You are on page 1of 6

Journal of Clinical Lipidology (2015) 9, 384–389

Review Article

Adenosine triphosphate citrate lyase: Emerging


target in the treatment of dyslipidemia
Hern!an N. Lemus, Carlos O. Mendivil, MD, PhD*

Universidad de los Andes Medical School, Bogot!


a, Colombia (Mr Lemus and Dr Mendivil); and Section of Endocrinology,
Department of Internal Medicine, Fundaci!on Santa Fe de Bogot!a, Bogot!a, Colombia (Dr Mendivil)

KEYWORDS: Abstract: Despite major advances in pharmacologic therapy over the last few decades, dyslipidemia
LDL; remains a prevalent, insufficiently recognized, and undercontrolled risk factor for cardiovascular
Lipid metabolism; disease. Statins are the mainstay of hypercholesterolemia treatment, but because of adherence and
Cholesterol; tolerability issues that limit dose titration, there is a need for additional therapies with good efficacy
Hypercholesterolemia; and better tolerability. Adenosine triphosphate (ATP) citrate lyase, a cytoplasmic enzyme responsible
Dyslipidemia; for the generation of acetyl coenzyme A for the de novo synthesis of fatty acids and cholesterol, is a
ATP citrate lyase; very interesting molecular target for the reduction of plasma lipids. Furthermore, ATP citrate lyase in-
AMPK; hibition may be accompanied by activation of 50 -adenosine monophosphate–activated protein kinase, a
ETC-1002; key signaling molecule that acts a central hub in cellular metabolic regulation. ETC-1002 is a small
Statin intolerance molecule inhibitor of ATP citrate lyase that also activates 50 -adenosine monophosphate–activated pro-
tein kinase, effectively reducing low-density lipoprotein cholesterol and inducing some other positive
metabolic changes. Recent evidence from phase I and II clinical trials in humans has shown a positive
efficacy and safety profile of ETC-1002, with low-density lipoprotein cholesterol reductions similar to
those attainable by usual doses of many statins and with no major apparent side effects. These results
potentially introduce a new family of medications that may expand our therapeutic arsenal against
hypercholesterolemia.
! 2015 National Lipid Association. All rights reserved.

Alterations of lipoprotein metabolism are highly preva- outcomes in both primary and secondary cardiovascular
lent risk factors for cardiovascular disease, and their (CV) prevention.1,2
appropriate treatment has the potential to generate a major Statins, a family of medications that work as inhibitors
public health impact. There is now abundant evidence to of 3-hydroxy-3-methylglutaryl (HMG) coenzyme A (CoA)
support the concept that among the different lipoprotein reductase (the rate-limiting enzyme in the cholesterol
fractions, tight control of plasma low-density lipoprotein biosynthesis pathway), are currently the mainstay of hy-
(LDL) cholesterol (LDL-C) has the most impact on hard percholesterolemia treatment, and their use has consistently
shown to reduce CV morbidity and mortality.1–3 Despite
the overall safety and efficacy of these medications, statin
Conflict of Interest: The authors have no conflict of interest to declare. intolerance remains a major clinical issue. The most impor-
* Corresponding author. Universidad de los Andes Medical School,
Carrera 7 # 116-05, Of. 413, Bogot!a, Colombia.
tant unwanted effects of statins can be grouped into 3 cat-
E-mail address: carlosolimpo@gmail.com egories: (1) overt myopathy and/or myalgia, even without
Submitted October 21, 2014. Accepted for publication January 9, 2015. significant elevations of plasma creatine phosphokinase4,5;

1933-2874/! 2015 National Lipid Association. All rights reserved.


http://dx.doi.org/10.1016/j.jacl.2015.01.002
Lemus and Mendivil ATP citrate lyase inhibition 385

