Accepted Manuscript Discovery of aliphatic-chain hydroxamates containing indole derivatives with potent class I histone deacetylase inhibitory activities Shi-Wei Chao, Liang-Chieh Chen, Chia-Chun Yu, Chang-Yi Liu, Tony Eight Lin, JihHwa Guh, Chen-Yu Wang, Chun-Yung Chen, Kai-Cheng Hsu, Wei-Jan Huang PII:
S0223-5234(17)30995-9
DOI:
10.1016/j.ejmech.2017.11.092
Reference:
EJMECH 9967
To appear in:
European Journal of Medicinal Chemistry
Received Date: 1 August 2017 Revised Date:
11 November 2017
Accepted Date: 28 November 2017
Please cite this article as: S.-W. Chao, L.-C. Chen, C.-C. Yu, C.-Y. Liu, T.E. Lin, J.-H. Guh, C.-Y. Wang, C.-Y. Chen, K.-C. Hsu, W.-J. Huang, Discovery of aliphatic-chain hydroxamates containing indole derivatives with potent class I histone deacetylase inhibitory activities, European Journal of Medicinal Chemistry (2018), doi: 10.1016/j.ejmech.2017.11.092. This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
AC C
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
ACCEPTED MANUSCRIPT
Discovery of aliphatic-chain hydroxamates containing indole derivatives with potent class I histone deacetylase inhibitory activities Shi-Wei Chaoa,b, Liang-Chieh Chena,b, Chia-Chun Yuc, Chang-Yi Liud, Tony Eight Lind, Jih-Hwa Guhc, Chen-Yu Wange, Chun-Yung Chenb, Kai-Cheng Hsud*, Wei-Jan Huangb,f,g,h∗ School of Pharmacy, Taipei Medical University, Taipei, Taiwan
b
Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
c
School of Pharmacy, National Taiwan University, Taipei, Taiwan
d
Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
RI PT
a
TaiMed Biologics Inc., Taipei, Taiwan
f
School of Pharmacy, National Defense Medical Center, Taipei, Taiwan
g
SC
e
Ph.D. Program for the Clinical Drug Discovery from Botanical Herbs, Taipei Medical
h
M AN U
University, Taipei, Taiwan
Ph.D. Program in Biotechnology Research and Development, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
Abstract
Histone deacetylase (HDAC) is a validated drug target for various diseases. This study combined indole recognition cap with SAHA, an FDA-approved HDAC inhibitor used to treat cutaneous T-cell
TE D
lymphoma (CTCL). The structure activity relationship of the resulting compounds that inhibited HDAC was disclosed as well. Some compounds exhibited much stronger inhibitory activities than SAHA. We identified two meta-series compounds 6j and 6k with a two-carbon linker had IC50 values of 3.9 and 4.5 nM for HDAC1, respectively. In contrast, the same oriented compounds with longer
EP
carbon chain linkers showed weaker inhibition. The result suggests that the linker chain length greatly contributed to enzyme inhibitory potency. In addition, comparison of enzyme-inhibiting activity
AC C
between the compounds and SAHA showed that compounds 6j and 6k displayed higher inhibiting activity for class I (HDAC1, -2, -3 and -8). The molecular docking and structure analysis revealed structural differences with the inhibitor cap and metal-binding regions between the HDAC isozymes that affect interactions with the inhibitors and play a key role for selectivity. Further biological evaluation showed multiple cellular effects associated with compounds 6j- and 6k-induced HDAC inhibitory activity. Keywords: Histone deacetylase; Structure activity relationship; Indole; Isozyme; Molecular docking
*Corresponding authors E-mail:
[email protected] (W.-J. Huang) E-mail:
[email protected] (K.-C. Hsu) 1
ACCEPTED MANUSCRIPT
1. Introduction
RI PT
Reversible histone acetylation is balanced by histone deacetylases (HDAC) and histone acetyltransferases (HAT). These epigenetic processes remodel the chromatin
SC
structure and control gene expression without changing the gene sequence [1]. Acetylation of the lysine tail of nucleosome-related histone neutralizes its positive
M AN U
charge, which causes the relaxed chromatin to induce transcriptional expression. In contrast, deacetylation of histone leads to the formation of condensed chromatin,
TE D
which causes transcriptional repression. Deregulation of such epigenetic systems may induce inappropriate gene expression associated with the pathogenesis of certain
EP
malignancies [2, 3]. These findings suggest HADCs are involved in regulating various cellular events. HDAC inhibitors are known to exhibit anticancer activity in many
AC C
tumor types as well as in-vivo models, and consequently, inhibition of HDACs has been emerged as an attractive strategy in cancer therapeutics [4, 5]. Mammalian HDACs can be classified into four groups according to their sequence homology, sub-cellular distribution, and catalytic activity. Class I (HDAC1, -2, -3, -8), class II (HDAC4, -5, -6, -7, -9, -10), and class IV (HDAC11) enzymes are
2
ACCEPTED MANUSCRIPT zinc-dependent HDACs, whereas class III (Sirtuins 1-7) enzymes require NAD+ as a cofactor for activity. Class II enzymes are further subdivided into class IIa (HDAC4,
RI PT
-5, -7, -9) and class IIb (HDAC6, -10) [6]. Class I and IV HDACs are ubiquitously expressed in the nucleus. In contrast, class II HDACs are located in some specific
SC
tissues and shuttle between the nucleus and cytoplasm [7]. Class IIa HDACs are shown to have certain biological functions. For example, HDAC4 performs a
M AN U
prominent neurological function and plays a distinct role in synaptic plasticity and memory [8, 9]. HDAC5 reportedly regulates differentiation-specific functions.
TE D
HDAC9, meanwhile, is highly expressed in the brain and skeletal muscle [8]. Study showed that HDAC5- or HDAC9-deficient mice has an increased incidence of the
EP
cardiac hypertrophy after stress [10]. HDAC7 is highly expressed in the vascular endothelium. HDAC7 deficiency leads to the embryonic lethality in mice due to
AC C
aberrant circulatory development [11]. These experimental facts revealed that inhibiting class IIa HDACs possibly causes some undesirable side effects. In contrast, extensive studies reported that class I enzymes (HDAC1, -2, -3, -8) are involved with cell cycle progression, metastasis, and apoptosis [12]. Moreover, these enzymes are highly expressed in certain cancer cells compared to their corresponding normal cells.
