81

    39. Grey A. Skeletal consequences of

    thiazolidinedione therapy. Osteoporos Int

    2008;19:129-37

    40. Chalasani N, Younossi Z, Lavine JE,

    et al. The diagnosis and management of

    non-alcoholic fatty liver disease: practice

    Guideline by the American Association

    New PPAR agonists: potential treatments for atherogenic dyslipidemia and NAFLD

    Expert Opin. Pharmacother. (2014) 15(4) 9

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • for the Study of Liver Diseases, American

    College of Gastroenterology, and the

    American Gastroenterological

    Association. Hepatology

    2012;55:2005-23

    41. Goldberg RB, Kendall DM, Deeg MA,

    et al. A comparison of lipid and glycemic

    effects of pioglitazone and rosiglitazone

    in patients with type 2 diabetes and

    dyslipidemia. Diabetes Care

    2005;28:1547-54

    42. Bojic LA, Huff MW. Peroxisome

    proliferator-activated receptor delta:

    amultifaceted metabolic player.

    Curr Opin Lipidol 2013;24:171-7

    43. Gervois P, Fruchart JC, Staels B. Drug

    insight: mechanisms of action and

    therapeutic applications for agonists of

    peroxisome proliferator-activated

    receptors. Nat Clin Pract

    Endocrinol Metab 2007;3:145-56

    44. Fruchart JC. Selective peroxisome

    proliferator-activated receptor alpha

    modulators (SPPARMalpha): the next

    generation of peroxisome proliferator-

    activated receptor alpha-agonists.

    Cardiovasc Diabetol 2013;12:82

    .. A helpful review on the concept

    of SPPARM.

    45. Zhu T, Awni WM, Hosmane B, et al.

    ABT-335, the choline salt of fenofibric

    acid, does not have a clinically significant

    pharmacokinetic interaction with

    rosuvastatin in humans.

    J Clin Pharmacol 2009;49:63-71

    46. Goldberg AC, Bays HE, Ballantyne CM,

    et al. Efficacy and safety of ABT-335

    (fenofibric acid) in combination with

    atorvastatin in patients with mixed

    dyslipidemia. Am J Cardiol

    2009;103:515-22

    . A proof-of-concept trial showing the

    incremental benefit of combination

    therapy with ABT-335

    and atorvastatin.

    47. Mohiuddin SM, Pepine CJ, Kelly MT,

    et al. Efficacy and safety of ABT-335

    (fenofibric acid) in combination with

    simvastatin in patients with mixed

    dyslipidemia: a phase 3, randomized,

    controlled study. Am Heart J

    2009;157:195-203

    . A proof-of-concept trial showing the

    incremental benefit of combination

    therapy with ABT-335

    and atorvastatin.

    48. Jones PH, Davidson MH, Kashyap ML,

    et al. Efficacy and safety of ABT-335

    (fenofibric acid) in combination with

    rosuvastatin in patients with mixed

    dyslipidemia: a phase 3 study.

    Atherosclerosis 2009;204:208-15

    . A proof-of-concept trial showing the

    incremental benefit of combination

    therapy with ABT-335

    and atorvastatin.

    49. Takizawa T. K-877, a highly potent and

    selective PPAR alpha agonist, improves

    dyslipidemia and atherosclerosis in

    experimental animal models [abstract

    787]. Avaialble from: http://www.kenes.

    com/eas2012/abstracts/pdf/787.pdf

    50. Ishibashi S, Arai H, Yamashita S, et al.

    Benefical effects of K-877, a potent and

    highly selective PPARalpha agonist, on

    plasma lipoprotein profile in patients

    with atherogenic dyslipidemia [abstract

    525]. Proceedings of 80th EAS Congress;

    26 -- 29 May 2012; Milan, Italy

    . Clinical evidence on the efficacy of

    GFT-505.

    51. Kendall DM, Rubin CJ, Mohideen P,

    et al. Improvement of glycemic control,

    triglycerides, and HDL cholesterol levels

    with muraglitazar, a dual (alpha/gamma)

    peroxisome proliferator-activated receptor

    activator, in patients with type 2 diabetes

    inadequately controlled with metformin

    monotherapy: a double-blind,

    randomized, pioglitazone-comparative

    study. Diabetes Care 2006;29:1016-23

    52. Wilding JPH, Gause-Nilsson I,

    Persson A. Tesaglitazar, as add-on

    therapy to sulphonylurea, dose-

    dependently improves glucose and lipid

    abnormalities in patients with

    type 2 diabetes. Diab Vasc Dis Res

    2007;4:194-203

    53. Nissen SE, Wolski K, Topol EJ. Effect

    of muraglitazar on death and major

    adverse cardiovascular events in patients

    with type 2 diabetes mellitus. JAMA

    2005;294:2581-6

    . This study showed serious adverse

    events after muraglitazar use, leading

    to the discontinuation of the agent

    from further research.