(2) asymptomatic increases in liver transaminases4,6; and


(3) an increase in the risk of developing type 2 diabetes
mellitus, or a slight impairment in glycated hemoglobin
A1c among patients already diagnosed with type 2 diabetes
mellitus.7,8 Although these adverse effects are only seldom
severe or life threatening, they do compromise patient
adherence or treatment continuation and therefore the ex-
pected CV benefit of therapy.
Furthermore, despite the wide availability and clinical
use of statins, the percentage of patients who reach LDL-C
goals remains worryingly low.9,10 This may be related to
the dose-dependent nature of statin-related adverse effects,
which prevents strict dose escalation when necessary.4 To
overcome the limitations inherent to statin intolerance, Figure 1 Crucial role of ACL as a precursor supplier for both
and to provide patients with the full benefit that can be fatty acid and cholesterol synthesis. ACC, acetyl-CoA carbox-
derived from LDL-C reduction, new families of ylase; ACL, ATP citrate lyase; ATP, adenosine triphosphate;
cholesterol-lowering medications are being developed, CoA, coenzyme A; HMG, hydroxymethylglutaryl; TCA, tricar-
exploiting molecular targets different from HMG-CoA boxylic acid.
reductase. One such pharmacologic family is proprotein
convertase subtilisin/kexin type 9 (PCSK9) inhibitors, but limiting enzyme in de novo fatty acid biosynthesis) carbox-
concerns still remain about subcutaneous administration, ylates acetyl CoA to produce malonyl CoA. Then, through
cost, and the potential for neurocognitive side effects.11 repetitions of the 4-reaction cycle of de novo lipogenesis,
Recently, an entirely new approach to cholesterol lowering palmitic acid is synthesized. Modifications of this primary
has been developed. product lead to other (shorter, longer, or unsaturated) fatty
acids.17 Depending on the set of biosynthetic enzymes that
is predominantly expressed, acetyl CoA from ACL can also
Adenosine triphosphate citrate lyase and its be used for cholesterol synthesis: HMG-CoA is formed by
role in lipid synthesis condensation of acetyl CoA and acetoacetyl CoA (cata-
lyzed by HMG-CoA synthase) and later reduced by
Adenosine triphosphate (ATP) citrate lyase (ACL) is a HMG-CoA reductase to mevalonate.18 Mevalonate is then
cytosolic enzyme most highly expressed in lipogenic converted into 3-isopentenyl pyrophosphate by 3 consecu-
tissues such as liver and white adipose tissue.12 ACL cata- tive reactions requiring ATP. Multiple extension steps
lyzes a reaction in which 2 carbons from citrate are trans- involving iterative addition of extra 5-carbon modules leads
ferred to CoA, with consumption of 1 ATP molecule and to squalene, which is cyclized to form lanosterol. The
generation of acetyl CoA and oxaloacetate. ACL is thus a removal of 3 methyl groups plus reduction of one double
major contributor to the cytosolic pool of acetyl CoA, the bond and migration of another double bond in lanosterol
fundamental building block for the biosynthesis of both finally yields cholesterol (Fig. 1).
fatty acids and cholesterol. X-ray diffraction analyses of
ACL have revealed that it is conformed by 4 polypeptide
chains of similar size.13 Recent studies have identified the ACL and AMPK have opposite effects on lipid
binding site of both citrate and ATP to ACL, as well as
biosynthesis
two thirds of the enzyme’s complete 3-dimensional struc-
ture.14 The ACL gene is under transcriptional regulation The enzyme 50 -adenosine monophosphate–activated
by sterol regulatory element binding protein 1 (SREBP- protein kinase (AMPK) is a serine/threonine kinase that
1).15 The mature protein seems to be stabilized by works as a sensor for cellular depletion of ATP, whose
posttranslational phosphorylation of specific serine and activation results in the simultaneous shutting down of
threonine residues,16 although the relevance of these mod- several energy-consuming pathways and in the activation of
ifications has not been tested in vivo. energy-generating pathways. Activation of the AMPK
In liver, the essential organ of lipoprotein synthesis, pathway also influences a plethora of cellular functions
ACL plays a fundamental role in lipogenesis and steroido- through phosphorylation of other enzymes and transcription
genesis by supplying cytosolic acetyl CoA to both path- factors, which ultimate leads to changes in expression of
ways. By using a tricarboxylic acid cycle intermediate multiple genes.19 Among these target genes are the rate-
(citrate) to produce acetyl CoA, ACL can be seen as an limiting enzymes for steroidogenesis and fatty acid synthe-
important bridge between carbohydrate and lipid meta- sis (HMG-CoA reductase and ACC), but also enzymes with
bolism. Intramitochondrial citrate is transported via the a crucial role in gluconeogenesis and liver glucose produc-
so-called citrate shuttle to the cytosol, where it encounters tion, phosphoenolpyruvate carboxykinase and glucose-6-
ACL (Fig. 1). Acetyl-CoA carboxylase (ACC—the rate- phosphatase. The role of AMPK in lipid metabolism also
386 Journal of Clinical Lipidology, Vol 9, No 3, June 2015