3
ACCEPTED MANUSCRIPT Therefore, the anticancer effects of class I HDAC enzymes have been studied intensively.
RI PT
Clinical studies have evaluated the use of HDAC inhibitors for treating various solid and hematologic malignancies. These structurally diverse compounds include compounds
such
as
SAHA,
panobinostat,
PCI-24781,
SC
hydroxamate-based
JNJ-26481585, benzamides such as MS-275 and MGCD0103 and cyclic tetrapeptides
M AN U
such as FK228 and apicidin [13]. The hydroxamate-based compounds are considered pan-HDAC inhibitors due to their broad-spectrum inhibition of HDAC isoforms (Fig.
TE D
1). In contrast, the other two groups are classified as class I-selective HDAC inhibitors because they display specific inhibiting effects on HDAC1, -2, -3 enzymes.
EP
Among these HDAC inhibitors, SAHA and FK228 were approved for the treatment of cutaneous T-cell lymphoma (CTCL) [13, 14] and PXD101 was used to treat refractory
AC C
peripheral T-cell lymphoma (PTCL) [15]. Recently, panobinostat has been approved for use in treating multiple myeloma [16]. However, in the clinical trials, SAHA and PXD101 reportedly cause more side effects for the patients compared to MS275, which suggested targeting class I HDAC may offer wider therapeutic window [14, 17].
4
ACCEPTED MANUSCRIPT The chemical features of all HDAC inhibitors can be divided into three groups: a zinc-binding motif (such as a hydroxamic acid), a hydrophobic cavity-binding linker,
RI PT
and a surface recognition cap also with hydrophobic properties. Each group interacts with a discrete region of the active site of HDAC (Fig. 1). Studies also indicate that
SC
modification of the recognition cap moiety may enable recognition of specific HDACs through binding to the rim surface of the enzyme active site, which
AC C
EP
TE D
M AN U
potentially generates selective HDAC inhibitors [18].
5
ACCEPTED MANUSCRIPT Figure 1. Examples of pan and class I-selective HDAC inhibitors in clinical trials. Trimethoxyindole exists in various anticancer agents (Fig. 1S). These compounds
RI PT
include synthesized combretastatin A-4 derivatives 1-3 [19-21] and a naturally occurring antibiotic, duocarmycin SA [22-24]. Our previous efforts sought to enhance
SC
the recognition caps that are fused with reported HDAC inhibitor scaffold such as N-hydroxycinnamide and aliphatic hydroxamate [25, 26]. Accordingly, we previously
M AN U
developed trimethoxyindole derivative 6d fused with the aliphatic-hydroxamate core of SAHA (Scheme 1). Comparison of the HDAC-inhibiting activities of compound 6d
TE D
and SAHA indicated that compound 6d displayed higher HDAC1 activity [27]. In addition to its potent enzyme inhibition, compound 6d, in human hormone-refractory
EP
prostate cancer cells, also significantly affected cell cycle regulators as well as apoptotic signaling, which are induced by its anti-HDAC activity. These experimental
AC C
results revealed such modifications can increase HDAC specificity and produces improved HDAC-related cellular effects. Moreover, the trimethoxyindole of compound 6d structurally resembles the indole cap used for HDAC inhibitors LBH-589, PCI-24781, and JNJ-26481585 currently tested in clinical trials (Fig. 1). Despite its strong anti-HDAC effect, the structure activity relationship of compound
6
ACCEPTED MANUSCRIPT 6d is still not disclosed. These results further motivated us to extensively investigate the effect of the chemical motif of 6d-derived compounds on HDAC-inhibiting
RI PT
activity such as the orientation of indole moiety added to aliphatic hydroxamate, the linker carbon-chain length between the cap and zinc-binding group, and the
SC
substituent of indole ring.
Using compound 6d as a lead, this study incorporated varied indole moieties into
M AN U
the core structure of SAHA via various carbon linker-chain lengths at different positions (Scheme 1). The HDAC inhibitory activities of the resulting compounds
TE D
were evaluated, which leads to identify their structure activity relationship. Several meta-substituted compounds had higher anti-HDAC activity compared to SAHA. Test
EP
of cytotoxicity revealed their cytotoxicity against human prostate cancer PC-3 cells was comparable to that of SAHA. Notably, two of these compounds, designated
AC C
compounds 6j and 6k, showed much higher potency than SAHA in class I HDAC-1, -2, -3 and -8. Further assays indicated that both compounds had class IIb HDAC6 inhibitory activities compatible to SAHA, but with only slight effects on most class IIa isoforms. Analysis of the binding pocket revealed structural differences between the HDAC isozymes. Further experiments were performed to profile acetylation of
7
ACCEPTED MANUSCRIPT histone and α-tubulin, cell cycle progression, and gene expression and revealed the cellular effects that are relevant to the HDAC inhibition of the resulting compounds.
H3CO
H3CO
OCH3 N
O 2O
N H
O 6
N H
OCH3
H3CO
OH
N
H N nO
H N
6
O
OH
O
RI PT
H3CO
n=2-4
6d
N
M AN U
R
SC
ortho-, meta-, para-(6a-i)
2O
O N H
O
N H n=4, 6-8 n
6j-r
AC C
EP
TE D
Scheme 1. Design of indole-containg aliphatic hydroxamates derived from compound 6d
8
OH
ACCEPTED MANUSCRIPT 2. Chemistry The hydroxamates 6a-6o were synthesized as described in Scheme 2.