    54. AstraZeneca Discontinues Development

    of GALIDA (tesaglitazar). AstraZeneca.

    2006-05-04. Retrieved 2013-11-11

    . This report concerns serious adverse

    events after tesaglitazar use, leading to

    the discontinuation of the agent from

    further research.

    55. Sanwald-Ducray P, Liogier Dardhuy X,

    Jamois C, Banken L. Pharmacokinetics,

    pharmacodynamics, and tolerability of

    aleglitazar in patients with

    type 2 diabetes: results from a

    randomized, placebo-controlled clinical

    study. Clin Pharmacol Ther

    2010;88:197-203

    56. Henry RR, Lincoff AM, Mudaliar S,

    et al. Effect of the dual peroxisome

    proliferator-activated receptor-alpha/

    gamma agonist aleglitazar on risk of

    cardiovascular disease in patients with

    type 2 diabetes (SYNCHRONY): a phase

    II, randomised, dose-ranging study.

    Lancet 2009;374:126-35

    57. Sprecher DL, Massien C, Pearce G, et al.

    Triglyceride:high-density lipoprotein

    cholesterol effects in healthy subjects

    administered a peroxisome proliferator

    activated receptor delta agonist.

    Arterioscler Thromb Vasc Biol

    2007;27:359-65

    58. Riserus U, Sprecher D, Johnson T, et al.

    Activation of peroxisome proliferator-

    activated receptor (PPAR)delta promotes

    reversal of multiple metabolic

    abnormalities, reduces oxidative stress,

    and increases fatty acid oxidation in

    moderately obese men. Diabetes

    2008;57:332-9

    59. Ooi EM, Watts GF, Sprecher DL, et al.

    Mechanism of action of a peroxisome

    proliferator-activated receptor (PPAR)-

    delta agonist on lipoprotein metabolism

    in dyslipidemic subjects with central

    obesity. J Clin Endocrinol Metab

    2011;96:E1568-76

    60. Olson EJ, Pearce GL, Jones NP,

    Sprecher DL. Lipid effects of peroxisome

    proliferator-activated receptor-delta

    agonist GW501516 in subjects with low

    high-density lipoprotein cholesterol:

    characteristics of metabolic syndrome.

    Arterioscler Thromb Vasc Biol

    2012;32:2289-94

    61. Bays HE, Schwartz S, Littlejohn T III,

    et al. MBX-8025, a novel peroxisome

    proliferator receptor-delta agonist: lipid

    and other metabolic effects in

    dyslipidemic overweight patients treated

    with and without atorvastatin. J Clin

    Endocrinol Metab 2011;96:2889-97

    . The first clinical evidence on the

    efficacy of MBX-8025.

    62. Choi YJ, Roberts BK, Wang X, et al.

    Effects of the PPAR-delta agonist MBX-

    8025 on atherogenic dyslipidemia.

    Atherosclerosis 2012;220:470-6

    A. Sahebkar et al.

    10 Expert Opin. Pharmacother. (2014) 15 (4)

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

  • 63. Hanf R, Darteil R, Hum DW, Staels B.

    GFT505 efficacy and safety in healthy

    volunteers and patients suffering from

    atherogenic dyslipidemia. Diabetologia

    [70th Annu Meet Sci Sess Am Diabetes

    Assoc (ADA) (June 25-29, Orlando)

    2010]. Diabetologia

    2010;59(Suppl 1):abstract 677-P

    . Clinical evidence on the efficacy of

    GFT-505.

    64. Cariou B, Zar Y, Staels B, Bruckert E.

    Effects of the new dual PPAR alpha/delta

    agonist GFT505 on lipid and glucose

    homeostasis in abdominally obese

    patients with combined dyslipidemia or

    impaired glucose metabolism.

    Diabetes Care 2011;34:2008-14

    . Clinical evidence on the efficacy of

    GFT-505.

    65. Cariou B, Hanf R, Lambert-Porcheron S,

    et al. Dual peroxisome proliferator-

    activated receptor alpha/delta agonist

    GFT505 improves hepatic and peripheral

    insulin sensitivity in abdominally obese

    subjects. Diabetes Care 2013;36:2923-30

    . Clinical evidence on the efficacy of

    GFT-505.