involves regulation of mitochondrial long-chain fatty acids and lowering of plasma LDL-C.25 Expression of the gene
oxidation. Malonyl CoA, the product of ACC, is a strong for HMG-CoA is also stimulated, but in the context of low
inhibitor of carnitin-palmitoil transferase 1 (CPT-1), an cytosolic acetyl CoA, this does not impact cholesterol pro-
enzyme responsible for fatty acid transport into the mito- duction. Activation of AMPK via liver kinase B1 (LKB1)
chondrial matrix for their subsequent beta-oxidation.20 by ETC-1002 leads to inhibition of ACC and HMG-CoA
Phosphorylation of ACC by AMPK reduces malonyl CoA reductase (Fig. 2) and could potentially increase nitric oxide
production in the vicinity of CPT-1, resulting in its disinhi- production23 and reduce inflammatory and oxidative stress.26
bition and in increased transport and oxidation of long- The pharmacokinetics of ETC-1002 seem to make it
chain fatty acids.21 Thus, ACL activation provides the suitable for once-daily administration. MacDougall et al27
building blocks for fatty acid biosynthesis, whereas reported in a phase I study that used doses between 20
AMPK activation stimulates their degradation. and 220 mg/d, a directly proportional increase in the
maximum plasma concentration and 24-hour area under
the curve with increasing doses of ETC-1002. The mean
Inhibition of ACL half-life of the drug ranged between 16 and 33 hours across
the dosing range.
Among several ACL inhibitors that were initially tested, Given that ETC-1002 would be used in patients with
ETC-1002 (8-hydroxy-2,2,14,14-tetramethylpentadecaned– comorbidities, many of which may also be receiving
ioic acid) is the one currently in a most advanced stage of a statin, a clinical trial was undertaken, aimed at evaluating
clinical development. The molecule was first reported in the safety and pharmacokinetic interactions of ETC-
2004 as ESP55016, after a search for long-chain hydrocar- 1002 in patients with hypercholesterolemia who were
bon derivatives with positive effects on lipid and/or
metabolic profiles in vivo. Initial experiments demonstrated
the ability of ETC-1002 to inhibit fatty acid and sterol
synthesis and to stimulate palmitate oxidation in primary
rat hepatocytes, as well as to lower plasma non–high-
density lipoprotein (HDL) cholesterol (HDL-C), triglycer-
ides, and free fatty acids in a rat model of obesity and
metabolic syndrome (fa/fa).22 Furthermore, in this initial
set of experiments, ETC-1002 also reduced weight gain
and insulin resistance, as reflected by lower fasting levels
of both serum insulin and glucose. No direct effect of
ACL inhibition on HMG-CoA reductase levels or activity
was observed. The increase in fatty acid oxidation was
CPT-1 dependent, but the degree of AMPK or ACC phos-
phorylation did not change significantly, suggesting that
ETC-1002 increased CPT-1 only through reduced malonyl
CoA availability. Nonetheless, more recent studies in chow-
fed rats did find an increase in phospho-AMPK and
phospho-ACC after 2 weeks of treatment with ETC-1002
(30 mg/kg/d), and also a transient (10 min) increase in Figure 2 Mechanism of action of the ATP citrate lyase (ACL)
inhibitor ETC-1002 as a hypolipidemic agent. Direct inhibition
phosphorylation of AMPK, ACC, and HMG-CoA reductase
of ACL by ETC-1002 lowers cytosolic acetyl CoA, reducing sub-
in primary hepatocytes in vitro.23 ETC-1002 inhibits de strate availability for acetyl-CoA carboxylase (ACC) and therefore
novo sterol and fatty acid synthesis in primary rat hepato- malonyl-CoA levels. Lower malonyl-CoA levels have 2 conse-
cytes, inducing dose-dependent reductions cellular acetyl quences: reduced de novo fatty acid synthesis (FAS) and derepres-
CoA, malonyl CoA, and HMG-CoA, and simultaneously sion of carnitin-palmitoil transferase 1 (CPT-1), which increases
increasing cellular citrate. These changes start to occur fatty acid transport into the mitochondrial matrix to undergo
just 5 minutes after administration of the drug.23 beta-oxidation. Less acetyl CoA is also available for
As cytosolic acetyl-CoA levels in hepatocytes descend, hydroxymethylglutaryl-CoA (HMG-CoA) synthesis, effectively
cellular production and concentration of cholesterol go reducing flow through the cholesterol biosynthesis pathway. In
down, a change that is known to be sensed by the Sterol addition, 50 -adenosine monophosphate–activated protein kinase
response element binding protein (SREBP)/Cleavage- (AMPK) activation by ETC-1002 reduces the activity of ACC
and HMG-CoA reductase, the rate-limiting enzymes of fatty
Activating Protein system, leading to SREBP escort to the
acid and cholesterol synthesis, respectively. The reduction in intra-
Golgi apparatus, cleavage, and nuclear translocation.24 In the cellular cholesterol levels stimulates membrane expression of
nucleus, SREBPs binds to sterol response elements in several LDLR, augmenting LDL uptake and reducing plasma LDL.
target genes, regulating their transcription. One of those ATP, adenosine triphosphate; CoA, coenzyme A; LDL, low-
genes is the LDL receptor (LDLR) gene, whose increased density lipoprotein; LDLR, LDL receptor; TCA, tricarboxylic
expression leads to increased uptake of circulating LDL acid.
Lemus and Mendivil ATP citrate lyase inhibition 387