RI PT
Monomethyl suburate was reacted with oxalyl chloride to produce the activated acid chloride in situ. Treatment with ortho-, meta- and para-aminophenol 1a-1c then gave
SC
corresponding methyl esters 2a-2c. Reaction of compounds 2a-2c with the appropriate alkyl bromides, such as 2-chloroethyl, 3-chloropropyl and 4-chlorobutyl,
M AN U
provided corresponding 3a-3i. Trimethoxyindole was synthesized according to the reaction approach as previously reported [27]. Compounds 3a-3i coupled with various
TE D
indoles yielded compounds 4a-4o, respectively. Saponification of compounds 4a-4o in the presence of LiOH gave corresponding acids 5a-5o in quantitative yields. To
EP
activate compounds 5a-5o, the corresponding mixed anhydrides were generated by
AC C
using ethyl chloroformate prior to reaction with NH2OH to give hydroxamates 6a-6o.
9
ACCEPTED MANUSCRIPT Scheme 2
HO
6
O
H N
a
O O
NH2
O
HO
H N
b
O
6
O
Br
Cl n
Cl
O
nO
H N
O
6
O
H N
d
O
R
O N
O
O
R
N
O
N
M AN U
O
N
Br N
O
nO
H N
6
OH
O
6a-o
O
O
H N
SC
:
e
5a-o
O R
6
O
nO
4a-o
OH
RI PT
n=2-4
1a: o-aminophenol 1b: m-aminophenol 1c: p-aminophenol
nO
O
3a-i
2a-c HO
R
c
O
6
N
N
O
TE D
Reagents and conditions: (a) 1) (COCl)2, CH2Cl2, RT; 2) DMF, 60 oC, N2, 42-46%; (b) K2CO3, MeCN, ∆, N2, 79-99%; (c) substituted indole or indole, K2CO3, DMF, RT, N2, 25-95%; (d) LiOH, MeOH, 95-99%; (e) 1) ClCO2Et, Et3N, THF, RT; 2) NH2OH-HCl, KOH, MeOH, 56-99%
The meta-substituted hydroxamates 6p-6r were synthesized as described in
EP
Scheme 3. Compounds 7a-7c reacted with oxalyl chloride prior to coupling with compound 1b gave 8a-c, respectively. Using the same method to compounds 6a-6o,
AC C
compounds 6p-6r were synthesized starting from compounds 8a-8c. Details on the synthesis, isolation and characterization of compounds 2-5 and 8-9 can be found in the supplementary material. The estimated purity of compounds 6a-6r is at least 97% as determined by HPLC analysis (supplementary material).
10
ACCEPTED MANUSCRIPT Scheme 3
O
H N
a
O
n
O
O
7a-c n=4, 7, 8
O
n O
N
Br
H N O
n O
N
n O
O
Cl
O
c
9a-c
d
O
O
N
H N
O
O
n O
OH
e
4p-r
4p-r
O
H N
O
Cl
8a-c
OH
O
b
O
RI PT
HO
H N
O
H N
OH
SC
O
n O
6p-r
3. Results and Discussion
M AN U
Reagents and conditions: (a) 1) (COCl)2, CH2Cl2, RT; 2) 1b, DMF, 60 oC, N2, 65-87%; (b) K2CO3, MeCN, ∆, N2, 8897%; (c) 5-Methoxyindole, K2CO3, DMF, RT, N2, 80-97%; (d) LiOH, MeOH, 97-98%; (e) 1) ClCO2Et, Et3N, THF, RT; 2) NH2OH-HCl, KOH, MeOH, 79-89%
3.1 HeLa nuclear HDAC inhibitory activity
TE D
The enzyme inhibitory activities of all synthesized compounds 6a-6m against HeLa nuclear HDACs were tested using SAHA as a reference (Table 1). The activities
EP
of compounds 6a-6c with ortho-substituted trimethoxyindole moiety were
AC C
approximately 2-fold lower than that of SAHA, which indicates that the ortho substitution weakens the enzyme binding affinity. The activities of para-substituted compounds 6g and 6i were comparable to that of SAHA, suggesting that para-substituted indole slightly enhances enzyme inhibition. In contrast, the activities of meta-substituted compounds 6d-6f were approximately two- to five-fold higher than that of SAHA. We speculate that the positive contribution of meta-substitution 11
ACCEPTED MANUSCRIPT inhibitory activity was due to increased contact between the indole motif and the rim of HDAC enzyme active site. Comparisons of the meta-series 6d-6f indicated that
RI PT
compound 6d had the strongest activity, which suggested that a two-carbon linker chain between the benzene and the recognition cap produced optimal anti-HDAC
SC
effect. Next, compounds 6j-6m were synthesized with different indoles at the meta-position using a two carbon chain length. These four compounds also showed
M AN U
either an equivalent or higher potency compared to compound 6d. Notably, the enzyme-inhibiting activity of compound 6k was within a range of single digit
TE D
nanomolar. In general, we observed that compounds that contained at least a two-carbon linker and indoles at the meta-position had better inhibiting activity. Table 1
EP
IC50 value for the inhibition against HeLa nuclear HDACa (nM) and cytotoxicity
AC C
against cancer cells (µM) by compounds 6a-m H N
H N 6
N
R
nO
O
OH
O
Substitution
Chain
Substitution
position
length (n)
(R)
6a
ortho
2
5,6,7-TriOMe
109.9±1.2
5.6
5.9
6b
ortho
3
5,6,7-TriOMe
111.0±5.2
3.9
5.0
6c
ortho
4
5,6,7-TriOMe
111.4±9.8
2.4
4.8
6d
meta
2
5,6,7-TriOMe
11.5±0.2
1.3
2.4
6e
meta
3
5,6,7-TriOMe
22.5±0.8
2.7
2.7
Compound
12
HDAC
PC-3
A549
ACCEPTED MANUSCRIPT meta
4
5,6,7-TriOMe
17.8±1.9
2.1
2.4
6g
para
2
5,6,7-TriOMe
57.2±3.3
4.4
4.9
6h
para
3
5,6,7-TriOMe
138.6±1.1
6.9
5.9
6i
para
4
5,6,7-TriOMe
46.4±0.6
7.7
9.7
6j
meta
2
5,6-DiOMe
11.2±1.3
1.2
6.4
6k
meta
2
5-OMe
8.6±0.7
1.3
1.7
6l
meta
2
6-OMe
14.9±0.1
1.8
4.2
6m
meta
2
7-OMe
13.5±0.4
1.4
1.9
51.6±3.6
1.5
1.7
SAHA Data was obtained from three independent experiments.