    66. Staels B, Rubenstrunk A, Noel B, et al.

    Hepatoprotective effects of the dual

    peroxisome proliferator-activated receptor

    alpha/delta agonist, GFT505, in rodent

    models of nonalcoholic fatty liver disease/

    nonalcoholic steatohepatitis. Hepatology

    2013;58:1941-52

    . A proof-of-concept animal study

    showing the potential of GFT-505 to

    attenuate hepatic steatosis.

    67. Whitehead JP. Diabetes: new conductors

    for the peroxisome proliferator-activated

    receptor gamma (PPARgamma)

    orchestra. Int J Biochem Cell Biol

    2011;43:1071-4

    68. Yew T, Toh SA, Millar JS. Selective

    peroxisome proliferator-activated

    receptor-gamma modulation to reduce

    cardiovascular risk in patients with

    insulin resistance. Recent Pat Cardiovasc

    Drug Discov 2012;7:33-41

    69. Dunn FL, Higgins LS, Fredrickson J,

    et al. INT131-004 study group. Selective

    modulation of PPARgamma activity can

    lower plasma glucose without typical

    thiazolidinedione side-effects in patients

    with type 2 diabetes.

    J Diabetes Complications 2011;25:151-8

    70. Naci H, Brugts JJ, Fleurence R, et al.

    Comparative benefits of statins in the

    primary and secondary prevention of

    major coronary events and all-cause

    mortality: a network meta-analysis of

    placebo-controlled and active-comparator

    trials. Eur J Prev Cardiol

    2013;20:641-57

    71. Fruchart JC, Sacks F, Hermans MP,

    et al. The Residual Risk Reduction

    Initiative: a call to action to reduce

    residual vascular risk in patients with

    dyslipidemia. Am J Cardiol

    2008;102:1K-34K

    72. Baigent C, Landray MJ, Reith C, et al.

    The effects of lowering LDL cholesterol

    with simvastatin plus ezetimibe in

    patients with chronic kidney disease

    (Study of Heart and Renal Protection):

    a randomised placebo-controlled trial.

    Lancet 2011;377:2181-92

    73. Stone NJ, Robinson J, Lichtenstein AH,

    et al. 2013 ACC/AHA guideline on the

    treatment of blood cholesterol to reduce

    atherosclerotic cardiovascular risk in

    adults: a report of the American college

    of cardiology/American heart association

    task force on practice guidelines. J Am

    Coll Cardiol 2013.

    [Epub ahead of print]

    74. Boden WE, Probstfield JL, Anderson T,

    et al. Niacin in patients with low HDL

    cholesterol levels receiving intensive statin

    therapy. N Engl J Med

    2011;365:2255-67

    75. Merck. Merck announces HPS2-

    THRIVE study of Tredaptive(Extended-Release Niacin/Laropiprant)

    did not achieve primary endpoint

    [online]. Available from: http://www.

    mercknewsroom.com/press-release/

    prescription-medicine-news/merck-

    announces-hps2-thrive-study-tredaptive-

    extended-relea

    76. Watts GF, Karpe F. Triglycerides and

    atherogenic dyslipidaemia: extending

    treatment beyond statins in the high-risk

    cardiovascular patient. Heart

    2011;97:350-6

    AffiliationAmirhossein Sahebkar1,2, Gerard T Chew3 &

    Gerald F Watts4

    Author for correspondence1Biotechnology Research Center,

    Mashhad University of Medical Sciences,

    Mashhad, Iran2Cardiovascular Research Center,

    Department of Medical Biotechnology,

    School of Medicine, Mashhad University of

    Medical Sciences, Mashhad, Iran3School of Medicine and Pharmacology,

    Royal Perth Hospital, University of

    Western Australia, Perth, Australia4Lipid Disorders Clinic, Cardiovascular

    Medicine, Royal Perth Hospital and Metabolic

    Research Centre, School of Medicine and

    Pharmacology, University of Western Australia,

    GPO Box X2213, Perth, Western Australia 6847,

    Australia

    Tel: +61 8 92240245;

    Fax: +61 8 92240246;

    E-mail: [email protected]

    New PPAR agonists: potential treatments for atherogenic dyslipidemia and NAFLD

    Expert Opin. Pharmacother. (2014) 15(4) 11

    Expe

    rt O

    pin.

    Pha

    rmac

    othe

    r. D

    ownl

    oade

    d fro

    m in

    form

    ahea

    lthca

    re.c

    om b

    y U

    nive

    rsity

    New

    Sou

    th W

    ales

    on

    01/2

    2/14

    For p

    erso

    nal u

    se o

    nly.

    AbstractIntroductionPPAR familyABT-335K-877Dual PPAR-/ agonistsGW501516MBX-8025GFT-505INT131ConclusionExpert opinionDeclaration of interestBibliography