Table 1 Clinical trials of ETC-1002


Placebo-subtracted
percentage
Ref Participants Doses used (mg/d) LDL-C change Comments
27
56 healthy subjects, 6 per dose 20 NS Dose-finding phase I study. No
group 1 8 in placebo group, 60 211.2 relevant adverse effects.
93% male 100 216.5
120 215.0
140 221.3
180 227.1
220 236.1
25
177 patients with LDL-C 130- 40 216 No significant change in HDL-C
220 mg/dL 80 223 or triglycerides. Similar results
120 225 among patients with or without
high triglycerides.*
31
60 patients with type 2 diabetes, Forced titration, 239 No significant change in HDL-C
mean LDL-C 126 mg/dL. 2 wks 80 mg/d and or triglycerides, 41% reduction
2 wks 120 mg/d in hsCRP*
32
56 patients with hypercholester 60 Up to 32%
olemia and a history of statin 120
intolerance 180
240
32
58 patients with hypercholester 60 Up to 22%
olemia, ETC-1002 used as add-on 120
to 10 mg/d atorvastatin 180
240
32
Patients with hypercholesterol 120 Up to 30% in monotherapy;
emia, with or without statin 180 up to 48% 1 ezetimibe
intolerance 120 1 ezetimibe
180 1 ezetimibe
HDL-C, high-density lipoprotein cholesterol; hsCRP, high-sensitivity C-reactive protein; LDL-C, low-density lipoprotein cholesterol; NS,
nonsignificant; Ref, reference.
*The rate of adverse events was not significantly different from placebo.

