Download Targeting Acetyl-CoA Carboxylases: Small

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Evolution of metal ions in biological systems wikipedia , lookup

Proteolysis wikipedia , lookup

MTOR inhibitors wikipedia , lookup

Peptide synthesis wikipedia , lookup

Metalloprotein wikipedia , lookup

Basal metabolic rate wikipedia , lookup

Point mutation wikipedia , lookup

Glycolysis wikipedia , lookup

Enzyme inhibitor wikipedia , lookup

Lipid signaling wikipedia , lookup

Artificial gene synthesis wikipedia , lookup

Ketosis wikipedia , lookup

Biochemistry wikipedia , lookup

Biosynthesis wikipedia , lookup

15-Hydroxyeicosatetraenoic acid wikipedia , lookup

Amino acid synthesis wikipedia , lookup

Butyric acid wikipedia , lookup

Metabolism wikipedia , lookup

Specialized pro-resolving mediators wikipedia , lookup

Citric acid cycle wikipedia , lookup

Hepoxilin wikipedia , lookup

Fatty acid synthesis wikipedia , lookup

Fatty acid metabolism wikipedia , lookup

Transcript
168
Recent Patents on Anti-Cancer Drug Discovery, 2012, 7, 168-184
Targeting Acetyl-CoA Carboxylases: Small Molecular Inhibitors and their
Therapeutic Potential
Di-Xian Luo1,2, Di-Jun Tong3, Sandeep Rajput2, Chun Wang4,5, Duan-Fang Liao4,5, Deliang Cao2
and Edmund Maser6,*
1
Institute of Translational Medicine & Department of Laboratory Medicine, The First People's Hospital of Chenzhou,
102 Luojiajing Road, Chenzhou 423000, Hunan, P.R. China; 2Department of Microbiology, Immunology and Cell Biology, Simmons Cancer Institute, Southern Illinois University School of Medicine. 913 N. Rutledge Street, Springfield, IL
62794, USA; 3Department of Cardiovascular Medicine, Central Hospital of Yiyang, Yiyang 41300, P.R. China;
4
Institute of Pharmacy and Pharmacology, College of Pharmaceutics and Life Science, University of South China, Hengyang 421001, P.R. China; 5Department of Traditional Chinese Diagnotics, School of Pharmacy, Hunan University of
Chinese Medicine, Changsha 420108, P.R. China; 6Institute of Toxicology and Pharmacology for Natural Scientists,
University Medical School Schleswig-Holstein, Campus Kiel. Brunswiker Str. 10 24105, Kiel, Germany
Received: November 1, 2011; Accepted: January 12, 2012; Revised: February 12, 2012
Abstract: Acetyl-CoA carboxylases (ACCs) play a rate-limiting role in fatty acid biosynthesis in plants, microbes, mammals and humans. ACCs have the activity of both biotin carboxylase (BC) and carboxyltransferase (CT), catalyzing carboxylation of Acetyl-CoA to malonyl-CoA. In the past years, ACCs have been used as targets for herbicides in agriculture
and for drug discovery and development of human diseases, such as microbial infections, diabetes, obesity and cancer. A
great number of small molecule ACC inhibitors have been developed, including natural and non-natural (artificial) products. These chemicals target BC reaction, CT reaction or ACC phosphorylation. This article provides a comprehensive review and updates of ACC inhibitors, with a focus on their therapeutic application in metabolic syndromes and malignant
diseases. The patent status of common ACC inhibitors is discussed.
Keywords: Acetyl-CoA carboxylases, cancer, fatty acid synthesis, inhibitors, metabolic syndromes, obesity.
1. INTRODUCTION
Obesity, a worldwide epidemic, not only impacts life
quality, but also leads to a variety of co-morbidities, such as
diabetes, hypertension, dyslipidemia, coronary heart disease,
stroke, atherosclerosis, and cancer, accelerating obesityrelated morbidity and mortality [1-6]. It is needed to develop
effective therapeutics of obesity and the ensuring comorbidities. Acetyl-CoA carboxylases (ACCs) are ratelimiting enzymes in fatty acid de novo biosynthesis, catalyzing ATP-dependent carboxylation of acetyl-CoA to malonylCoA [7-9]. This reaction continuously proceeds in two steps
in participation of biotin prosthetic group, i.e., an ATPdependent biotin carboxylation and an ATP-independent
transfer of the carboxyl group Fig. (1) [10].
In humans and other mammals, there are two ACCs:
ACC1 (also called ACC-) with 265kDa and ACC2 (also
known as ACC-) with 280kDa. ACC1 and ACC2 are encoded by different genes, but share 75% amino acid sequence similarity except for extra 114 amino acids in the Nterminus of ACC2, in which the first 20 amino acid residues
constitute a signal peptide targeting mitochondrial membrane
[11]. Thereby, these two ACCs have distinct subcellular
*Address correspondence to this author at the Institute of Toxicology and
Pharmacology for Natural Scientists, University Medical School SchleswigHolstein, Campus Kiel. Brunswiker Str. 10 24105, Kiel, Germany;
Tel: 0431-597-3540; Fax: 0431-597-3558; E-mail: [email protected]
-9/12 $100.00+.00
distribution and function although they both catalyze the
production of malonyl-CoA Fig. (1) [10]. ACC1 is expressed
mostly in lipogenic tissues (the liver, adipose and lactating
mammary gland) and catalyzes the rate-limiting reaction in
the biosynthesis of long-chain fatty acids in cytosol. The
product malonyl-CoA is used for the elongation of acyl
chains by fatty acid synthase (FAS) [12-15]. In contrast,
ACC2 is expressed mainly in the liver, skeletal muscle and
heart with high energy metabolic activity, where its product
malonyl-CoA participates in the regulation of fatty acid oxidation by inhibiting carnitine palmitoyltransferase I
(CPT-I) that catalyzes the transition of long-chain acyl-CoA
across mitochondrial membranes [16]. Therefore, malonylCoA is a dual functional metabolite involved in both fatty
acid synthesis and oxidation, and ACC1/2 isozyme-nonselective inhibitors may selectively reduce fatty acid synthesis in lipogenic tissues and increase fatty acid oxidation in
energy production organs [17, 18].
In view of the importance of ACCs in fatty acid synthesis
and oxidation, the investigation of ACC inhibitors have been
attracting the interest of researchers and many promising
inhibitors have been developed and used in preclinical and
clinical studies for the treatment of obesity and metabolic
syndromes or in the management of malignancies [19]. This
article reviews the recent updates of ACC inhibitor exploitation and their patent situations.
© 2012 Bentham Science Publishers
Acetyl-CoA Carboxylase Inhibitors
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
169
O
O
O
O
HN
S
O
N
C
C
O
se
Biotin Carboxyl
Carrier protein
se
era
rase
nsf
oxyla
NH
ansfe
ytra
O
S
oxytr
Biotin
translocation
O
N
Bioti
n Ca
rb
Carb
box
Biotin Carboxyl
Carrier protein
B io ti n C ar b
o xy la se
Car
NH
HN
C
H2
S
Malonyl-CoA
CoA
O
C
S
xytran
C
Carbo
O
O
NH
sferas
e
Biotin Carboxyl
Carrier protein
HO
HN
Acetyl-CoA
ADP + Pi
C
N
CoA
O
O
O
S
B io ti n C ar b
o xy la se
C
H3
O
O
C
ATP
Fig. (1). ACC work mode. ACC has three functional domains and its catalyzation reaction occurs in two steps. The initial reaction is an
ATP-dependent transfer of CO2 from HCO3- to a nitrogen atom of the biotin prosthetic group of ACCs, and the 2nd step is an transfer of the
activated CO2 from biotin to acetyl-CoA, forming malonyl-CoA [10].
2. STRUCTURE OF ACETYL-COA CARBOXYLASES
ACCs are conserved in their amino acid sequence and
function in most living organisms, such as archaea (~34%
similarity in amino acid sequence), bacteria (~34% similarity), yeast (~56% similarity), plants (~54% similarity), rodents (~98% similarity), and mammals (~98% similarity),
compared to humans. However, genes encoding ACCs are
varied Fig. (2A). In mammals and most eukaryotic organisms, ACCs are a multiple domain polypeptide composed of
biotin carboxylase (BC), biotin carboxyl carrier (BCCP), and
carboxyltransferase (CT) domains that are encoded by a single gene. ACCs from Streptomyces coelicolor (S. coelicolor)
comprise subunit containing BC and BCCP domains and subunit (CT domain), encoded by an accA1(A2) and pccB
gene, respectively [20-22]. ACC in Archaeal Acidianus brierleyi consists of 3 subunits encoded by accC, accB, and
pccB gene, respectively [23]. In Escherichia coli (E. coli),
carboxyltransferase of ACCs consists of subunit and subunit encoded by accA and accD, respectively [24]. Therefore, in many low grade organisms, ACC is an unstable
multi-submit enzyme comprised of BC, BCCP and CT
subunits. BC domain/subunit catalyzes carboxylation of N1
atom in ureido ring of biotin covalently linked to a lysine
residue in BCCP with bicarbonate as a donor of carboxyl
group and ATP as an energy source. CT domain/subunit
catalyzes the transfer of the carboxyl group from the N1
atom to the methyl group of acetyl-CoA [17]. Significant
sequence homology exist between the BC subunit and eukaryotic BC domain, but the conservation of the CT compo-
nent is much lower [25]. An exception appears in plants
where ACCs exist as a multi-functional single protein (MFACC) and a multi-subunit heteromeric complex (MS-ACC)
Fig. (2B) [26, 27].
2.1. BC Domain of acetyl-CoA Carboxylases
Crystal structure shows that yeast BC domain consists of
20 -strands (1-20) and 21 -helices (A–U), forming
three sub-domains (A, B, and C) and an ATP-grasp fold [2831] Fig. (3A). The A-domain (residues 1-175) consists of
helices A-G and strands 1-5; B-domain (residues 234293) holds helices K and L and strands 9-11; and Cdomain (residues 294-566) is composed of anti-parallel sheet (12-20) flanked with helices M-U. Residues 176233) comprise an AB linker (helices H-J and 6-8). A-,
C-domains and AB-linker form a cylindrical structure with
ATP located at one end and B-domain acts as a lid at another
end Fig. (3B). ATP binding site, i.e., the active site of enzyme is located at the interface of the B-domain and cylinder
[30]. The B-domain keeps open conformation to the entry of
substrate or the release of product, but is closed during the
catalytic process.
2.2. CT Domain of Acetyl-CoA Carboxylases