SC
a
RI PT
6f
M AN U
We further synthesized meta-substituted compounds 6n-6o to investigate the effect of indole’s substituent on HDAC inhibition. Table 2 shows that compared to compound 6n with unsubstituted indole, compound 6o with bromo-substituted indole
TE D
exhibited lower enzyme inhibition, which suggested that an electron-withdrawing group on indole negatively contributed to the activity. However, compound 6k
EP
showed greater activity, which suggested that an electron-donating group
AC C
strengthened the activity. Finally, this study synthesized compounds 6p-6r with the linker-chain length of 4, 7, and 8 carbons, respectively, between the amide and hydroxamate group. Comparisons of the enzyme-inhibiting activities between compounds 6k and 6p-6r show that compound 6k had the most potent activity, which suggested that for this series of compounds, the optimal length of linker-chain is six carbons. 13
ACCEPTED MANUSCRIPT Table 2. IC50 value for the inhibition against HeLa nuclear HDACa (nM) by compounds 6n-6r H N
O
N
O
OH
Chain length (n)
Substitution (R)
6n
6
H
6o
6
5-Br
6p
4
5-OMe
185.8±10.3
6q
7
5-OMe
40.0±3.2
6r
8
5-OMe
298.0±14.2
6k
6
5-OMe
8.6±0.7
M AN U
SC
Compound
SAHA a
n O
H N
RI PT
R
HDAC
23.1±0.3
41.3±0.9
51.6±3.6
Data was obtained from three independent experiments.
TE D
3.2 Cytotoxicities of potential inhibitors
The cell growth inhibiting effects of compounds 6a-6m were compared in human
EP
prostate cancer PC-3 cells and lung cancer A549 cells. Table 1 showed that, in both cancer cell lines, all compounds except 6c and 6i had cytotoxic effects consistent with
AC C
the HDAC enzyme inhibition. In contrast to its weak anti-HDAC activity, the cytotoxicity of compound 6c was moderate. The experimental result suggested that, in addition to HDAC, compound 6c likely affects other cell death-related cellular events in the signaling pathway. Although the enzyme inhibitory activity of compound 6i was comparable to that of SAHA, compound 6i had much lower cytotoxicity than
14
ACCEPTED MANUSCRIPT SAHA. Compound 6j preferentially inhibited PC-3 cells (IC50=1.2 µM) over A549 cells (IC50= 6.4 µM) when compared to SAHA. The HDAC enzyme inhibitory
RI PT
activities of compounds 6d and 6k-6m were much higher than that of SAHA; however, their cytotoxicities were only comparable or equivalent to that of SAHA.
SC
This may be due to their poor cell membrane permeability. 3.3 HDAC isoforms inhibition
M AN U
Next, we selected four compounds (6d, 6f, 6j, and 6k) that showed high anti-HeLa nuclear HDAC activity and determined their enzyme inhibiting effects
TE D
against a panel of HDAC isoforms. These isozymes include class I HDACs 1, 2, 3, 8, class IIa HDACs 4, 5, 7, 9, and Class IIb HDAC6. Table 3 shows that the HDAC1
EP
inhibiting effects of most compounds were superior to that of SAHA. For example, the activities of compounds 6j and 6k were 14- and 16-fold higher than that of
AC C
SAHA[28]. Similarly, they also had stronger anti-HDAC8 effect than did SAHA with four- to eight-fold greater activities. The activities of compounds 6d, 6j and 6k against HDAC2 were approximately two-, twelve-and seven-fold higher than that of SAHA, respectively; however, the activity of compound 6f was five-fold lower. Compared to SAHA, compounds 6d, 6j and 6k had higher or comparable potency
15
ACCEPTED MANUSCRIPT against HDAC3, whereas compound 6f had lower potency. The compounds showed weak activity against class IIa HDACs 4, 5, 7, and 9 (Table 3). Moreover, increasing
RI PT
concentrations as high as 10 µM reveal low inhibitory effects. In contrast, the HDAC6 inhibitory activities of compounds 6d, 6f, 6j and 6k were comparable to that of
SC
SAHA (Table 3). These experimental results indicated the methoxy group when added
and 8. Table 3
M AN U
to the indole cap can affect the enzyme-inhibiting activity in class I HDACs 1, 2, 3
IC50a (nM) value for the inhibition against class I and class II HDACs by compounds 6d, 6f, 6j and 6k Compound
TE D
Class I HDAC1 HDAC2 HDAC3
HDAC8
Class II
HDAC4 HDAC5 HDAC7 HDAC9 HDAC6
11.1±1.7 111.2±0.2 72.5±0.5 420.9±3.6 >10000
>10000
>10000 >10000 9.6±0.2
6f
29.3±0.9 999.6±5.9 108.2±0.4 752.0±5.2 >10000
>10000
>10000 >10000 8.5±0.7
6j
3.9±0.3 16.6±1.4 29.7±0.7 398.3±2.3 >10000
>10000
>10000 >10000 7.1±1.0
6k
4.5±0.1 33.1±0.5 35.3±0.2 560.9±1.2 >10000
>10000
>10000 >10000 5.9±2.0
>10000
>10000 >10000 9.1±0.3
AC C
SAHA a
EP
6d
63.5±1.9 214.0±0.1 72.8±1.8 3400.1±6.2 >10000
Each value is obtained by three independent experiments.
A phylogenetic tree based on the sequence homology between HDAC isozymes separates the HDACs by class (Fig. 2S). When the IC50 values of the identified compounds were placed onto the trees, a bias for class I and HDAC6 isozymes was 16
ACCEPTED MANUSCRIPT observed. This suggested a possible discrepancy between the isozyme structures that determines inhibitor potency. Therefore, we identified inhibitors that are specific for
RI PT
class I HDACs.