concomitantly receiving atorvastatin 10 mg (clinicaltrials. cholesterol and fatty acid synthesis.22 In rats, the compound
gov NCT01779453). According to preliminary results also reduced plasma cholesterol and very low–density lipo-
announced by the manufacturer, ETC-1002 showed only protein production, albeit to a smaller extent than in vitro.
a small pharmacokinetic interaction with atorvastatin. In dogs, the effect of SB-204990 on plasma triglycerides
Several other compounds have proven capable of and cholesterol was still more modest, yet still significant.22
inhibiting ACL in vitro, including (2), (2), 2,2 difluoroci-
trate, several benzonesulfonamides, and the naturally
occurring compound hydroxycitrate. Nevertheless, their Clinical efficacy of ACL inhibition
development as pharmacologic agents has been limited by
issues related to poor ability to cross cell membranes, poor There is currently a quite efficacious and extensively
affinity for ACL requiring too high tissue concentrations, used family of LDL-lowering medications, the HMG-CoA
or, as in the case of hydroxycitrate, poor specificity leading reductase inhibitors, commonly referred to as statins.
to undesirable inhibition of other essential enzymes (iso- Despite their efficacy, many patients do not reach LDL
citrate dehydrogenase).28 The compound bb’-methyl- treatment goals because titration of statin doses may lead to
substituted a,w-dicarboxylic acid of 16-carbon length dose-dependent adverse effects.4–8 Against the backdrop of
(MEDICA 16) was identified as a potent inhibitor of neutral this unmet need, a number of studies have evaluated the
lipid synthesis in the rat,29 with additional direct inhibitory ACL inhibitor ETC-1002 as a lipid-lowering agent in hu-
activity over acyl-CoA carboxylase.30 However, MEDICA mans. Table 1 recapitulates the key features and results of
16 did not continue its development into the clinical phase. clinical phase I and phase II studies involving ETC-1002.
Yet, other studies have evaluated ACL inhibitor pre- The lowest dose of ETC-1002 with a significant LDL-C
cursors that can penetrate the plasma membrane, including reducing effect has been 60 mg/d, and the maximum dose
SB-204990, the lactone prodrug of the ACL inhibitor SB- tested so far for clinical efficacy is 240 mg/d (Table 1).
201076. In HepG2 cells, SB-204990 strongly inhibited both In adult patients with hypercholesterolemia, ETC-1002
388 Journal of Clinical Lipidology, Vol 9, No 3, June 2015

Table 2 Adverse events observed with ETC-1002 in 2 published clinical trials using different doses
25 25 25 31
Reference number
Daily dose of ETC-1002 40 mg 80 mg 120 mg 2 wk 80 mg
and then 2
wk 120 mg
Number of patients receiving this dose 45 44 44 30
Patients with any adverse event in ETC-1002 group, n (%) 34 (76) 32 (73) 31 (71) 14 (47)
Patients with any adverse event in placebo group, n (%) 33 (75) 33 (75) 33 (75) 21 (70)
Serious adverse events 0 0 0 —
Discontinuation because of adverse events in ETC-1002 group, n (%) 3 (7) 4 (9) 3 (7) 0
Discontinuation because of adverse events in placebo group, n (%) 4 (9) 4 (9) 4 (9) 1 (3)
Headache, n (%) 5 (11) 5 (11) 7 (16) 6 (20)
Dry eyes, n (%) — — — 2 (7)
Viral upper respiratory infection — — — 2 (7)
Abdominal pain — — — 0
Arthralgia, n (%) 2 (4) 2 (5) 1 (2) 2 (7)
Pruritus, n (%) — — — 1 (3)
Constipation, n (%) — — — 2 (7)
Dizziness — — — 0
Nausea, n (%) 3 (7) 3 (7) 4 (9) 0
Urinary tract infection, n (%) 4 (9) 1 (2) 0 —
Diarrhea, n (%) 3 (7) 3 (7) 1 (2) —
Fatigue, n (%) 3 (7) 1 (2) 1 (2) —
Cough, n (%) 2 (4) 3 (7) 1 (2) —
Myalgia, n (%) 2 (4) 2 (5) 3 (7) 0
Bronchitis, n (%) 0 3 (7) 1 (2) —
Pain in extremity, n (%) 0 0 4 (9) —
AST/ALT . 3 ! ULN, n (%) 0 1 (2) 0 —
CK . 5 ! ULN 0 0 0 —
AST, aspartate amino transferase; ALT, alanine amino transferase; CK, creatine kinase; ULN, upper limit of normal.