Crystal structures of human ACC CT domain in complex
with CP-640186 and bovine CT domain in complex with
novel inhibitors have been identified [32, 33]. Yeast CT domain dimer was also identified as a free enzyme or in complexes with CoA [25], herbicides haloxyfop/diclofop [34], or
170 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
A
BC
Mammalian/Wheat/Yeast
Luo et al.
a subunit
S. coelicolor
Encoding gene:
BC
Encoding gene:
accC
MF-ACC
b subunit
accA1 (A2)
BC
Acidianus brierleyi
CT
BCCP
BCCP
BCCP
accB
CT
MF/MS-ACC
CT
MS-ACC
pccB
pccB
CT
BC
E. coli
Encoding gene:
B
accC
Gramineae
MF-ACC
Plastid
MF-ACC
BCCP
accB
MS-ACC
accA
accD
Dicot
MF-ACC
Plastid
MS-ACC
Fig. (2). ACC function domain. (A) ACC protein domains and encoding genes. ACCs of mammalian, wheat and yeast are composed of BC,
BCCP, and CT domains encoded by a single gene. ACC from S. coelicolor consists of subunit consisting of BC and BCCP domains and subunit, encoded by an accA1 (A2) and pccB gene, respectively. ACC form Acidianus brierleyi consists of 3 subunits encoded by an accC,
accB, and pccB gene, respectively. In E. coli, carboxyltransferase of ACCs consists of subunit and subunit encoded by accA and accD
gene. (B) ACC forms in plant, a multi-functional single protein (MF-ACC) and a multi-subunit heteromeric complex (MS-ACC).
an inhibitor CP-640186 [35]. This yeast CT domain dimer is
formed by a side-to-side reverse arrangement of two monomers [36]. A yeast CT domain monomer comprises 24 helices and 29 -strands, constructing two sub-domains (Nand C-domains) intimately associated with each other Fig.
(3C). The N-domain consists of residues 1484-1824 (strands
1-13 and helices 1-8), and the C-domain is composed of
residues 1825-2202 (1-12 and helices 1-8). The N- and
C-domains share similar polypeptide backbone folds with a
central -- superhelix (strands 5, 7, 9, and 11 and
helix 6). Catalytic pocket/cavity is formed by small sheets and 6 helix of -- superhelix of two domains, featured with additional binding surface for CoA Fig. (3C and
3D). The active site is located at the middle site of the interface of N- and C-domains in the dimer. Conserved residues
in the active site, Arg 1954 and Arg 1731 in particular, are
important for carboxyl group recognition of malonyl-CoA,
and the N1 atom of biotin itself functions as a general base
[25].
2.3. BCCP Subunit/Domain of Acetyl-CoA Carboxylases
BCCP subunit in E. coli contains the essential biotin covalently bound to lys 35 from the C-terminus, and the integral BCCP has strong tendency to aggregate [37, 38]. The
molar ratio of BC to BCCP subunits in E. coli is 1:2 [39].
The N-terminus (residues 1-30) of BCCP is responsible for
the interaction with BC, and the BC·BCCP complex could be
biotinylated in vitro.
3. REGULATION OF ACETYL-COA CARBOXYLASE
ACTIVITY
Due to the importance in energy and lipid metabolism,
ACCs activity is regulated at multiple levels, including transcriptional, posttranslational, and metabolite-allosteric regulations. Transcription of ACC1/2 genes is controlled by
sterol-regulatory-element binding protein 1 (SREBP-1), liver
X receptor, retinoid X receptor, peroxisome-proliferationactivated receptors (PPARs), forkhead box O (FOXO),
C/EBP and PPAR co-activator (PGC) [40-44]. By stimulating these signalings, a variety of factors and hormones, such
as glucose, insulin, and thyroid hormones regulate ACC expression. Please refer to the recent review articles for more
details [45-52].
Posttranslational regulation of ACC activity includes
phosphorylation and stabilization [53]. ACCs are phosphorylated as inactive monomers. On the contrary, dephosphorylation activates ACCs that self-associates for a functional multimeric filamentous complex. Induced polymerization of
mammalian acetyl-CoA carboxylase by MIG12 provides a
tertiary level of regulation of fatty acid synthesis [54]. AMPactivated kinase (AMPK), regulated by a variety of stress
signals and adipokines, (e.g. leptin and adiponectin), mediates the phosphorylation of ACC at Ser 79, Ser 1200, and
Ser 1215 [45, 55-57], and protein kinase A (PKA) activated
by low blood glucose phosphorylates ACCs at Ser 77, and
Ser 1200 [42, 58]. Additionally, breast cancer protein 1
Acetyl-CoA Carboxylase Inhibitors
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
A
B
B-Domain
C-Domain
C
b11 b6
ATP
aR
B-Domain
aK
b9
aT
aS
b15 b16
b17 b14 b12
aI
b13
b19
Sor aN
b18
aM
b20
aD
aE
C
AB
linker
linker
aG
b1
aP
b 3 b 2 aF b 4
b5
aO
A-Domain
ATP
aJ
b8 b7 AB
aH
aC
B-Domain
aL
b10
aU
aQ
C-Domain
Sor
aA
aB
N
N
D
C
N Domain
CoA
N
Mol2
C domain
C Domain
b7D
A-Domain
Mol1
b7C
N domain b7A
b7B a3A
171
a6C
a6
a 6B
CoA a4
a6A
a5
a3
b11 b 9
a7
b6
b10 b
8
b12
a8
b13
b4
b3
a1
b2
a6
a6D
a7
b 10
a8A
N
a2
b8 b6
b 4A
a3
b4
b3
a2
a8
a8B
C
b 4B
a4
b9
b 11
b 12
b1
a5
N
a8C
b1
N
A
C-Domain
N Domain
C
Fig. (3). 3D structure of ACC domains. A and B: Crystal structure of BC domain of yeast ACC 1. C and D: Crystal structure of CT domain
of ACC 2. The crystal structure figures were produced with permission as indicated in footnote.
(BRCA1) prevents ACCs from dephosphorylation at Ser 79
and Ser 1263 [59, 60].
Recent studies from our laboratory have revealed a novel
regulatory mechanism on ACC activity. Aldo-keto reductase
family 1 member B10 (AKR1B10), a NADPH-dependent
xenobiotic reductase primarily expressed in the colon and
small intestine [61-63], upregulated simultaneously with
ACC1 in tumorigenic transformation of human mammary
epithelial cells. Through direct association with ACC1,
AKR1B10 blocks its ubiquitin-dependent degradation, mediating fatty acid synthesis and lipid metabolism [64, 65].
ACC activity is also regulated in molecular conformation
by local metabolites. Citrate, a precursor of acetyl-CoA, allosterically activates ACCs, stimulating conversion of excessive acetyl-CoA to malony-CoA [66]. In contrast, palmitoylCoA, an end-product of fatty acid synthesis, promotes the
inactive conformation of ACCs, diminishing malonyl-CoA
production [67].
1
Reprinted from Molecular Cell, Vol. 16, Shen Y, Volrath SL, Weatherly SC, Elich
TD, Tong L, A mechanism for the potent inhibition of eukaryotic acetyl-coenzyme A
carboxylase by soraphen A, a macrocyclic polyketide natural product, 881-91., Copyright (2004) , with permission from Elsevier.
2
, From Zhang H, Yang Z, Shen Y, Tong L, Crystal structure of the carboxyltransferase
domain of acetyl-coenzyme A carboxylase, 2003; 299: 2064-2067. Reprinted with
permission from AAAS.
4. ACETYL-COA CARBOXYLASE INHIBITORS
As critical enzymes in fatty acid synthesis and energy
metabolism, ACCs are pathogenically implicated in several
human diseases, including metabolic syndromes and deadly
172 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
malignancies, and thus may be potent therapeutic targets. In
the past decades, ACC inhibitors have been extensively explored in preclinical and clinical trials. Discussed below are
updates on the investigation and development of ACC inhibitors.
Luo et al.
domain [110]. These inhibitors include sulfonamide-containing spirochromane derivatives (JP119987, US20110077262A1 and US7935712) [113, 114], non-spirocyclic matter
(JP119987), spiro [chromene-2,4-piperidin]-4(3H)-ones
(WO07011809 and WO07011811), and pyrazolospiroketone
(US 20110028390) [115].
4.1. Categories of Acetyl-CoA Carboxylase Inhibitors
ACC inhibitors include two main classes: natural and
non-natural (artificial) compounds. As summarized in Table
1 [17, 68-72], natural ACC inhibitors are isolated from natural products, such as Soraphen A (WO03011867) from Sporangium cellulosum [73-75], avenaciolide (WO03094912
and JP035998) from Aspergillus avenaceus, chloroacetylated
biotin (US6242610 and US6485941) from beans, egg yolks,
and cauliflower [72, 76, 77], pseudopeptide pyrrolidine dione antibiotics (Moiramide and Andrimid, US7544709 and
US20050080129) from bacteria [78, 79], curcumin
(WO05113069 and US20050267221) from turmeric [80].
The natural products inhibit ACC activity by three modes.
Curcumin phosphorylates and inactivates ACC via activating
AMPK [81-83], Moiramide B and Andrimid act as a CT
inhibitor, and other natural products inhibit the BC activity
by interacting with the allosteric site [78, 79].
Chemically synthesized inhibitors are composed of three
subclasses based on their chemical structures Table 2 [18,
33, 84-111]. The first subclass possess commonly extended
linear aliphatic region. Inhibitory activity of these compounds depends on their intracellular conversion to CoA
thioesters, inhibiting ACC activity by competing with acetylCoA in the CT catalyst. This subclass of ACC inhibitors
includes anthranilic acid derivatives (2-amino-,,trifluoro-p-toluic acids; US4307113 and JP11171848), sulfonamide derivatives (N-(1-[4-[2-(4-isopropoxyphenoxy)1,3-thiazol-5-yl]phenyl]ethyl)ethyl) acetamide; US0191323
and WO0202101), benzodioxepine derivatives (A1, 3,3Dimethyl-7-(4-methylsulfanyl-phenylethynyl)-3,4-dihydro2H-benzo[b][1,4] dioxepine; US0113374), alkynyl-substituted thiazole derivatives (A1, N-[4-(2-furyl)-5-(4-pyridyl)
thiazol-2-yl]pyridine-4-carboxamide; US0105919), heteroaryl-substituted thiazole derivatives (A1, 4-(6-((dimethylamino)methyl)pyridin-3-yl)-N-(4-(pentyloxy)-3(trifluoromethyl) phenyl) thiazol-2-amine; US0041720), ,
-dicarboxylic acid derivatives (MEDICA 16 and ESP55016; US4711896), benzoic acid derivative (S-2E;
US5145865), furan-2,5-dicarboxylic acid diamides (TOFA;
US3546255), aryloxyphenoxypropionate derivatives (Haloxyfop; US0014643), and cyclohexanedione derivatives
(sethoxydim, US4640706).
The second subclass of ACC inhibitors is bipiperidinylcarboxamide pharmacophores or cyclohexyl. They are potent, reversible, isozyme-nonselective inhibitors targeting the
CT domain of ACC [112], and however, the cyclohexyl derivatives exhibit potent inhibition of human ACC2, 10-fold