3.4 Molecular docking analysis
SC
We performed an interaction analysis to determine key interacting residues between the strongest (6j) and the weakest (6f) inhibitors for HDAC1 (Table 3). The
M AN U
inhibitors were docked into HDAC1 (PDB ID: 4KBX) using the software SYBYL [29] to obtain the binding conformations. The interaction analysis between the enzyme
TE D
residues and inhibitors were determined using the program iGEMDOCK [30]. The modeling of compounds 6j and 6f showed the locations of the indole cap along the
EP
surface (Fig. 2A-B). Superimposing the two compounds in HDAC1 showed different spatial positions in the surface (Fig. 2C). Within the binding site, the two compounds
AC C
created similar hydrogen bonds with residues H140, H141, D176, H178, D264, and Y303 (Fig. 2D-E). This is due to their hydroxamate moiety. In contrast, compounds 6j and 6f formed different van der Waals interactions with surface residues (Fig. 2C, 2E). Compound 6f contains a four-carbon linker chain. The longer linker allowed the indole of compound 6f to display a higher degree of rotation to form interactions with
17
ACCEPTED MANUSCRIPT residues H28 and P29. Compound 6j with a two-carbon linker positioned the indole to form van der Waals interactions with residues H28 and P29 (Fig. 2C). These results
RI PT
suggested that the linker length and the interactions between the indole cap and
EP
TE D
M AN U
SC
surface residues H28 and P29 are important for the potency of the inhibitors.
Figure 2. Molecular docking analysis of inhibitors 6f and 6j in HDAC1. Compound
AC C
6f and 6j docked into HDAC 1 (PDB ID: 4BKX) crystal structure. (A-B) Surface view of compound 6j (yellow) and compound 6f (purple) docked in HDAC 1. (C-D) Compounds 6j (yellow) and compound 6f (purple) docking pose stick view of the HDAC1 (grey) active site reveal interactions with residues. (E) Compounds 6f and 6j were superimposed in HDAC 1 catalytic center. Residues contributed hydrogen bond
18
ACCEPTED MANUSCRIPT are colored in green, residues contributed van der Waals interaction are colored in brown. H178 contain hydrogen donor and pi-donor is colored in yellow. (F) Heat map
RI PT
of corresponding residue interactions and compounds 6j and 6f. Hydrogen bonds are represented in shades of green and van der Waals in shades of grey.
SC
3.5 Structural differences between HDAC isozymes
To determine discrepancies between residue interactions and HDAC class
M AN U
potency, we compared and analyzed the HDAC isozyme structures. As seen in Table 3, the identified inhibitors show a low IC50 value for class I isozymes, but a high value
TE D
for class IIa isozymes. The identified compounds had a IC50 value comparable to SAHA for HDAC6, a class IIb isozyme (Table 2). HDAC sequences was obtained
EP
from Uniprot database and a multiple sequence alignment was performed in Jalview [31]. We observed that the histidine residue is relatively conserved across the HDAC
AC C
family; however, the tyrosine and histidine amino acid differs between class I and class IIa HDAC, respectively (Fig. 3A). While residues were relatively conserved, the surface view of the binding site revealed different cavity sizes between the HDAC classes. The crystal structures for HDAC5, 9 and 10 are currently unavailable; as a result, structural analysis and modeling of these structures was not performed. Class I
19
ACCEPTED MANUSCRIPT and HDAC6 contain a tyrosine residue, whereas class IIa HDACs contain a histidine residue in the same location [8, 32]. The aromatic ring of the tyrosine residue extends
RI PT
into the binding site and creates a narrow pocket for the inhibitor (Fig. 3B). Furthermore, our interaction analysis revealed a hydrogen bond between the tyrosine
SC
residue and inhibitor 6j (Fig. 2F). The amount of interactions for inhibitor 6j is reduced in class IIa HDACs due to the histidine residue substitution, which produces
M AN U
a larger binding site (Fig. 3C). The difference in residues between class IIa and class I and HDAC6 is further amplified when the HDAC isozymes are aligned with inhibitor
TE D
6j (Fig. 3D). The sequence alignment of HDAC8 showed that it contains a leucine and alanine instead of the conserved histidine and proline residues for class I HDAC
EP
(Fig. 3A). This may explain why the inhibitors and SAHA produced a higher IC50
AC C
value compared to the other class I HDACs (Table 3).
20
Figure
3.
Interaction
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
analysis
of
inhibitors
6f
and
6j.
(A)
Multiple Sequence Alignment (MSA) of HDAC family. Residues were aligned to
TE D
HDAC1. Amino acids labeled using Crustal X color scheme. (B) The docking pose of inhibitor 6j in HDAC1 (yellow) was aligned with class I and class IIb HDAC6 and (C)
EP
class IIa HDAC4 and HDAC7. (D) All HDAC isozyme structures used previously are
AC C
aligned. Red circle highlights contrasting residues in alignment. HDACs color is labeled as shown.
We next looked at specific indole interactions with surface residues. The HDAC1
and inhibitor 6j complex was aligned to class I isozymes and HDAC6 (Fig. 4A). The inhibitor cap is situated near the HDAC1 residue H28. In contrast, when the 6j-HDAC1 complex is superimposed to class IIa HDACs, the histidine residue is 21
ACCEPTED MANUSCRIPT located on a different plane (Fig. 4B). Alignment of available HDAC structures reveals the histidine residue of class IIa situated away from class I and HDAC6
RI PT
isozymes (Fig. 4C). When the surface is compared, the histidine of HDAC1 forms interactions with the inhibitor cap, whereas HDAC4 contains a non-interacting pocket
SC
due to its histidine residue location (Fig. 4D-E). This observation accounts for the
AC C
EP
TE D
M AN U
increased potency of the compound 6j for class I isozymes and HDAC6.