has induced dose-dependent LDL-C reductions between has provided an additional LDL-C reduction of up to
16% and 25%, regardless of plasma triglycerides.25 22%. Similarly, combined use with ezetimibe 10 mg/d
Non–HDL-C (18%-27%) and plasma apolipoprotein B has lowered LDL-C levels up to 48% (180 mg of ETC-
(15%-22%) were reduced to a similar degree, whereas 1002 plus 10 mg of ezetimibe), an effect similar to that ob-
plasma triglycerides were reduced nonsignificantly and tained with substantial doses of most available statins.
only by the 40 mg (15%) and 80 mg (12%) doses. Levels In all clinical studies undertaken so far, the rate of
of lipoprotein(a) were completely unaltered with any of adverse effects and/or discontinuation in the ETC-1002
the doses of ETC-1002 in this study. An even more marked treatment groups has been equal to or lower than that in the
hypocholesterolemic effect was observed in a study that placebo group of each study. This was also true in the
included only patients with type 2 diabetes and hypercho- patients with a documented history of intolerance to statins,
lesterolemia31; a 4-week treatment course reduced LDL-C suggesting that muscle-related adverse events are not
levels by 39% on average, with a simultaneous reduction necessarily a direct consequence of reductions in LDL-C,
of circulating high-sensitivity C-reactive protein of 41%. but instead an effect related to other properties of statins as
Non–HDL-C was reduced by 20%, with no significant a pharmacologic group. Treatment with ETC-1002 has not
changes in HDL-C or triglycerides. This increased efficacy increased plasma levels of liver transaminases, creatine
in the context of insulin resistance and/or type 2 diabetes kinase, bilirubin, or creatinine (Table 2).
may be related to activation of the AMPK pathway by
ETC-1002. Conclusions
Preliminary results of studies sponsored by the manu-
facturer of ETC-1002 have also shown positive results for There is currently a need for cholesterol-lowering drugs
monotherapy in statin-intolerant patients and for the com- that can be used to replace statins in intolerant patients or to
bined use of the ACL inhibitor plus other cholesterol- complement their effect when treatment goals are not
reducing agents. In patients with a documented history reached. Given its central role in modulating availability
of intolerance to statins, ETC-1002 60 to 240 mg/d has of the key substrate for cellular lipid synthesis, ACL is a
reduced plasma LDL-C up to 32%.29 Addition of the new very attractive target for the development of such agents.
agent in the same dose range to 10 mg/d of atorvastatin ETC-1002 is currently the ACL inhibitor whose clinical
Lemus and Mendivil ATP citrate lyase inhibition 389