selectivity over inhibition of human ACC1 [33]. These compounds
include
bipiperidinylcarboxamide
analogs
(CP640186), (4-piperidinyl)-piperazine, pseudopeptide pyrrolidine dione antibiotics, benzthiazolylamide analogs, and
cyclohexyl derivative.
The third subclass of ACC inhibitors is spirochromanone
pharmacophores and they may inhibit ACC by targeting CT
4.2. Popular Acetyl-CoA Carboxylase Inhibitors
Although thousands of ACC inhibitors have been developed thus far, most published reports are based on studies
with these compounds Table 3, which are actively applied/investigated in agriculture for weeding and in laboratory animals for the treatment of obesity, type 2 diabetes
mellitus, or cancer.
4.2.1. Soraphen A
Soraphen A (1S,2S,3E,5R,6S,11S,14S,15R,16R,17S,
18S)-15,17-dihydroxy-5,6,16-trimethoxy-2,14,18-trimethyl11-phenyl-12,19-dioxabicyclo[13.3.1] nonadec-3-en-13-one)
(WO03011867) was isolated from the culture broth of
Sorangium cellulosum, a soil-dwelling myxobacterium [73,
74]. This polyketide natural product contains an unsaturated
18-membered lactone ring, an extracyclic phenyl ring, two
hydroxyl groups, three methyl groups, and three methoxy
groups [74, 116, 117]. Soraphen A is an allosteric inhibitor
of the BC domain, binding to the interface between the Adomain and C-domain, 25Å away from the putative ATP
binding site. ATP and Soraphen A molecules are located at
opposite ends of the cylindrical structure of the BC domain.
Soraphen A interacts with residues from helices N and O
and strands 17-20 in the C-domain, as well as several residues of helix C in the A-domain. Soraphen A is a noncompetitive eukaryotic ACC inhibitor with sensitivity at a
nanomolar level; soraphen A has no inhibitory activity towards the bacterial BC subunits [74, 118-120]. This species
selectivity of soraphen A is explained by the amino acid sequence and structural difference of the binding sites, e.g. the
absence of 18 in E. coli BC) [117].
4.2.2. Haloxyfop
Haloxyfop (2-[4-[3-chloro-5-(trifluoromethyl)pyridin-2yl] oxyphenoxy]propanoic acid) contains pyridine moiety,
and two forms of Haloxyfop are synthesized i.e. haloxyfopmethyl and haloxyfop-ethoxyethyl (US0184980). Haloxyfops are commercially used as pre- and post-emergence selective herbicides in broad leaf crops. They are absorbed by
the foliage and roots and hydrolyzed to haloxyfop, inhibiting
growth of meristematic tissues. The (R)-isomer, not (S)isomer, of haloxyfop is herbicidally active [34]. Another
derivative, diclofop (2-(4-(2,4-dichlorophenoxy) phenoxy)propionate) (US0184980) inhibits fatty acid synthesis
in Zea mays. Haloxyfop or diclofop binds to the active site at
the interface of CT dimer and leads to large conformational
changes of several residues, creating a highly conserved hydrophobic pocket extended into the core of the dimer [34].
Two residues Leu 1705 and Val 1967 that affect herbicide
sensitivity are located in this binding site, and their mutation
disrupts the structure of the domain and affect the response
to this inhibitor.
Acetyl-CoA Carboxylase Inhibitors
Table 1.
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
173
Natural ACC Inhibitors.
ACC Inhibitors
Chemical Formula
Action Mechanism
Sources
Active Species
Selectivity
Ref.
Soraphen
OMe
Inhibit BC reaction
Myxobacteria
Sorangium
Cellulosum
Fungal
ACC1
[17, 68]
Eukaryote
ACC2
Bacteria
ACC2?
[69-71]
[72]
(e.g. Soraphen A)
OMe
O
O
O
OH
OMe
OH
O
Avenaciolide
O
H
O
O
ACC1
Eukaryote
ACC2
Inhibit CT reaction
Bacteria
Bacteria
--
[131,
133, 138]
Activate AMPK
Turmeric
Mammalian
ACC2
[82]
Eukaryote
O
O
R2
R3
O
NH
N
H
N
H
O
Curcumin derivative
O
O
O
HO
OH
OCH3
Table 2.
Bacteria
O
H
Pseudopeptide
pyrrolidine dione
R1
Beans, egg
yolks, and
cauliflower
NH
N
H
S
(e.g. Andrimid,
Moiramide B)
Inhibit BC reaction
Fungal
C8H17
O
Cl
Aspergillus
avenaceus
H
O
Chloroacetylated
biotin
Inhibit glutamate
transport in
mitochondria ?
OCH3
Non-natural ACC Inhibitors.
ACC Inhibitors
Chemical Formula
Action Mechanism
Active Species
Selectivity
Ref.
Inhibitors with Linear Aliphatic Region
Anthranilic acid derivatives
O
Inhibit carboxylase
reaction?
Bacteria
ACC1, ACC2
[84-85]
Suppress
Eukaryote
ACC1, ACC2
[86]
Mammalian
ACC2
[87]
Eukaryote
NH
OR1
H
N R1
Sulfonamide derivatives
O
H
S N
O2
Benzodioxepine derivatives
R
O
O
ACC activation?
O
R2
?
174 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
Luo et al.
(Table 2) Contd….
ACC Inhibitors
Chemical Formula
O
Alkynyl-substituted thiazole derivatives
Action Mechanism
S
Selectivity
Ref.
?
Eukaryote?
ACC2
[88-91]
?
Eukaryote?
ACC1, ACC2
[92]
Inhibit CT reaction
Eukaryote
ACC1, ACC2
[93-94]
Noncompetitively
inhibit ACC by S-2ECoA
Eukaryote
ACC1, ACC2
[95-97]
Inhibit CT reaction
Bacteria
ACC1, ACC2
[111]
NH
R1
O
O
Heteroaryl-substituted
thiazole derivatives
R3
N
R2
N
R1
Alkanedicarboxylic derivatives
Active Species
O
O
O
OH
HO
(e.g. MEDICA 16,
ESP-55016)
R2
Benzoic acid derivative
(e.g.S-2E)
O
O
R1
R2
Furan-2,5-dicarboxylic
acid diamides
Eukaryote
R1
(e.g. TOFA)
O
Aryloxyphenoxypropionates
R1
R2
(e.g. Haloxyfop)
Cyclohexanediones
N
O
COOH
Inhibit CT reaction
Grass
--
[98-100]
Inhibit CT reaction
Grass
--
[98-101]
Inhibit CT reaction
Eukaryote
ACC1, ACC2
[18, 102103]
Inhibit CT reaction
Eukaryote
ACC1, ACC2
[104]
Me
O
R1
O
(e.g. Sethoxydim)
N
O
O
R3
R2
Inhibitors with Bipiperidinylcarboxamide Pharmacophore or Cyclohexyl
Bipiperidinylcarboxamide analog (e.g.
CP640186)
O
N
O
N
N
O
O
(4-Piperidinyl)-piperazine
N
N
O
N
N
N
O
Acetyl-CoA Carboxylase Inhibitors
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
175
(Table 2) Contd….
ACC Inhibitors
Chemical Formula
Benzthiazolylamide analogs
Action Mechanism
R2
O
Active Species
Selectivity
Ref.
Inhibit CT reaction
Eukaryote
ACC1, ACC2
Inhibit CT reaction
Mammalian
ACC1, ACC2
(10-fold selectivity over inhibition of human
ACC1)
[33]
Inhibit CT reaction
Mammalian
ACC1, ACC2
[107]
Inhibit CT reaction
Mammalian
ACC1, ACC2
[108]
Inhibit CT reaction
Mammalian
ACC1, ACC2
[109]
Inhibit CT reaction
Mammalian
ACC1, ACC2
[110]
N
[18, 105106]
NH
S
NHR1
O
O
Cyclohexyl derivatives
O
NH
HN
O
R
Inhibitors with Spirochromaone Pharmacophore
Sulfonamide-containing
Spirochromane Derivatives
O
R1
R2
O
H
N
A
R3
R4
O
R3
Non-spirocyclic matter
R1
R4
R2
H
N
R5
R6
O
Spiro[chromene-2,4piperidin]-4(3H)-ones
O
R1
O
N
R2
O
O
Azaspirochromanones
R1
N
O
N
R2
O
176 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
Table 3.
Luo et al.
Popular ACC Inhibitors.
Inhibitors
Chemical
(Abbr.)
Formula
Soraphen A
O
O
Chemical Name
Molecular
Weight
Molecular
Formula
(1S,2S,3E,5R,6S,11S,14S,15R,16R,17S,18S
)-15,17-Dihydroxy-5,6,16-trimethoxy2,14,18-trimethyl-11-phenyl-12,19dioxabicyclo [13.3.1]nonadec-3-en-13-one
520.65
C29H 44O8
(2E,4E,6E)-N-[(1S)-3-[[(2S)-3-Methyl-1[(3R,4S)-4-methyl-2,5-dioxopyrrolidin-3yl]-1-oxobutan-2-yl]amino]-3-oxo-1phenylpropyl]octa-2,4,6-trienamide
479.57
C27H 33N3 O5
(2E,4E)-N-[(1S)-3-[[(2S)-3-Methyl-1[(3R,4S)-4-methyl-2,5-dioxopyrrolidin-3yl]-1-oxobutan-2-yl]amino]-3-oxo-1phenylpropyl]hexa-2,4-dienamide
453.53
C25H 31N3 O5
2-[4-[3-Chloro-5-(trifluoromethyl)pyridin2-yl]oxyphenoxy]propanoic acid
361.70
C15H 11ClF3NO4
2-[1-(Ethoxyamino)butylidene]-5-(2ethylsulfanylpropyl)cyclohexane-1,3-dione
327.48
C17H 29NO 3S
5-Tetradecoxyfuran-2-carboxylic acid
324.45
C19H 32O4
OH
O
O
O
HO
Andrimid
O
H
N
O
O
H
N
O
H
N
O
O
Moiramide
B
H
N
O
O
H
N
O
H
N
O
O
Haloxyfop
Cl
HO
O
O
N
F
F
F
O
Sethoxydim
S
HN
O
O
ToFA
OH
O
O
O
Acetyl-CoA Carboxylase Inhibitors
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
177
(Table 3) Contd….
Inhibitors
Chemical
(Abbr.)
Formula
O
S-2E
N
Chemical Name
Molecular
Weight
Molecular
Formula
(+)-(S)p
-[1-(
p-Tertbutylphenyl)-2-oxo-4-pyrrolidinyl]methoxybenzoic acid
519.61
C28 H 42NO 6
3,3,14,14Tetramethylhexadecanedioic acid
342.51
C20H 38O4
8-Hydroxy-2,2,14,14-Tetramethylpentadecanedioic acid
344.49
C19H 36O5
[(3R)-1-[1-(Anthracene-9carbonyl)piperidin-4-yl]piperidin3-yl]-morpholin-4-ylmethanone
485.62
C30H 35N3 O3
O
OH
O
MEDICA
16
O
OH
HO
O
O
ESP55016
OH
HO
O
OH
CP640186
O
N
O
N
N
O
4.2.3. Sethoxydim
Sethoxydim (2-[1-(ethoxyamino)butylidene]-5-(2-ethylsulfanylpropyl)cyclohexane-1,3-dione) (US4602935 and
US0033897) inhibits lipid synthesis in two dicot species,
Nicotiana sylvestris (wild tobacco) and Glycine max (soybean) [121]. It is a selective post-emergence herbicide used
to control annual and perennial grass weeds in broad-leaved
vegetables, fruits, fields, and ornamental crops. Sethoxydim
is rapidly absorbed through the leaf surfaces, transported in
the xylem and phloem, and accumulated in the meristematic
tissues. Non-susceptible broadleaf species have a different
acetyl-CoA carboxylase binding site resistant to sethoxydim.
Sethoxydim is water-soluble and does not bind readily with
soils, thus being mobile.
4.2.4. TOFA
Five-tetradecyloxy-2-furoic acid (TOFA) (US3546255)
itself has no activity. In adipocytes and hepatocytes, TOFA
is converted to 5-tetradecyloxy-2-furoyl-CoA (TOFyl-CoA)
that binds to CT domain and exerts an allosteric inhibition on
ACCs. TOFA is a mammalian ACC inhibitor. The inhibitory
activity of TOFA depends on its concentration relative to
fatty acids, cells, and nutritional state. In isolated rat adipocytes, TOFA inhibits fatty acid synthesis and leads to accumulation of lactate and pyruvate, and release of CO2 by
blocking synthesis of malonyl-CoA [122]. Ketogenesis from
palmitate was slightly inhibited (~ 20%) by TOFA at a concentration less than CoA, but the inhibition was almost complete (up to 90%) at a concentration equal to or greater than
the CoA [15]. In some conditions, TOFA may inhibit fatty
acid synthesis, but not affect fatty acid oxidation [122-124].
TOFA can also inhibit glycolysis as a secondary effect of
fatty acid synthesis inhibition and resultant citrate accumulation, a metabolite inhibitor of phosphofructokinase [125].