Figure 4. Surface residues interacts with the inhibitor cap. The surface residues of class I and HDAC6 (A) and class IIa (B) isozymes, as described in Figure 3, are compared. (C) Alignment of all HDAC structures used with Inhibitor 6j docked in HDAC1, were transposed. Red circle denotes contrasting histidine locations between 22
ACCEPTED MANUSCRIPT isozymes. HDACs labeled as shown. The surface model of surface residues of HDAC 1 (D) and HDAC4 (E) reveal non-interacting pocket (red circle).
RI PT
3.6 Hyperacetylation of histone and α-tubulin Compounds 6j and 6k at various concentrations (0.1, 0.3, 1, 3 and 10 µM) were
SC
compared in terms of their enhancement of acetylation of histone and α-tubulin, which are two biomarkers for intracellular HDAC inhibition. Figure 5 shows that, in
M AN U
PC-3 cells, both compounds promoted histone acetylation in a dose-dependent manner. Notably, compounds 6j and 6k at concentrations as low as 1 µM induced higher levels of histone acetylation than did SAHA. In addition to histone acetylation, similar
TE D
concentrations of compounds 6j and 6k enhanced α-tubulin acetylation. The potency
EP
of their enhancing effects was comparable to that of SAHA. These experimental data had strong correlations with the HDAC isoform inhibitory activities of compounds 6j
AC C
and 6k. However, in A-549 cells, compound 6k slightly induced histone acetylation when concentration was increased up to 10 µM (Fig. 3S), which confirmed its preferential cytotoxicity towards PC-3 cells.
23
RI PT
ACCEPTED MANUSCRIPT
Figure 5. Effect of compounds 6j and 6k on induction of histone acetylation and
SC
α-tubulin acetylation in cultured human prostate cancer PC-3 cells. Cells were incubated with the HDAC inhibitors at the indicated concentrations for 16 h. The
M AN U
whole cell lysates were analyzed for histone and α-tubulin acetylation by SDS-PAGE and Western blot for either acetylated histone or α-tubulin.
TE D
3.7 Cell cycle progression
HDACs have been shown to regulate the cell cycle [12]. The effects of
EP
compounds 6j and 6k on cell cycle progression were then investigated by flow cytometry. Different concentrations (1, 3, and 10 µM) of the two compounds were
AC C
tested in PC-3 cells (Fig. 6A). After 24 h incubation, compounds 6j and 6k increased the G2/M population in a dose-dependent manner (Fig. 6B). (A)
24
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
(B)
Figure 6. Effect of compounds 6j and 6k on cell cycle progression in cultured human prostate PC-3 cells. (A) Cells were treated without (ctrl) or with the HDAC inhibitors at the indicated concentrations for 24 h and were analyzed by flow cytometry for cell cycle distribution. (B) Data were presented by mean value of three independent experiments. 25
ACCEPTED MANUSCRIPT
3.8 Gene expression
RI PT
The HDAC inhibitors reportedly affect a spectrum of genes in transformed cells [33]. To compare the effects of compounds 6j, 6k, and SAHA on gene expression
SC
associated with cell proliferation, these three compounds were evaluated for the levels of representative genes in human prostate cancer PC-3 cells and lung cancer A549
M AN U
cells. These genes included p21 (inducing cell cycle arrest) [34], CCND1 (inducing cell cycle progression) [35], RARβ (mediating differentiation and growth arrest) [36],
TE D
TNFα (inducing cell apoptosis) [38], Bcl-2 (inhibiting cell apoptosis) [40], CDH15 [42] (inhibiting migration) as well as TIMP3 [44-46] and CDH1 [47] (supressing
EP
tumor growth). While we observed high RARβ levels induced by compound 6k in PC3 cells, the levels of most genes in PC-3 and A549 cell lines by compounds 6j and
AC C
6k were comparable to those by SAHA (Fig. 4SA). In A549 cells, the TNFα levels induced by compounds 6j and 6k were higher than the TNFα levels induced by SAHA (Fig. 4SB). The levels of compound 6k were the highest. Additionally, compounds 6j and 6k were comparable to SAHA in terms of enhancing effects on TIMP3 expression and CDH1 expression in A549 cells (Fig. 4SC), In contrast, compound 6j, 6k and
26
ACCEPTED MANUSCRIPT SAHA in PC-3 cells showed no significant mediating effects on TNFα, TIMP3 and CDH1 expression (data not shown).
RI PT
4. Conclusion This study synthesized a novel series of aliphatic hydroxamates by incorporating
SC
varied indoles as a recognition cap into SAHA core. The structure activity relationship of the resulting compounds in HDAC-inhibiting activity was extensively disclosed.
M AN U
These findings showed the favored orientation of indole cap, the optimal carbon linker-chain length between the cap and zinc-binding group and the effect of the
TE D
substituent of indole ring in inhibiting HDAC. In class I HDAC enzymes, several of these compounds had much higher enzyme inhibition compared to SAHA. Thus, they
EP
are identified to be potent class I HDAC inhibitors. Notably, two of these compounds, namely 6j and 6k, significantly induced histone and tubulin acetylation.
AC C
Additionally, both compounds arrested cell cycle progression in G2/M phase in a dose-dependent manner. Further mechanism studies associated with the identified inhibitors are being investigated.
27
ACCEPTED MANUSCRIPT 5. Experimental 5.1 General procedures
RI PT
The IR spectra (KBr disk) were recorded using a Jasco Fourier Transform infrared spectrometer (Jasco FT/IR-410). The 1H-NMR spectrum was obtained on a Bruker
SC
AV400 or AV500 spectrometer using standard pulse programs. Melting point was recorded on Fisher-Johns apparatus (uncorrected). The MS data were measured on
M AN U
JEOL JMX-HX110 mass spectrometer (HREIMS and HRFABMS), JMS-SX102A mass spectrometer (EIMS and FABMS), and Finnigan Mat TSQ-7000 mass
TE D
spectrometer (ESIMS). The TLC analyses were performed on silica gel plates (KG60-F254, Merck). The microplate spectrophotometer Victor 2x (Perkin Elmer,
EP
Fremont, CA, USA) was used for fluorometric analysis. Unless otherwise mentioned, all chemicals and materials were used as received from commercial suppliers without
AC C
further purification. Dichloromethane was distilled from calcium hydride under nitrogen. Tetrahydrofuran was distilled from sodium and benzophenone under N2. 5.2 N-Hydroxy-7-(2-(2-(5,6,7-trimethoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptana mide (6a)
28
ACCEPTED MANUSCRIPT After adding KOH (224 mg, 3.98 mmol) to a solution of NH2OH-HCl (277 mg, 3.98 mmol) in MeOH (5 mL), the resulting solution was stirred in an ice bath for 1 h.