development program is most advanced. ETC-1002 is also 12. Wang Q, Li S, Jiang L, et al. Deficiency in hepatic ATP-citrate lyase
an AMPK activator, an additional molecular target that affects VLDL-triglyceride mobilization and liver fatty acid composi-
tion in mice. J Lipid Res. 2010;51:2516–2526.
potentially expands the impact provided by ACL inhibition 13. Sun T, Hayakawa K, Fraser ME. ADP-Mg21 bound to the ATP-grasp
by modulating fatty acid oxidation and carbohydrate domain of ATP-citrate lyase. Acta Crystallogr Sect F Struct Biol Cryst
metabolism. Despite the small number of patients studied Commun. 2011;67:1168–1172.
so far, results of clinical trials with the new agent look 14. Sun T, Hayakawa K, Bateman KS, Fraser ME. Identification of the cit-
promising and validate the mechanism, possibly paving the rate binding site of human ATP-citrate lyase using X-ray crystallog-
raphy. J Biol Chem. 2010;285:27418–27428.
way for a new family of hypolipidemic drugs. 15. Zaidi N, Swinnen JV, Smans K. ATP-citrate lyase: a key player in can-
Further studies are needed to test whether ACL inhibition cer metabolism. Cancer Res. 2012;72:3709–3714.
and/or AMPK activation will reduce systemic inflammation 16. Wang Y, Wang Y, Shen L, Pang Y, Qiao Z, Liu P. Prognostic and ther-
and ameliorate other metabolic disturbances beyond pure apeutic implications of increased ATP citrate lyase expression in hu-
hypercholesterolemia, to assess the benefits and risks of its man epithelial ovarian cancer. Oncol Rep. 2012;27:1156–1162.
17. Ameer F, Scandiuzzi L, Hasnain S, Kalbacher H, Zaidi N. De novo
use in the long term, and to fully characterize the lipogenesis in health and disease. Metabolism. 2014;63:895–902.
pharmacokinetics and drug interaction profile of ETC-1002. 18. Gorin A, Gabitova L, Astsaturov I. Regulation of cholesterol biosyn-
thesis and cancer signaling. Curr Opin Pharmacol. 2012;12:710–716.
19. Ruderman NB, Carling D, Prentki M, Cacicedo JM. AMPK, insulin resis-
Acknowledgments tance, and the metabolic syndrome. J Clin Invest. 2013;123:2764–2772.
20. Schreurs M, Kuipers F, van der Leij FR. Regulatory enzymes of mito-
Both authors contributed equally to the literature search, chondrial beta-oxidation as targets for treatment of the metabolic syn-
article selection, analysis of the information, and writing drome. Obes Rev. 2010;11:380–388.
21. O’Neill HM, Holloway GP, Steinberg GR. AMPK regulation of fatty
and revision of the article. Both authors approved the final
acid metabolism and mitochondrial biogenesis: implications for
version of this article and its submission. obesity. Mol Cell Endocrinol. 2013;366:135–151.
22. Filippov S, Pinkosky SL, Newton RS. LDL-cholesterol reduction in
patients with hypercholesterolemia by modulation of adenosine
References triphosphate-citrate lyase and adenosine monophosphate-activated
protein kinase. Curr Opin Lipidol. 2014;25:309–315.
1. Cholesterol Treatment Trialists’ (CTT) Collaborators, Mihaylova B, 23. Pinkosky SL, Filippov S, Srivastava RA, et al. AMP-activated protein
Emberson J, Blackwell L, et al. The effects of lowering LDL choles- kinase and ATP-citrate lyase are two distinct molecular targets for
terol with statin therapy in people at low risk of vascular disease: ETC-1002, a novel small molecule regulator of lipid and carbohydrate
meta-analysis of individual data from 27 randomised trials. Lancet. metabolism. J Lipid Res. 2013;54:134–151.
2012;380:581–590. 24. Sharpe LJ, Brown AJ. Controlling cholesterol synthesis beyond
2. Cholesterol Treatment Trialists’ (CTT) Collaboration, Baigent C, 3-hydroxy-3-methylglutaryl-CoA reductase (HMGCR). J Biol Chem.
Blackwell L, Emberson J, et al. Efficacy and safety of more intensive 2013;288:18707–18715.