TOFA inhibition of ACCs in human cancer cells is controversial. It has been reported that TOFA induces the apoptosis
of lung cancer cells NCI-H460 and colon carcinoma cells
HCT-8 and HCT-15, but not of some breast and ovary cancer
cells, such as MCF-7 [15, 126-129].
4.2.5. Andrimid
Andrimid
((2E,4E,6E)-N-[(1S)-3-[[(2S)-3-methyl-1[(3R,4S)-4-methyl-2,5-dioxopyrrolidin-3-yl]-1-oxobutan-2yl]amino]-3-oxo-1-phenylpropyl]octa-2,4,6-trienamide;
JP11171848) is a hybrid non-ribosomal peptide-polyketide
178 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
antibiotic that can block the carboxyl-transfer reaction.
Structure–activity studies have led to development of new
analogues with modified pseudopeptide motifs and improved
efficacies in vivo and in vitro [130].
Andrimid and moiramide (A, B and C) are both natural
antibiotics with a hybrid non-ribosomal peptide polyketide
scaffold that is acylated at the N-terminus and modified by a
pyrrolidine dione moiety at the C-terminus. This class of
molecules is widely distributed in nature and has received
considerable attention after their cellular target is discovered
as ACCs [131]. Low primary sequence homology and structural distinction between the prokaryotic and eukaryotic
ACCs has made bacterial ACCs a long appreciated target for
antibacterial drug development. The andrimid biosynthetic
gene cluster from Pantoea agglomerans encodes an admT
with homology to the arboxyltransferase (CT) -subunit encoded by accD. E. coli cells with admT overexpression are
resistant to andrimid. When AdmT and CT -subunit AccA
are overexpressed in E. coli cells, an active heterologous
tetrameric CT A2T2 complex is formed. Andrimid-inhibition
assay shows an IC50 of 500 nM for the A2T2 complex, compared to 12 nM for E. coli CT A2D2. These data suggested
that AdmT, as an AccD homolog, confers resistance to andrimid [130].
4.2.6. Moiramide B
Pseudopeptide pyrrolidine dione antibiotics are isolated
from three distinct bacterial species of proteobacteria: Enterobacter sp., Vibrio sp., and Pseudomonas fluorescens
[132]. These antibiotics have broad antibacterial activity
against Gram-positive bacteria, such as Staphylococci and
Bacilli, and Gram-negatives, such as E. coli. The chemical
Moiramide B ((2E,4E)-N-[(1S)-3-[[(2S)-3-methyl-1-[(3R,
4S)-4-methyl-2,5-dioxopyrrolidin-3-yl]-1-oxobutan-2yl]amino]-3-oxo-1-phenylpro-pyl]hexa-2,4-dienamide;
US7544709) inhibits carboxyltransferase activity of ACCs
[131, 133]. In vitro and bacterial studies indicates that
Moiramide B demonstrates inhibitory activity at nanomolar
levels[39, 133, 134].
4.2.7. ESP-55016
Luo et al.
enantiomers (1-(4-tert-butylphenyl)-2-oxo-pyrrolidine-4-carboxylic acid, N-[(S)-(-)-[4-methyl-(alpha-methyl)benzyl]]-1(4-tert-butylphenyl)-2-oxo-pyrrolidine-4-carboxyamide, 4bromo-2-fluorobenzamide of (+)-4-[1-(4-tert-butylphenyl)-2oxo-pyrrolidine-4-yl]-methyloxy-benzoic acid) (US4831055)
showed an essentially equipotent activity in inhibiting fatty
acid- and sterol-biosynthesis, lowering down blood cholesterol and triglyceride levels [136, 137]. The in vivo active
form is S-2E ((+)-(S)-p-[1-(p-tert-butylphenyl)-2-oxo-4pyrrolidinyl] methoxybenzoic acid) that is converted into S2E-CoA in the liver, exerting non-competitive inhibition of
ACCs at Ki = 69.2M. S-2E-CoA also effectively inhibits 3hydroxy-3-methylglutaryl coenzyme-A (HMG-CoA) reductase at Ki = 18.11M [136]. When administered at 10 mg/kg
orally, liver S-2E-CoA are sufficient to inhibit HMG-CoA
reductase and ACCs, and therefore S-2E may be useful in the
treatment of familial hypercholesterolemia and mixed hyperlipidemia [95, 96, 137].
4.2.9. CP-640186
CP-640186 ([(3R)-1-[1-(anthracene-9-carbonyl) piperidin-4-yl]piperidin-3-yl]-morpholin-4-ylmethanone; WO03072197) is a potent inhibitor of mammalian ACCs, which can
reduce body weight and improve insulin sensitivity in animals. It is non-selective towards two isoforms ACC1 and
ACC2 with an IC50 at 50nM in rat, mouse and monkey [68].
In experimental animals, CP-640186 decreases malonyl-CoA
in lipogenic and oxidative tissues, reducing fatty acid synthesis and stimulating fatty acid oxidation [18]. In sucrose-fed
rats, CP-640186 reduces triglycerides in the liver, muscle
and adipose, and body weight by selectively reducing fats
but not lean body mass. In these animals, CP-640186 also
reduces leptin levels and induces hyperinsulinemia stemmed
from high sucrose diet [18].
5. APPLICATIONS OF ACETYL-COA CARBOXYLASE INHIBITORS
5.1. Herbicides in Agriculture
The earliest application of ACC inhibitors is in agriculture. Two classes of herbicides targeting ACCs, aryloxyphenoxypropionates (Haloxyfop) and cyclohexanediones
(Sethoxydim), have been commercially used for more than
twenty years [118].
ESP-55016 (8-hydroxy-2,2,14,14-tetramethyl-pentadecanedioic acid; US4711896), a -hydroxy-alkanedicarboxylic acid, is converted in vivo into a CoA derivative, ESP55016-CoA, which markedly inhibits the activity of ACC.
ESP55016 dually inhibits fatty acid and sterol synthesis
in vivo and in primary rat hepatocyte culture [135]. In obese
female Zucker (fa/fa) rats [135], ESP55016 favorably reduces serum non-HDL-cholesterol (non-HDL-C), triglyceride,
and non-esterified fatty acid levels, but increases serum
HDL-C and -hydroxybutyrate in a dose-dependent manner.
ESP55106 also increases the oxidation of [14C]-palmitate in a
carnitine palmitoyl transferase-I (CPT-I)-dependent manner
[135]. This indicates that ESP-55016 affects both fatty acid
and sterol synthesis and fatty acid oxidation through the
ACC/malonyl-CoA/CPT-I axis.
ACC inhibitors are also used to control invading organisms that depend on lipid synthesis for rapid proliferation
[131, 138, 139]. Antibacterial ACC inhibitors include (-)avenaciolide, chloroacetylated biotin, and pseudopeptide
pyrrolidine dione antibiotics (Andrimid and Moiramide B).
Especially, Andrimid and Moiramide B are natural antibiotics with broad antibacterial activity against Gram-positive
and negative bacteria, such as Staphylococci, Bacilli, and E.
coli [131, 138]. Antifungal ACC inhibitors include soraphen
A, (-)-avenaciolide, CP640186, MEDICA-16, and TOFA
[39, 69, 73, 74].
4.2.8. S-2E
5.3. Treatment of Metabolic Syndromes
S-2 ((+/-)-4-[1-(4-tert-Butylphenyl)-2-oxo-pyrrolidine-4yl]methyloxybenzoic acid), a anti-lipidemic agent, and its
Over the past 30 years, a variety of ACC inhibitors have
demonstrated treatment activity in animal models of dyslipi-
5.2. Antibiotics in Infection Diseases
Acetyl-CoA Carboxylase Inhibitors
demia [140, 141]. For example, alkyloxyarylcarboxylic acids
(e.g., TOFA) showed marked hypolipidemic activity in both
rats and monkeys [142, 143], and MEDICA, MEDICA-16 in
particular, displayed hypolipidemic, anti-diabetic, and antiatherosclerotic activity in relevant animal models [144-146].
The alkylthioacrylic acids appear to have similar activity to
MEDICA in intervening dyslipidemia in animals [147-149].
In addition, S-2E may hold the promise for the treatment of
familial hypercholesterolemia and mixed hyperlipidemia [95,
96], and -hydroxy-alkanedicarboxylic acid, ESP 55016,
favorably alters serum lipid profile of the Zucker rat diabetic
dyslipidemia [135]. CP-640186 is an isozyme-nonselective
ACC inhibitor, equally inhibiting ACC1 and ACC2 in rat,
mouse, monkey, and human. This chemical reduces fatty
acid synthesis and triglyceride secretion without affecting
cholesterol synthesis, thus decreasing apo-B secretion but
not apo-A1; CP-640186 also stimulates fatty acid oxidation
[17, 18].
5.4. Cancer Therapy
Exploration of ACC inhibitors for anticancer therapy is a
novel attracting field. De novo fatty acid synthesis is required for carcinogenesis, and in cancer cells, up to 95%
fatty acids used are newly synthesized despite adequate nutritional lipid supplies [150]. The newly synthesized fatty
acids in malignant cells are used for biomembrane synthesis
and lipid second messengers, promoting cell growth and
proliferation [151]. Therefore, the lipogenesis pathway is a
cancer target [152]. As a critical, rate-limiting enzyme in
fatty acid de novo biosynthesis, ACC1 is upregulated in multiple types of human cancers, such as breast and prostate
tumors. ACC is a novel target for cancer therapy [153]. Specific silencing of ACC1 by RNA interference (RNAi) results
in caspase-mediated apoptosis of breast, colon and prostate
cancer cells by depleting fatty acids/lipids and inducing oxidative stress derived from membrane damage of mitochondria [154, 155]. Similarly, our studies revealed that
AKR1B10 mediates ACC1 stability and fatty acid synthesis,
and silencing of AKR1B10 triggers ACC1-mediated breast
cancer cell growth inhibition and apoptosis [65]. Therefore,
fatty acid synthesis is essential for cancer cell growth and
survival, and ACC inhibitors may represent a novel class of
potent antitumor agents [66, 155, 156].
TOFA (5-tetradecyl-oxy-2-furoic acid) decreases fatty
acid synthesis and induces caspase activation and cell death
in most PCa cell lines by reducing ACC-mediated fatty acid
synthesis and inhibiting expression of androgen receptor
(AR), neuropilin-1 (NRP1) and Mcl-1 [157]. ACC inhibitor
soraphen A at nanomolar levels can block fatty acid synthesis and stimulate fatty acid oxidation in LNCaP and PC-3M
prostate cancer cells, inducing apoptosis [19, 158], and
TOFA showed capability of triggering apoptosis of lung
(NCI-H460) and colon (HCT-8 and HCT-15) carcinoma
cells through disturbing their fatty acid synthesis [126]. It is
obvious that further studies are merited to develop ACC inhibitors as anticancer agents.
6. CURRENT & FUTURE DEVELOPMENTS
ACCs catalyze the production of malonyl-CoA from acetyl-CoA and malonyl-CoA is an essential component for
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
179