RI PT
Free NH2OH in MeOH solution was obtained by filtering out the white salt. A solution of 5a (120 mg, 0.24 mmol) in freshly distilled THF (20 mL) was treated with
SC
ethyl chloroformat (0.047 mL, 0.48 mmol) and triethylamine (0.066 mL, 0.48 mmol), and the resulting solution was stirred at RT for 1 h. The prepared free NH2OH
M AN U
solution was then added to the reaction. After 3 h stirring, the reaction mixture was concentrated in vacuo, and the residue was diluted with distd H2O (50 mL), acidified
TE D
with 1N HCl to pH 2~3 and extracted with EtOAc (50 mL x 3). The organic layer was dried over Na2SO4, and the solvent was removed in vacuo. The residue was purified
EP
by silica gel chromatography (CH2Cl2: MeOH =47:3) to give 6a (124 mg, 56%) as a solid. Mp 95-100 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.32 (s, 1H), 8.64 (s, 1H),
AC C
8.48 (s, 1H), 7.93 (d, J = 7.4 Hz, 1H), 7.40 (d, J = 2.7 Hz, 1H), 6.97 (d, J = 3.8 Hz, 2H), 6.86 (t, J = 5.4 Hz, 2H), 6.29 (d, J = 3.0 Hz, 1H,), 4.67 (t, J = 4.7 Hz, 2H), 4.27 (t, J = 4.7 Hz, 2H), 3.94 (s, 3H), 3.76 (s, 3H), 3.75 (s, 3H), 2.32 (t, J = 7.2 Hz, 2H), 1.95 (t, J = 7.2 Hz, 2H), 1.54 (p, J = 7.0 Hz, 2H), 1.51 (p, J = 7.0 Hz, 2H), 1.30 (m, 4H).
13
C NMR (125 MHz, DMSO-d6): δ 171.2, 169.6, 154.4, 148.9, 140.2, 138.6,
29
ACCEPTED MANUSCRIPT 133.3, 130.7, 125.5, 123.2, 121.2, 115.0, 101.5, 98.4, 68.3, 62.0, 61.3, 56.5, 47.5, 36.7, 32.8, 28.9, 25.5. HR-ESI-MS m/z: (M+Na)+ caclcd for C27H35N3O7Na 536.2373,
RI PT
found 536.2377. 5.3
SC
N-Hydroxy-7-(2-(3-(5,6,7-trimethoxy-1H-indol-1-yl)propoxy)phenylcarbamoyl)hepta namide (6b)
M AN U
Following the procedure as described for 6a, reaction of 5b (311 mg, 0.61 mmol) in THF (20 mL) with ethyl chloroformate (0.12 mL, 1.22 mmol) and triethylamine
TE D
(0.17 mL, 1.22 mmol) gave 6b (301 mg, 94%) as a solid. Mp 55-60 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.29 (s, 1H), 8.79 (s, 1H), 8.62 (s, 1H), 7.80 (d, J = 7.7 Hz, 1H),
EP
7.16 (d, J = 3.0 Hz, 1H), 7.01 (t, J = 7.6 Hz, 1H), 6.91 (d, J = 8.0 Hz, 1H), 6.87 (t, J =7.8 Hz, 1H), 6.83 (s, 1H), 6.26 (d, J = 2.9 Hz, 1H), 4.43 (t, J = 6.7 Hz, 2H), 3.92 (s,
AC C
3H), 3.89 (t, J = 6.0 Hz, 2H), 3.76 (s, 3H), 3.72 (s, 3H), 2.36 (t, J = 7.2 Hz, 2H), 2.19 (p, J = 6.2 Hz, 2H), 1.91 (t, J = 7.3 Hz, 2H), 1.57 (p, J = 7.0 Hz, 2H), 1.47 (p, J = 7.4 Hz, 2H), 1.26 (4H, m). 13C NMR (125 MHz, DMSO-d6): δ 171.5, 170.0, 154.8, 148.8, 140.2, 138.5, 133.0, 130.3, 125.5, 123.2, 121.2, 115.0, 101.4, 98.3, 65.3, 62.1, 61.3, 56.5, 46.2, 45.2, 36.7, 32.7, 31.4, 28.9, 25.6. HR-ESI-MS m/z: (M+Na)+ caclcd for
30
ACCEPTED MANUSCRIPT C28H37N3O7Na 550.2529, found 550.2525. 5.4
RI PT
N-Hydroxy-7-(2-(4-(5,6,7-trimethoxy-1H-indol-1-yl)butoxy)phenylcarbamoyl)heptana mide (6c)
SC
Following the procedure as described for 6a, reaction of 5c (395 mg, 0.75 mmol) in THF (20 mL) with ethyl chloroformate (0.15 mL, 1.50 mmol) and triethylamine
M AN U
(0.21 mL, 1.50 mmol) gave 6c (320 mg, 81%) as a solid. Mp 65-68 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.30 (s, 1H), 8.77 (s, 1H), 8.62 (s, 1H), 7.81 (d, J = 7.6 Hz, 1H),
TE D
7.19 (d, J = 3.0 Hz, 1H), 7.02 (t, J = 7.6 Hz, 1H), 6.96 (d, J = 7.7 Hz, 1H), 6.86 (t, J = 7.6 Hz, 1H), 6.83 (s, 1H), 6.27 (d, J = 3.0 Hz, 1H), 4.29 (t, J = 7.0 Hz, 2H), 3.99 (t, J
EP