lowering of LDL cholesterol: a meta-analysis of data from 170,000 25. Ballantyne CM, Davidson MH, Macdougall DE, et al. Efficacy and
participants in 26 randomised trials. Lancet. 2010;376:1670–1681. safety of a novel dual modulator of adenosine triphosphate-citrate
3. Taylor F, Huffman MD, Macedo AF, et al. Statins for the primary pre- lyase and adenosine monophosphate-activated protein kinase in pa-
vention of cardiovascular disease. Cochrane Database Syst Rev. 2013; tients with hypercholesterolemia: results of a multicenter, randomized,
1:CD004816. double-blind, placebo-controlled, parallel-group trial. J Am Coll Car-
4. Matteucci E, Giampietro O. Statin intolerance: why and what to do— diol. 2013;62:1154–1162.
with a focus on diabetic people. Curr Med Chem. 2013;20:1397–1408. 26. Filippov S, Pinkosky SL, Lister RJ, et al. ETC-1002 regulates immune
5. Rallidis LS, Fountoulaki K, Anastasiou-Nana M. Managing the under- response, leukocyte homing, and adipose tissue inflammation via
estimated risk of statin-associated myopathy. Int J Cardiol. 2012;159: LKB1-dependent activation of macrophage AMPK. J Lipid Res.
169–176. 2013;54:2095–2108.
6. Alberton M, Wu P, Druyts E, Briel M, Mills EJ. Adverse events asso- 27. MacDougall D, Vanderlugt J, Rosenberg N, et al. ETC-1002, a modu-
ciated with individual statin treatments for cardiovascular disease: an lator of adenosine monophosphate-activated protein kinase and aden-
indirect comparison meta-analysis. QJM. 2012;105:145–157. osine triphosphate-citrate lyase, was safe and reduced low-density
7. Swerdlow DI, Preiss D, Kuchenbaecker KB, et al, DIAGRAM Con- lipoprotein-cholesterol in healthy volunteers. J Am Coll Cardiol.
sortium, MAGIC Consortium, InterAct Consortium. HMG-coenzyme 2013;61(Suppl 1):E1463.
A reductase inhibition, type 2 diabetes, and bodyweight: evidence 28. Abramson HN. The lipogenesis pathway as a cancer target. J Med
from genetic analysis and randomised trials. Lancet. 2014. Chem. 2011;54:5615–5638.
http://dx.doi.org/10.1016/S0140-6736(14)61183-1. 29. Rose-Kahn G, Bar-Tana J. Inhibition of lipid synthesis by beta beta’-
8. Erqou S, Lee CC, Adler AI. Statins and glycaemic control in individ- tetramethyl-substituted, C14-C22, alpha, omega-dicarboxylic acids in
uals with diabetes: a systematic review and meta-analysis. Diabetolo- cultured rat hepatocytes. J Biol Chem. 1985;260:8411–8415.
gia. 2014. http://dx.doi.org/10.1007/s00125-014-3374-x. 30. Rose-Kahn G, Bar-Tana J. Inhibition of rat liver acetyl-CoA carbox-
9. Hermans MP, Castro Cabezas M, Strandberg T, et al. Centralized ylase by beta, beta’-tetramethyl-substituted hexadecanedioic acid
Pan-European survey on the under-treatment of hypercholesterolaemia (MEDICA 16). Biochim Biophys Acta. 1990;1042:259–264.
(CEPHEUS): overall findings from eight countries. Curr Med Res 31. Gutierrez MJ, Rosenberg NL, Macdougall DE, et al. Efficacy and
Opin. 2010;26:445–454. safety of ETC-1002, a novel investigational low-density lipoprotein-
10. Leiter LA, Lundman P, da Silva PM, Drexel H, J€unger C, Gitt AK, cholesterol-lowering therapy for the treatment of patients with hyper-
DYSIS investigators. Persistent lipid abnormalities in statin-treated cholesterolemia and type 2 diabetes mellitus. Arterioscler Thromb
patients with diabetes mellitus in Europe and Canada: results of the Vasc Biol. 2014;34:676–683.
Dyslipidaemia International Study. Diabet Med. 2011;28:1343–1351. 32. Esperion Therapeutics: ETC-1002 phase 1 and phase 2 clinical trials,
11. Dadu RT, Ballantyne CM. Lipid lowering with PCSK9 inhibitors. Nat as of October 2014. Available at: http://www.esperion.com/therapies-
Rev Cardiol. 2014;11:563–575. progress/etc-1002/. Accessed last time on October 10, 2014.

You might also like