fatty acid elongation and yet is an inhibitor of fatty acid oxidation. Therefore, ACCs are critical for fatty acid metabolism and organism growth and viability, explaining their
broadly conservation from bacteria to humans. Inhibition of
ACC activity is a potential treatment strategy for microbial
infections, metabolic syndromes, and cancer in humans. Currently available ACC inhibitors inhibit ACC activity through
binding to the carboxyltransferase-domain or the biotin carboxylase-domain. Among mechanistically distinct ACC inhibitors, isozyme-nonselective ACC inhibitors may hold
better therapeutic potential via decreasing fatty acid synthesis and enhancing oxidation. Isozyme-selective inhibitors
should have advantages and liabilities associated with a single isozyme inhibition. However, it should be noted that
isozyme nonselective ACC inhibitors may also have issues,
e.g. ACC1 knockout mice show embryonic lethality [159]
and also ACC2 inhibition may be ineffective [160]. The current intensive research on ACCs and inhibitors could lead to
the development of novel therapeutic agents against metabolic syndromes and cancers. Clinical efficacy of the ACC
inhibitors should be expected. Tissue fat reduction, weight
loss, improved insulin sensitivity, and relief of dyslipidemia
were observed in ACC2 knockout mice [161-163] and in
experimental animals treated with isozyme-nonselective
ACC inhibitors [18, 68] or ACC antisense oligonucleotides
[164], which compels a study in human clinics. The clinic
efficacy evaluation could focus initially on approved endpoints for obesity and dyslipidemia, improved insulin sensitivity, and reduction in hyperinsulinemia. Weight loss, percentage body fat, and regional fat distribution may be evaluated in longer-term clinical trials. The treatment outcomes
of coronary heart diseases and type 2 diabetes or other
significant health outcomes related to metabolic syndromes
could also be assessed [17].
Several potential hurdles hamper the application of ACC
inhibitors in humans. First, malonyl-CoA is important in
controlling insulin secretion in the pancreas. Reduction of
malonyl-CoA levels via ACC inhibition could offset beneficial effects on glucose-stimulated insulin secretion due to
reduction of pancreatic -cell fat contents [165-167]. It
remains to be determined whether ACC inhibition-induced
malonyl-CoA and subsequent fat content decrease in
pancreatic -cells adversely influence insulin secretion and
thus offsets beneficial effects in clinic [17]. Second, hypothalamic malonyl-CoA is a negative regulator of food intake
in feeding behavior [168], and thus reduction of malonylCoA in the hypothalamus may be undesirable. Consistent
with this observation, ACC2 knockout mice consume more
food even though they weigh less than wild-type animals
[161, 162], and Leptin-deficient Lepob/Lepob (ob/ob) mice
treated with CP-640186 increased food consumption concomitant with weight loss [68]. Therefore, weight reduction in
clinical trials with ACC inhibitors may occur together with
increased food consumption, and what is worse is that it
remains unknown whether this phenomenon counteracts the
positive metabolic effects of an ACC inhibitor, especially for
the inhibitors crossing the blood-brain barrier [169-172].
Third, studies in ex vivo working hearts suggest that elevated fatty acid oxidation during and after ischemia may
contribute to contractile dysfunction and increase ischemic
injury [173]. It remains to be understood whether ACC
180 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
inhibition could potentially increase myo-cardial injury after
an ischemic event for fatty acid oxidation enhanced by ACC
inhibition [17]. Obviously, these concerns need to be taken
into consideration in the future efforts to develop ACC inhibitors for the treatment of metabolic syndromes and cancers,
and more ACC inhibitors are being invented for high
efficiency and low toxicity [174-176].
Luo et al.
[5]
[6]
[7]
ABBREVIATIONS
ACCs
=
Acetyl-CoA carboxylase
AKR1B10
=
Aldo-keto reductase family 1 member
B10
AMPK
=
AMP-activated kinase
AR
=
Androgen receptor
BC
=
Biotin carboxylase
BCCP
=
Biotin carboxyl carrier protein
BRCA1
=
Breast cancer protein 1
C/EBP
=
CCAAT-enhancer-binding proteins
CPT-I
=
Carnitine palmitoyltransferase I
CT
=
Carboxyltransferase
FOXO
=
Forkhead box O
HMG-CoA =
3-Hydroxy-3-methylglutaryl coenzymeA
NRP1
=
Neuropilin-1
PKA
=
Protein kinase A
PPARs
=
Peroxisome-proliferation-activated receptors
SREBP-1
=
Sterol-regulatory-element binding protein 1
TOFA
=
Five-tetradecyloxy-2-furoic acid
ACKNOWLEDGEMENTS
We thank Dr. Zhangyong Li in Department of Laboratory
Medicine, the First People’s Hospital of Chenzhou for drawing chemical structures in ChemDraw in this article.
[8]
[9]
[10]
[11]
[12]
[13]
[14]
[15]
[16]
[17]
[18]
CONFLICT OF INTEREST
The authors declare no potential conflict of interest.
REFERENCES
[1]
[2]
[3]
[4]
Mojiminiyi OA, Al Mulla FAbdella NA. Which obesity index best
explains the link between adipokines, coronary heart disease risk
and metabolic abnormalities in type 2 diabetes mellitus? Med Princ
Pract 2009; 18: 123-9.
Rana JS, Nieuwdorp M, Jukema JW, Kastelein JJ. Cardiovascular
metabolic syndrome - an interplay of, obesity, inflammation,
diabetes and coronary heart disease. Diabetes Obes Metab 2007; 9:
218-32.
Wild SH, Byrne CD. ABC of obesity. Risk factors for diabetes and
coronary heart disease. BMJ 2006; 333: 1009-11.
Plecka Ostlund M, Marsk R, Rasmussen F, Lagergren JNaslund E.
Morbidity and mortality before and after bariatric surgery for
morbid obesity compared with the general population. Br J Surg
2011; 98: 811-6.
[19]
[20]
[21]
[22]
[23]
Losina E, Walensky RP, Reichmann WM, Holt HL, Gerlovin H,
Solomon DH, et al. Impact of obesity and knee osteoarthritis on
morbidity and mortality in older Americans. Ann Intern Med 2011;
154: 217-26.
Reilly JJ, Kelly J. Long-term impact of overweight and obesity in
childhood and adolescence on morbidity and premature mortality in
adulthood: Systematic review. Int J Obes (Lond) 2011; 35: 891-8.
Moore F, Brophy PJ. Regulation of acetyl-CoA carboxylase (ACC)
by ATP depletion in developing oligodendrocytes mimics the
action of AMP-activated protein kinase (AMPK). Biochem Soc
Trans 1994; 22: 416S.
Witters LA, Nordlund AC, Marshall L. Regulation of intracellular
acetyl-CoA carboxylase by ATP depletors mimics the action of the
5'-AMP-activated protein kinase. Biochem Biophys Res Commun
1991; 181: 1486-92.
Rabinkov AG, Amontov SV, Buneva VN, Tarussova NB.
Interaction of ATP with acetyl-CoA carboxylase from rat liver. The
role of the polyphosphate chain. Affinity labelling with alkylating
amides of ATP and ADP. Biochimie 1990; 72: 719-24.
Ohlrogge J, Browse J. Lipid biosynthesis. Plant Cell 1995; 7: 95770.
Boone AN, Rodrigues B, Brownsey RW. Multiple-site
phosphorylation of the 280kDa isoform of acetyl-CoA carboxylase
in rat cardiac myocytes: Evidence that cAMP-dependent protein
kinase mediates effects of beta-adrenergic stimulation. Biochem J
1999; 341 ( Pt 2): 347-54.
Gurvitz A. Physiological function of mycobacterial mtFabD, an
essential malonyl-CoA: AcpM transacylase of type 2 fatty acid
synthase FASII, in yeast mct1Delta cells. Comp Funct Genomics
2009: 836172.
Arthur CJ, Williams C, Pottage K, Ploskon E, Findlow SC, Burston
SG, et al. Structure and malonyl CoA-ACP transacylase binding of
Streptomyces coelicolor fatty acid synthase acyl carrier protein.
ACS Chem Biol 2009; 4: 625-36.
Huang YS, Ge J, Zhang HM, Lei JQ, Zhang XL, Wang HH.
Purification and characterization of the Mycobacterium
tuberculosis FabD2, a novel malonyl-CoA:AcpM transacylase of
fatty acid synthase. Protein Expr Purif 2006; 45: 393-9.
Thupari JN, Pinn ML, Kuhajda FP. Fatty acid synthase inhibition
in human breast cancer cells leads to malonyl-CoA-induced
inhibition of fatty acid oxidation and cytotoxicity. Biochem
Biophys Res Commun 2001; 285: 217-23.
Ventura FV, Costa CG, IJlst L, Dorland L, Duran M, Jakobs C,
et al. Broad specificity of carnitine palmitoyltransferase II towards
long-chain acyl-CoA beta-oxidation intermediates and its practical
approach to the synthesis of various long-chain acylcarnitines. J
Inherit Metab Dis 1997; 20: 423-6.
Tong L, Harwood HJ, Jr. Acetyl-coenzyme A carboxylases:
Versatile targets for drug discovery. J Cell Biochem 2006; 99:
1476-88.
Harwood HJ, Jr., Petras SF, Shelly LD, Zaccaro LM, Perry DA,
Makowski MR, et al. Isozyme-nonselective N-substituted
bipiperidylcarboxamide acetyl-CoA carboxylase inhibitors reduce
tissue malonyl-CoA concentrations, inhibit fatty acid synthesis, and
increase fatty acid oxidation in cultured cells and in experimental
animals. J Biol Chem 2003; 278: 37099-111.
Beckers A, Organe S, Timmermans L, Scheys K, Peeters A,
Brusselmans K, et al. Chemical inhibition of acetyl-CoA
carboxylase induces growth arrest and cytotoxicity selectively in
cancer cells. Cancer Res 2007; 67: 8180-7.
Diacovich L, Peiru S, Kurth D, Rodriguez E, Podesta F, Khosla C,
et al. Kinetic and structural analysis of a new group of Acyl-CoA
carboxylases found in Streptomyces coelicolor A3(2). J Biol Chem
2002; 277: 31228-36.
Rodriguez E, Banchio C, Diacovich L, Bibb MJ, Gramajo H. Role
of an essential acyl coenzyme A carboxylase in the primary and
secondary metabolism of Streptomyces coelicolor A3(2). Appl
Environ Microbiol 2001; 67: 4166-76.
Rodriguez E, Gramajo H. Genetic and biochemical characterization
of the alpha and beta components of a propionyl-CoA carboxylase
complex of Streptomyces coelicolor A3(2). Microbiology 1999;
145 ( Pt 11): 3109-19.
Chuakrut S, Arai H, Ishii MIgarashi Y. Characterization of a
bifunctional archaeal acyl coenzyme A carboxylase. J Bacteriol
2003; 185: 938-47.
Acetyl-CoA Carboxylase Inhibitors
[24]
[25]
[26]
[27]
[28]
[29]
[30]
[31]
[32]
[33]
[34]
[35]
[36]
[37]
[38]
[39]
[40]
[41]
[42]
[43]
[44]
[45]
[46]
Kiatpapan P, Kobayashi H, Sakaguchi M, Ono H, Yamashita M,
Kaneko Y, et al. Molecular characterization of Lactobacillus
plantarum genes for beta-ketoacyl-acyl carrier protein synthase III
(fabH) and acetyl coenzyme A carboxylase (accBCDA), which are
essential for fatty acid biosynthesis. Appl Environ Microbiol 2001;
67: 426-33.
Zhang H, Yang Z, Shen Y, Tong L. Crystal structure of the
carboxyltransferase domain of acetyl-coenzyme A carboxylase.
Science 2003; 299: 2064-7.
Leibundgut M, Maier T, Jenni S, Ban N. The multienzyme
architecture of eukaryotic fatty acid synthases. Curr Opin Struct
Biol 2008; 18: 714-25.
Maier T, Leibundgut M, Ban N. The crystal structure of a
mammalian fatty acid synthase. Science 2008; 321: 1315-22.
Artymiuk PJ, Poirrette AR, Rice DW, Willett P. Biotin carboxylase
comes into the fold. Nat Struct Biol 1996; 3: 128-32.
Galperin MY, Koonin EV. A diverse superfamily of enzymes with
ATP-dependent carboxylate-amine/thiol ligase activity. Protein Sci
1997; 6: 2639-43.
Thoden JB, Blanchard CZ, Holden HM, Waldrop GL. Movement
of the biotin carboxylase B-domain as a result of ATP binding. J
Biol Chem 2000; 275: 16183-90.
Waldrop GL, Rayment I, Holden HM. Three-dimensional structure
of the biotin carboxylase subunit of acetyl-CoA carboxylase.
Biochemistry 1994; 33: 10249-56.
Madauss KP, Burkhart WA, Consler TG, Cowan DJ, Gottschalk
WK, Miller AB, et al. The human ACC2 CT-domain C-terminus is
required for full functionality and has a novel twist. Acta
Crystallogr D Biol Crystallogr 2009; 65: 449-61.
Bengtsson C, Blaho S, Saitton DB, Brickmann K, Broddefalk J,
Davidsson O, et al. Design of small molecule inhibitors of acetylCoA carboxylase 1 and 2 showing reduction of hepatic malonylCoA levels in vivo in obese Zucker rats. Bioorg Med Chem 2011;
19: 3039-53.
Zhang H, Tweel B, Tong L. Molecular basis for the inhibition of
the carboxyltransferase domain of acetyl-coenzyme-A carboxylase
by haloxyfop and diclofop. Proc Natl Acad Sci USA 2004; 101:
5910-5.
Zhang H, Tweel B, Li J, Tong L. Crystal structure of the
carboxyltransferase domain of acetyl-coenzyme A carboxylase in
complex with CP-640186. Structure 2004; 12: 1683-91.
Tong, L., Zhang, H., Yang, Z. Carboxyltransferase domain of
acetyl-CoA carboxylase. US7884189 (2011).
Fall RR, Vagelos PR. Acetyl coenzyme A carboxylase. Molecular
forms and subunit composition of biotin carboxyl carrier protein. J
Biol Chem 1972; 247: 8005-15.
Nenortas EBeckett D. Purification and characterization of intact
and truncated forms of the Escherichia coli biotin carboxyl carrier
subunit of acetyl-CoA carboxylase. J Biol Chem 1996; 271: 755967.
Choi-Rhee E, Cronan JE. The biotin carboxylase-biotin carboxyl
carrier protein complex of Escherichia coli acetyl-CoA
carboxylase. J Biol Chem 2003; 278: 30806-12.
Swinnen JV, Ulrix W, Heyns W, Verhoeven G. Coordinate
regulation of lipogenic gene expression by androgens: Evidence for
a cascade mechanism involving sterol regulatory element binding
proteins. Proc Natl Acad Sci USA 1997; 94: 12975-80.
Lopez JM, Bennett MK, Sanchez HB, Rosenfeld JM, Osborne TF.
Sterol regulation of acetyl coenzyme A carboxylase: A mechanism
for coordinate control of cellular lipid. Proc Natl Acad Sci USA
1996; 93: 1049-53.
Barber MC, Price NT, Travers MT. Structure and regulation of
acetyl-CoA carboxylase genes of metazoa. Biochim Biophys Acta
2005; 1733: 1-28.
Liu QS, Pu L, Poo MM. Repeated cocaine exposure in vivo
facilitates LTP induction in midbrain dopamine neurons. Nature
2005; 437: 1027-31.
Talukdar S, Hillgartner FB. The mechanism mediating the
activation of acetyl-coenzyme A carboxylase-alpha gene
transcription by the liver X receptor agonist T0-901317. J Lipid
Res 2006; 47: 2451-61.
Hardie DG, Pan DA. Regulation of fatty acid synthesis and
oxidation by the AMP-activated protein kinase. Biochem Soc Trans
2002; 30: 1064-70.
Xiao CW, Wood C, Huang W, L'Abbe MR, Gilani GS, Cooke GM,
et al. Tissue-specific regulation of acetyl-CoA carboxylase gene
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
[47]
[48]
[49]
[50]
[51]
[52]
[53]
[54]
[55]
[56]
[57]
[58]
[59]
[60]
[61]
[62]
[63]
[64]
[65]
[66]
[67]
181
expression by dietary soya protein isolate in rats. Br J Nutr 2006;
95: 1048-54.
Cheng HL, Ji NJ, Peng YX, Shen X, Xu JH, Dong ZG, et al.
Molecular characterization and tissue-specific expression of the
acetyl-CoA carboxylase alpha gene from Grass carp,
Ctenopharyngodon idella. Gene 2011; 487: 46-51.
Luo DX, Peng XH, Xiong Y, Liao DF, Cao D, Li L. Dual role of
insulin-like growth factor-1 in acetyl-CoA carboxylase-alpha
activity in human colon cancer cells HCT-8: Downregulating its
expression and phosphorylation. Mol Cell Biochem 2011; 357:
255-62.
Li ZG, Yin WB, Song LY, Chen YH, Guan RZ, Wang JQ, et al.
Genes encoding the biotin carboxylase subunit of acetyl-CoA
carboxylase from Brassica napus and parental species: Cloning,
expression patterns, and evolution. Genome 2011; 54: 202-11.
Shi X, Liu S, Metges CC, Seyfert HM. C/EBP-beta drives
expression of the nutritionally regulated promoter IA of the acetylCoA carboxylase-alpha gene in cattle. Biochim Biophys Acta
2010; 1799: 561-7.
Li ZG, Yin WB, Guo H, Song LY, Chen YH, Guan RZ, et al.
Genes encoding the alpha-carboxyltransferase subunit of acetylCoA carboxylase from Brassica napus and parental species:
Cloning, expression patterns, and evolution. Genome 2010; 53:
360-70.
Chen XJ, Mao HL, Ma XM, Liu JX. Effects of dietary corn oil and
vitamin E supplementation on fatty acid profiles and expression of
acetyl CoA carboxylase and stearoyl-CoA desaturase gene in Hu
sheep. Anim Sci J 2010; 81: 165-71.
Yao N, Lu CL, Zhao JJ, Xia HF, Sun DG, Shi XQ, et al. A network
of miRNAs expressed in the ovary are regulated by FSH. Front
Biosci 2009; 14: 3239-45.
Kim CW, Moon YA, Park SW, Cheng D, Kwon HJ, Horton JD.
Induced polymerization of mammalian acetyl-CoA carboxylase by
MIG12 provides a tertiary level of regulation of fatty acid
synthesis. Proc Natl Acad Sci USA 2010; 107: 9626-31.
Kahn BB, Alquier T, Carling D, Hardie DG. AMP-activated
protein kinase: Ancient energy gauge provides clues to modern
understanding of metabolism. Cell Metab 2005; 1: 15-25.
Kemp BE, Stapleton D, Campbell DJ, Chen ZP, Murthy S, Walter
M, et al. AMP-activated protein kinase, super metabolic regulator.
Biochem Soc Trans 2003; 31: 162-8.
Wong GW, Wang J, Hug C, Tsao TS, Lodish HF. A family of
Acrp30/adiponectin structural and functional paralogs. Proc Natl
Acad Sci USA 2004; 101: 10302-7.
Munday MR. Regulation of mammalian acetyl-CoA carboxylase.
Biochem Soc Trans 2002; 30: 1059-64.
Magnard C, Bachelier R, Vincent A, Jaquinod M, Kieffer S, Lenoir
GM, et al. BRCA1 interacts with acetyl-CoA carboxylase through
its tandem of BRCT domains. Oncogene 2002; 21: 6729-39.
Shen Y, Tong L. Structural evidence for direct interactions between
the BRCT domains of human BRCA1 and a phospho-peptide from
human ACC1. Biochemistry 2008; 47: 5767-73.
Cao D, Fan ST, Chung SS. Identification and characterization of a
novel human aldose reductase-like gene. J Biol Chem 1998; 273:
11429-35.
Zu X, Yan R, Robbins S, Krishack PA, Liao DF, Cao D. Reduced
293T cell susceptibility to acrolein due to aldose reductase-like-1
protein expression. Toxicol Sci 2007; 97: 562-8.
Yan R, Zu X, Ma J, Liu Z, Adeyanju M, Cao D. Aldo-keto
reductase family 1 B10 gene silencing results in growth inhibition
of colorectal cancer cells: Implication for cancer intervention. Int J
Cancer 2007; 121: 2301-6.
Wang C, Yan R, Luo D, Watabe K, Liao DF, Cao D. Aldo-keto
reductase family 1 member B10 promotes cell survival by
regulating lipid synthesis and eliminating carbonyls. J Biol Chem
2009; 284: 26742-8.
Ma J, Yan R, Zu X, Cheng JM, Rao K, Liao DF, et al. Aldo-keto
reductase family 1 B10 affects fatty acid synthesis by regulating the
stability of acetyl-CoA carboxylase-alpha in breast cancer cells. J
Biol Chem 2008; 283: 3418-23.
Moreau K, Dizin E, Ray H, Luquain C, Lefai E, Foufelle F, et al.
BRCA1 affects lipid synthesis through its interaction with acetylCoA carboxylase. J Biol Chem 2006; 281: 3172-81.
Grantham BD, Zammit VA. Binding of [14C]malonyl-CoA to rat
liver mitochondria after blocking of the active site of carnitine
182 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
[68]
[69]
[70]
[71]
[72]
[73]
[74]
[75]
[76]
[77]
[78]
[79]
[80]
[81]
[82]
[83]
[84]
[85]
[86]
[87]
[88]
palmitoyltransferase I. Displacement of low-affinity binding by
palmitoyl-CoA. Biochem J 1986; 233: 589-93.
Harwood HJ, Jr. Treating the metabolic syndrome: Acetyl-CoA
carboxylase inhibition. Expert Opin Ther Targets 2005; 9: 267-81.
Castelo-Branco PA, Rubinger MM, Alves Lde C, de Barros PM,
Pereira SG, de Melo VJ, et al. Synthesis and antifungal activity of
aromatic bis-gamma-lactones analogous to avenaciolide. Chem
Biodivers 2007; 4: 2745-54.
Yu CM, Youn J, Jung J. Asymmetric sequential allylic transfer
reaction for the synthesis of 2-(1-stannylvinyl)-1,3-diols: Concise
synthesis of (-)-avenaciolide and (-)-isoavenaciolide. Angew Chem
Int Ed Engl 2006; 45: 1553-6.
Castelo-Branco PA, Rubinger MM, Guilardi S, Leite VM, dos
Santos AR, de C Alves L, et al. Synthesis, characterization,
absolute structural determination and antifungal activity of a new
chlorinated aromatic avenaciolide analogue. Pest Manag Sci 2009;
65: 34-40.
Levert KL, Waldrop GL, Stephens JM. A biotin analog inhibits
acetyl-CoA carboxylase activity and adipogenesis. J Biol Chem
2002; 277: 16347-50.
Gerth K, Pradella S, Perlova O, Beyer S, Muller R. Myxobacteria:
Proficient producers of novel natural products with various
biological activities--past and future biotechnological aspects with
the focus on the genus Sorangium. J Biotechnol 2003; 106: 233-53.
Gerth K, Bedorf N, Irschik H, Hofle G, Reichenbach H. The
soraphens: A family of novel antifungal compounds from
Sorangium cellulosum (Myxobacteria). I. Soraphen A1 alpha:
Fermentation, isolation, biological properties. J Antibiot (Tokyo)
1994; 47: 23-31.
Anderson, R., Hokama, T., Lee, S., Oey, R., Elich, T., Breazeale, S.
Modulators of acetyl-coenzyme A carboxylase and methods of use
thereof. US20080200461 (2008).
Srtrongin, R.M., Saraiva, M.C., Waldrop, G.L., Amspacher, D.R.
Derivatized biotin compounds and methods of use. US6242610
(2001).
Waldrop, G.L., Stephens, J.M., Levert, K.L. Inhibition of the
carboxyltransferase component of acetyl-CoA carboxylase, and the
use of such inhibition in anti-cancer and anti-lipogenic therapies.
US6485941(2002).
Brunner, N., Freiberg, C., Lampe, T., Newton, B., Otteneder, M.,
Pernerstorfer, J., Pohlmann, J., Schiffer, G., Shimada, M.
Derivatives of andrimide and moiramide B having antibacterial
properties. US7544709 (2009).
Brunner, N., Freiberg, C., Lampe, T., Newton, B., Otteneder, M.,
Pernerstorfer, J., Pohlmann, J., Schiffer, G., Shimada, M.
Derivatives of andrimide and moiramide b having antibacterial
properties. US20050080129 (2005).
Wellen, C.W. Use of curcumin and analogues thereof as inhibitors
of ACC2. US20050267221 (2005).
Lee YK, Park SY, Kim YM, Park OJ. Regulatory effect of the
AMPK-COX-2 signaling pathway in curcumin-induced apoptosis
in HT-29 colon cancer cells. Ann N Y Acad Sci 2009; 1171: 48994.
Kim T, Davis J, Zhang AJ, He X, Mathews ST. Curcumin activates
AMPK and suppresses gluconeogenic gene expression in hepatoma
cells. Biochem Biophys Res Commun 2009; 388: 377-82.
Pan W, Yang H, Cao C, Song X, Wallin B, Kivlin R, et al. AMPK
mediates curcumin-induced cell death in CaOV3 ovarian cancer
cells. Oncol Rep 2008; 20: 1553-9.
Naito J, Sasaki E, Ohta Y, Shinohara R, Ishiguro I. Anthranilic acid
metabolism in the isolated perfused rat liver: Detection and
determination of anthranilic acid and its related substances using
high-performance liquid chromatography with electrochemical
detection. Biochem Pharmacol 1984; 33: 3195-200.
Braun K, Gibson DT. Anaerobic degradation of 2-aminobenzoate
(anthranilic acid) by denitrifying bacteria. Appl Environ Microbiol
1984; 48: 102-7.
Ikawa, H.M., Nishimura, K., Okada, T., Nakamura, N., Inaba, K.,
Kojima, H., Kosono, H. Hypoglycemic agent. US20030191323
(2003).
Gobbi, L.C., Gubler, M., Neidhart, W., Nettekoven, M.H.
Benzodioxepine derivatives. US7459480 (2008).
Clark RF, Zhang T, Wang X, Wang R, Zhang X, Camp HS, et al.
Phenoxy thiazole derivatives as potent and selective acetyl-CoA
carboxylase 2 inhibitors: Modulation of isozyme selectivity by
Luo et al.
[89]
[90]
[91]
[92]
[93]
[94]
[95]
[96]
[97]
[98]
[99]
[100]
[101]
[102]
[103]
[104]
[105]
[106]
[107]
.[108]
[109]
[110]
[111]
incorporation of phenyl ring substituents. Bioorg Med Chem Lett
2007; 17: 1961-5.
Gu YG, Weitzberg M, Clark RF, Xu X, Li Q, Zhang T, et al.
Synthesis and structure-activity relationships of N-{3-[2-(4alkoxyphenoxy)thiazol-5-yl]-1methylprop-2-ynyl}carboxy
derivatives as selective acetyl-CoA carboxylase 2 inhibitors. J Med
Chem 2006; 49: 3770-3.
Clark RF, Zhang T, Xin Z, Liu G, Wang Y, Hansen TM, et al.
Structure-activity relationships for a novel series of thiazolyl
phenyl ether derivatives exhibiting potent and selective acetyl-CoA
carboxylase 2 inhibitory activity. Bioorg Med Chem Lett 2006; 16:
6078-81.
Xu X, Weitzberg M, Keyes RF, Li Q, Wang R, Wang X, et al. The
synthesis and structure-activity relationship studies of selective
acetyl-CoA carboxylase inhibitors containing 4-(thiazol-5-yl)but-3yn-2-amino motif: Polar region modifications. Bioorg Med Chem
Lett 2007; 17: 1803-7.
Gu YG, Weitzberg M, Clark RF, Xu X, Li Q, Lubbers NL, et al. N{3-[2-(4-Alkoxyphenoxy)thiazol-5-yl]-1-methylprop-2ynyl}carboxy derivatives as acetyl-coA carboxylase inhibitors-improvement of cardiovascular and neurological liabilities via
structural modifications. J Med Chem 2007; 50: 1078-82.
Henderson, S.T. Inhibitors of acetyl-CoA carboxylase for treatment
of neuronal hypometabolism. US20110003767 (2011).
Dang, Q., Gibson, T.S., Jiang, H., Chuang, D.M., Bao, J., Jiang, J.,
Kassick, A., Ketec, A., Lan, P., Lu, H., Makara, G.M., Romero,
A.F., Sebhat, I., Wilson, D., Wodka, D., Bookser, B.C. Novel
cyclic benzimidazole derivatives useful as anti-diabetic agents.
US20110263533 (2011).
Ohmori K, Yamada H, Yasuda A, Yamamoto A, Matsuura N,
Kiniwa M. Anti-hyperlipidemic action of a newly synthesized
benzoic acid derivative, S-2E. Eur J Pharmacol 2003; 471: 69-76.
Ohmori K, Yamada H, Yasuda A, Yamamoto A, Matsuura N,
Kiniwa M. Effects of a novel antihyperlipidemic agent, S-2E, on
the blood lipid abnormalities in homozygous WHHL rabbits.
Metabolism 2004; 53: 680-5.
Ohmori K, Yamada H, Yasuda A, Yamamoto A, Matsuura N,
Kiniwa M. Effects of a novel anti-hyperlipidemic agent, S-2E, on
blood lipid levels in rats with fructose-induced hypertriglyceridemia. Pharmacology 2004; 72: 240-6.
Zuther E, Johnson JJ, Haselkorn R, McLeod R, Gornicki P. Growth
of Toxoplasma gondii is inhibited by aryloxyphenoxypropionate
herbicides targeting acetyl-CoA carboxylase. Proc Natl Acad Sci
USA 1999; 96: 13387-92.
Alban C, Job D, Douce R. Biotin metabolism in plants. Annu Rev
Plant Physiol Plant Mol Biol 2000; 51: 17-47.
Nikolau BJ, Ohlrogge JB, Wurtele ES. Plant biotin-containing
carboxylases. Arch Biochem Biophys 2003; 414: 211-22.
Brunner, H. Cyclohexenonecarboxylic acid derivatives with
herbicidal and plant growth regulating properties. US4640706
(1987).
Munger, J., Bennett, B., Shenk, T., Rabinowitz, J. Treatment of
viral infections by modulation of host cell metabolic pathways.
US20090239830 (2009).
Fukatsu, K., Kamata, M., Yamashita, T., Endo, S. Nitrogencontaining heterocyclic compound. WO2007119833 (2007).
Chonan T, Oi T, Yamamoto D, Yashiro M, Wakasugi D, Tanaka
H, et al. (4-Piperidinyl)-piperazine: A new platform for acetyl-CoA
carboxylase inhibitors. Bioorg Med Chem Lett 2009; 19: 6645-8.
Perry, D.A., Harwood, H.J. ACC inhibitors. WO03072197 (2003).
Fukatsu, K., Kamata, M., Yamashita, T. Heterocyclic compound.
EP2123652 (2009).
Corbett, J.W., Elliott, R.L., Bell, A. Spiroketone acetyl-CoA
carboxylase inhibitors. WO08065508 (2008).
Iino, T., Jona, H., Kurihara, H., Nakamura, M., Niiyama, K., Shibata, J., Shimamura, T., Watanabe, H., Yamakawa, T., Yang, L.
Spirochromanon derivatives. WO2008088692 (2008).
Iino, T., Jona, H., Shibata, J., Shimamura, T., Yamakawa, T., Yang,
L. Substituted spirochromanone derivatives as ACC inhibitors.
WO2008088688 (2008).
Corbett JW, Freeman-Cook KD, Elliott R, Vajdos F, Rajamohan F,
Kohls D, et al. Discovery of small molecule isozyme non-specific
inhibitors of mammalian acetyl-CoA carboxylase 1 and 2. Bioorg
Med Chem Lett 2010; 20: 2383-8.
Duennenberger, M., Schellenbaum, M. Furan-2,5-dicarboxylic acid
diamides. US3546255 (1970).
Acetyl-CoA Carboxylase Inhibitors
[112]
[113]
[114]
[115]
[116]
[117]
[118]
[119]
[120]
[121]
[122]
[123]
[124]
[125]
[126]
[127]
[128]
[129]
[130]
[131]
[132]
[133]
Waldrop, G.L., Stephens, J.M., Levert, K.L. Inhibition of the
carboxyltransferase component of acetyl-CoA carboxylase, and the
use of such inhibition in anti-cancer and anti-lipogenic therapies.
US6485941(2002).
Yamakawa, T., Jona, H., Niiyama, K., Yamada, K., Lino, T.,
Ohkubo, M., Imamura, H., Shibata, J., Kusunoki, J., Yang, L.
Spirochromanone derivatives as acetyl coenzyme A carboxylase
(ACC) inhibitors. US7935712 (2011).
Jona, H., Shibata, Y., Yamakawa, T. Novel spirochromanone
carboxylic acids. US20110077262 (2011).
Corbett, J.W., Elliott, R., Freeman-cook, K., Griffith, D.A.,
Philipion, D.P. Pyrazolospiroketone acetyl-CoA carboxylase
inhibitors. US20110028390 (2011).
Ligon J, Hill S, Beck J, Zirkle R, Molnar I, Zawodny J, et al.
Characterization of the biosynthetic gene cluster for the antifungal
polyketide soraphen A from Sorangium cellulosum So ce26. Gene
2002; 285: 257-67.
Shen Y, Volrath SL, Weatherly SC, Elich TD, Tong L. A
mechanism for the potent inhibition of eukaryotic acetyl-coenzyme
A carboxylase by soraphen A, a macrocyclic polyketide natural
product. Mol Cell 2004; 16: 881-91.
Tong L. Acetyl-coenzyme A carboxylase: Crucial metabolic
enzyme and attractive target for drug discovery. Cell Mol Life Sci
2005; 62: 1784-803.
Vahlensieck HF, Pridzun L, Reichenbach H, Hinnen A.
Identification of the yeast ACC1 gene product (acetyl-CoA
carboxylase) as the target of the polyketide fungicide soraphen A.
Curr Genet 1994; 25: 95-100.
Weatherly SC, Volrath SL, Elich TD. Expression and
characterization of recombinant fungal acetyl-CoA carboxylase and
isolation of a soraphen-binding domain. Biochem J 2004; 380: 10510.
Belkebir A, De Paepe R, Tremolieres A, Aid F, Benhassaine-Kesri
G. Sethoxydim affects lipid synthesis and acetyl-CoA carboxylase
activity in soybean. J Exp Bot 2006; 57: 3553-62.
Halvorson DL, McCune SA. Inhibition of fatty acid synthesis in
isolated adipocytes by 5-(tetradecyloxy)-2-furoic acid. Lipids 1984;
19: 851-6.
Otto DA, Chatzidakis C, Kasziba E, Cook GA. Reciprocal effects
of 5-(tetradecyloxy)-2-furoic acid on fatty acid oxidation. Arch
Biochem Biophys 1985; 242: 23-31.
Malewiak MI, Griglio S, Le Liepvre X. Relationship between
lipogenesis, ketogenesis, and malonyl-CoA content in isolated
hepatocytes from the obese Zucker rat adapted to a high-fat diet.
Metabolism 1985; 34: 604-11.
Sola MM, Oliver FJ, Salto R, Gutierrez M, Vargas A. Citrate
inhibition of rat-kidney cortex phosphofructokinase. Mol Cell
Biochem 1994; 135: 123-8.
Wang C, Xu C, Sun M, Luo D, Liao DF, Cao D. Acetyl-CoA
carboxylase-alpha inhibitor TOFA induces human cancer cell
apoptosis. Biochem Biophys Res Commun 2009; 385: 302-6.
Zhou W, Simpson PJ, McFadden JM, Townsend CA, Medghalchi
SM, Vadlamudi A, et al. Fatty acid synthase inhibition triggers
apoptosis during S phase in human cancer cells. Cancer Res 2003;
63: 7330-7.
Pizer ES, Thupari J, Han WF, Pinn ML, Chrest FJ, Frehywot GL,
et al. Malonyl-coenzyme-A is a potential mediator of cytotoxicity
induced by fatty-acid synthase inhibition in human breast cancer
cells and xenografts. Cancer Res 2000; 60: 213-8.
Zhou W, Han WF, Landree LE, Thupari JN, Pinn ML, Bililign T,
et al. Fatty acid synthase inhibition activates AMP-activated
protein kinase in SKOV3 human ovarian cancer cells. Cancer Res
2007; 67: 2964-71.
Liu X, Fortin PD, Walsh CT. Andrimid producers encode an
acetyl-CoA carboxyltransferase subunit resistant to the action of
the antibiotic. Proc Natl Acad Sci USA 2008; 105: 13321-6.
Pohlmann J, Lampe T, Shimada M, Nell PG, Pernerstorfer J,
Svenstrup N, et al. Pyrrolidinedione derivatives as antibacterial
agents with a novel mode of action. Bioorg Med Chem Lett 2005;
15: 1189-92.
Campbell JW, Cronan JE, Jr. Bacterial fatty acid biosynthesis:
targets for antibacterial drug discovery. Annu Rev Microbiol 2001;
55: 305-32.
Freiberg C, Brunner NA, Schiffer G, Lampe T, Pohlmann J, Brands
M, et al. Identification and characterization of the first class of
Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
[134]
[135]
[136]
[137]
[138]
[139]
[140]
[141]
[142]
[143]
[144]
[145]
[146]
[147]
[148]
[149]
[150]
[151]
[152]
[153]
[154]
[155]
183
potent bacterial acetyl-CoA carboxylase inhibitors with
antibacterial activity. J Biol Chem 2004; 279: 26066-73.
Solbiati J, Chapman-Smith A, Cronan JE, Jr. Stabilization of the
biotinoyl domain of Escherichia coli acetyl-CoA carboxylase by
interactions between the attached biotin and the protruding "thumb"
structure. J Biol Chem 2002; 277: 21604-9.
Cramer CT, Goetz B, Hopson KL, Fici GJ, Ackermann RM, Brown
SC, et al. Effects of a novel dual lipid synthesis inhibitor and its
potential utility in treating dyslipidemia and metabolic syndrome. J
Lipid Res 2004; 45: 1289-301.
Ohno T, Yano S, Yamada H, Shirasaka T, Yamamoto A,
Kobayashi K, et al. Synthesis of the optical isomers of 4-[1-(4-tertbutylphenyl)-2-oxo- pyrrolidine-4-yl]methyloxybenzoic acid (S-2)
and their biological evaluation as antilipidemic agent. Chem Pharm
Bull (Tokyo) 1999; 47: 1549-54.
Berg IA, Ivanovskii RN. [Enzymes of the citramalate cycle in
Rhodospirillum rubrum]. Mikrobiologiia 2009; 78: 22-31.
Jin M, Fischbach MA, Clardy J. A biosynthetic gene cluster for the
acetyl-CoA carboxylase inhibitor andrimid. J Am Chem Soc 2006;
128: 10660-1.
Zhao L, Chen YX, Varghese Z, Huang AL, Tang RK, Jia B, et al.
Murine gamma herpes virus 68 infection promotes fatty liver
formation and hepatic insulin resistance in C57BL/6J mice.
Hepatol Int 2011 [Epub ahead of print].
Sirtori CR, Galli C, Franceschini G. Fraudulent (and non
fraudulent) fatty acids for human health. Eur J Clin Invest 1993;
23: 686-9.
Yamashita, T., Kamata, M., Hirose, H., Murakami, M., Fujimoto,
T., Ikeda, Z., Yasuma, T., Fujimori, I., Mizojiri, R., Yukawa, T.
Bicyclic compound. US20110263562 (2011).
McCune SA, Harris RA. Mechanism responsible for 5(tetradecyloxy)-2-furoic acid inhibition of hepatic lipogenesis. J
Biol Chem 1979; 254: 10095-101.
Parker RA, Kariya T, Grisar JM, Petrow V. 5-(Tetradecyloxy)-2furancarboxylic acid and related hypolipidemic fatty acid-like
alkyloxyarylcarboxylic acids. J Med Chem 1977; 20: 781-91.
Bar-Tana J, Ben-Shoshan S, Blum J, Migron Y, Hertz R, Pill J,
et al. Synthesis and hypolipidemic and antidiabetogenic activities
of beta,beta,beta',beta'-tetrasubstituted, long-chain dioic acids. J
Med Chem 1989; 32: 2072-84.
Tzur R, Rose-Kahn G, Adler JH, Bar-Tana J. Hypolipidemic,
antiobesity, and hypoglycemic-hypoinsulinemic effects of
beta,beta'-methyl-substituted hexadecanedioic acid in sand rats.
Diabetes 1988; 37: 1618-24.
Russell JC, Amy RM, Graham SE, Dolphin PJ, Wood GO, BarTana J. Inhibition of atherosclerosis and myocardial lesions in the
JCR:LA-cp rat by beta, beta'-tetramethylhexadecanedioic acid
(MEDICA 16). Arterioscler Thromb Vasc Biol 1995; 15: 918-23.
Berge RK, Skorve J, Tronstad KJ, Berge K, Gudbrandsen OA,
Grav H. Metabolic effects of thia fatty acids. Curr Opin Lipidol
2002; 13: 295-304.
Skrede S, Sorensen HN, Larsen LN, Steineger HH, Hovik K,
Spydevold OS, et al. Thia fatty acids, metabolism and metabolic
effects. Biochim Biophys Acta 1997; 1344: 115-31.
Bremer J. The biochemistry of hypo- and hyperlipidemic fatty acid
derivatives: Metabolism and metabolic effects. Prog Lipid Res
2001; 40: 231-68.
Menendez JA, Colomer R, Lupu R. Why does tumor-associated
fatty acid synthase (oncogenic antigen-519) ignore dietary fatty
acids? Med Hypotheses 2005; 64: 342-9.
Chajes V, Lanson M, Fetissof F, Lhuillery C, Bougnoux P.
Membrane fatty acids of breast carcinoma: Contribution of host
fatty acids and tumor properties. Int J Cancer 1995; 63: 169-75.
Abramson HN. The lipogenesis pathway as a cancer target. J Med
Chem 2011; 54: 5615-38.
Wang C, Rajput S, Watabe K, Liao DF, Cao D. Acetyl-CoA
carboxylase-a as a novel target for cancer therapy. Front Biosci
(Schol Ed) 2010; 2: 515-26.
Chajes V, Cambot M, Moreau K, Lenoir GM, Joulin V. AcetylCoA carboxylase alpha is essential to breast cancer cell survival.
Cancer Res 2006; 66: 5287-94.
Brusselmans K, De Schrijver E, Verhoeven G, Swinnen JV. RNA
interference-mediated silencing of the acetyl-CoA-carboxylasealpha gene induces growth inhibition and apoptosis of prostate
cancer cells. Cancer Res 2005; 65: 6719-25.
184 Recent Patents on Anti-Cancer Drug Discovery, 2012, Vol. 7, No. 2
[156]
[157]
[158]
[159]
[160]
[161]
[162]
[163]
[164]
[165]
[166]
Sinilnikova OM, Ginolhac SM, Magnard C, Leone M, Anczukow
O, Hughes D, et al. Acetyl-CoA carboxylase alpha gene and breast
cancer susceptibility. Carcinogenesis 2004; 25: 2417-24.
Guseva NV, Rokhlin OW, Glover RA, Cohen MB. TOFA (5Tetradecyl-oxy-2-furoic acid) reduces fatty acid synthesis, inhibits
expression of AR, neuropilin-1 and Mcl-1 and kills prostate cancer
cells independent of p53 status. Cancer Biol Ther 2011; 12: 80-5.
Jump DB, Torres-Gonzalez M, Olson LK. Soraphen A, an inhibitor
of acetyl CoA carboxylase activity, interferes with fatty acid
elongation. Biochem Pharmacol 2011; 81: 649-60.
Abu-Elheiga L, Matzuk MM, Kordari P, Oh W, Shaikenov T, Gu
Z, et al. Mutant mice lacking acetyl-CoA carboxylase 1 are
embryonically lethal. Proc Natl Acad Sci USA 2005; 102: 12011-6.
Olson DP, Pulinilkunnil T, Cline GW, Shulman GI, Lowell BB.
Gene knockout of ACC2 has little effect on body weight, fat mass,
or food intake. Proc Natl Acad Sci USA 2010; 107: 7598-603.
Abu-Elheiga L, Matzuk MM, Abo-Hashema KA, Wakil SJ.
Continuous fatty acid oxidation and reduced fat storage in mice
lacking acetyl-CoA carboxylase 2. Science 2001; 291: 2613-6.
Abu-Elheiga L, Oh W, Kordari P, Wakil SJ. Acetyl-CoA
carboxylase 2 mutant mice are protected against obesity and
diabetes induced by high-fat/high-carbohydrate diets. Proc Natl
Acad Sci USA 2003; 100: 10207-12.
Oh W, Abu-Elheiga L, Kordari P, Gu Z, Shaikenov T, Chirala SS,
et al. Glucose and fat metabolism in adipose tissue of acetyl-CoA
carboxylase 2 knockout mice. Proc Natl Acad Sci USA 2005; 102:
1384-9.
Savage DB, Choi CS, Samuel VT, Liu ZX, Zhang D, Wang A,
et al. Reversal of diet-induced hepatic steatosis and hepatic insulin
resistance by antisense oligonucleotide inhibitors of acetyl-CoA
carboxylases 1 and 2. J Clin Invest 2006; 116: 817-24.
Zhang S, Kim KH. Essential role of acetyl-CoA carboxylase in the
glucose-induced insulin secretion in a pancreatic beta-cell line. Cell
Signal 1998; 10: 35-42.
Zhang S, Kim KH. Glucose activation of acetyl-CoA carboxylase
in association with insulin secretion in a pancreatic beta-cell line. J
Endocrinol 1995; 147: 33-41.
Luo et al.
[167]
[168]
[169]
[170]
[171]
[172]
[173]
[174]
[175]
[176]
Chen J, Jeppesen PB, Nordentoft I, Hermansen K. Stevioside
improves pancreatic beta-cell function during glucotoxicity via
regulation of acetyl-CoA carboxylase. Am J Physiol Endocrinol
Metab 2007; 292: E1906-16.
Lane MD, Hu Z, Cha SH, Dai Y, Wolfgang M, Sidhaye A. Role of
malonyl-CoA in the hypothalamic control of food intake and
energy expenditure. Biochem Soc Trans 2005; 33: 1063-7.
Lane MD, Wolfgang M, Cha SH, Dai Y. Regulation of food intake
and energy expenditure by hypothalamic malonyl-CoA. Int J Obes
(Lond) 2008; 32 Suppl 4: S49-54.
Cesquini M, Stoppa GR, Prada PO, Torsoni AS, Romanatto T,
Souza A, et al. Citrate diminishes hypothalamic acetyl-CoA
carboxylase phosphorylation and modulates satiety signals and
hepatic mechanisms involved in glucose homeostasis in rats. Life
Sci 2008; 82: 1262-71.
Wolfgang MJ, Cha SH, Sidhaye A, Chohnan S, Cline G, Shulman
GI, et al. Regulation of hypothalamic malonyl-CoA by central
glucose and leptin. Proc Natl Acad Sci USA 2007; 104: 19285-90.
Gao S, Kinzig KP, Aja S, Scott KA, Keung W, Kelly S, et al.
Leptin activates hypothalamic acetyl-CoA carboxylase to inhibit
food intake. Proc Natl Acad Sci USA 2007; 104: 17358-63.
Sambandam N, Lopaschuk GD. AMP-activated protein kinase
(AMPK) control of fatty acid and glucose metabolism in the
ischemic heart. Prog Lipid Res 2003; 42: 238-56.
Defossa, E., Keil, S., Dietrich, V., Stengelin, S., Herling, A.,
Haschke, G., Klabunde, T. (2-aryloxyacetylamino) phenylpropionic
acid derivatives, processes for preparation thereof and use thereof
as medicaments. US20120004167 (2012).
Keli, S., Defossa, E., Dietrich, V., Stengelin, S., Herling, A.,
Haschke, G., Klabunde, T. Heterocyclically substituted methoxyphenyl derivatives with an oxo group, processes for preparation
thereof and use thereof as medicaments. US20120004165 (2012).
Keil, S., Defossa, E., Dietrich, V., Stengelin, S., Herling, A.,
Haschke, G., Klabunde, T. Aryloxyalkylene-substituted hydroxyphenylhexynoic acids, process for preparation thereof and use
thereof as a medicament. US20120004166 (2012).