= 6.1 Hz, 2H), 3.89 (s, 3H), 3.76 (s, 3H), 3.73 (s, 3H), 2.29 (t, J =7.1 Hz, 2H), 1.91 (t, J = 7.4 Hz, 2H), 1.88 (t, J = 7.7 Hz, 2H), 1.71 (p, J = 6.4 Hz, 2H), 1.50 (p, J = 6.6 Hz,
AC C
2H), 1.45 (p, J = 7.0 Hz, 2H), 1.23 (m, 4H). 13C NMR (125 MHz, DMSO-d6): δ 171.3, 169.7, 154.8, 148.8, 140.2, 138.4, 132.9, 130.2, 125.5, 123.2, 121.1, 114.9, 101.2, 98.3, 67.7, 61.9, 61.3, 56.5, 48.0, 36.7, 32.8, 28.9, 28.5, 26.6, 25.6. HR-ESI-MS m/z: (M+Na)+ caclcd for C29H39N3O7Na 564.2686, found 564.2682. 5.5
31
ACCEPTED MANUSCRIPT N-Hydroxy-7-(3-(2-(5,6,7-trimethoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptana mide (6d)
RI PT
Following the procedure as described for 6a, reaction of 5d (329 mg, 0.66 mmol) in THF (20 mL) with ethyl chloroformate (0.12 mL, 1.32 mmol) and triethylamine
SC
(0.18 mL, 1.32 mmol) gave 6d (277 mg, 82%) as a solid. Mp 105-108 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.30 (s, 1H), 9.77 (s, 1H), 8.62 (t, J = 0.9 Hz, 1H), 7.26 (s,
M AN U
1H), 7.23 (d, J = 3.1 Hz, 1H), 7.13 (t, J = 8.2 Hz, 1H), 7.07 (d, J = 8.2 Hz, 1H), 6.84 (s, 1H), 6.55 (dd, J = 2.1, 8.1 Hz, 1H), 4.61 (t, J = 5.4 Hz, 2H), 4.20 (t, J = 5.4 Hz, 2H), 3.94 (s, 3H), 3.77 (s, 3H), 3.75(s, 3H), 2.24 (t, J = 7.4 Hz, 2H), 1.92 (t, J = 7.4
TE D
Hz, 2H), 1.52 (p, J = 6.8 Hz, 2H), 1.47 (p, J = 7.0 Hz, 2H), 1.25 (m, 4H). 13C NMR
EP
(125 MHz, DMSO-d6): δ 171.8, 169.6, 158.8, 148.9, 141.0, 140.2, 138.6, 130.7, 130.0, 125.5, 123.2, 112.1, 109.2, 106.0, 101.5, 98.4, 68.0, 62.0, 61.3, 56.5, 47.5, 36.9, 32.8,
AC C
28.9, 25.5. HR-ESI-MS m/z: (M+Na)+ caclcd for C27H35N3O7Na 536.2373, found 536.2369. 5.6 N-Hydroxy-7-(3-(3-(5,6,7-trimethoxy-1H-indol-1-yl)propoxy)phenylcarbamoyl)hepta namide (6e)
32
ACCEPTED MANUSCRIPT Following the procedure as described for 6a, reaction of 5e (711 mg, 1.39 mmol) in THF (20 mL) with ethyl chloroformate (0.27 mL, 2.78 mmol) and triethylamine
RI PT
(0.39 mL, 2.78 mmol) gave 6e (566 mg, 77%) as a solid. Mp 108-110 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.31 (s, 1H), 9.80 (s, 1H), 8.63 (s, 1H), 7.15 (t, J = 8.2 Hz,
SC
2H), 7.13 (s, 1H), 7.07 (d, J = 8.0 Hz, 1H), 6.83 (s, 1H), 6.56 (dd, J = 3.9, 8.0 Hz, 1H), 6.26 (d, J = 3.0 Hz1H,), 4.39 (t, J = 6.9 Hz, 2H), 3.93 (s, 3H), 3.85 (t, J = 6.0 Hz, 2H),
M AN U
3.76 (s, 3H), 3.74 (s, 3H), 2.26 (t, J = 7.4 Hz, 2H), 2.15 (p, J = 6.2 Hz, 2H), 1.93 (t, J = 7.3 Hz, 2H), 1.53 (p, J = 6.8 Hz, 2H), 1.48 (p, J = 7.1 Hz, 2H), 1.26 (m, 4H). 13C
TE D
NMR (125 MHz, DMSO-d6): δ 171.8, 169.6, 159.2, 148.8, 141.0, 140.2, 138.5, 130.2, 129.9, 125.5, 123.2, 111.9, 109.3, 106.0, 101.4, 98.4, 65.1, 62.0, 61.2, 56.5, 45.3, 36.9,
EP
32.8, 31.4, 28.9, 25.5. HR-ESI-MS m/z: (M+Na)+ caclcd for C28H37N3O7Na 550.2529, found 536.2369.
AC C
5.7
N-Hydroxy-7-(3-(4-(5,6,7-trimethoxy-1H-indol-1-yl)butoxy)phenylcarbamoyl)heptana mide (6f) Following the procedure as described for 6a, reaction of 5f (587 mg, 1.11 mmol) in THF (20 mL) with ethyl chloroformate (0.22 mL, 2.22 mmol) and triethylamine
33
ACCEPTED MANUSCRIPT (0.31 mL, 2.22 mmol) gave 6f (476 mg, 79%) as a solid. Mp 106-110 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.32 (s, 1H), 9.79 (s, 1H), 8.64 (s, 1H), 7.27 (s, 1H), 7.19 (t,
RI PT
J = 3.0 Hz, 1H), 7.13 (t, J = 8.1 Hz, 1H), 7.05 (d, J = 8.1 Hz, 1H), 6.82 (s, 1H), 6.54 (dd, J = 2.1, 8.0 Hz, 1H), 6.26 (d, J = 3.0 Hz, 1H), 4.28 (t, J = 7.0 Hz, 2H), 3.90 (s,
SC
3H), 3.90 (t, J = 7.4 Hz, 2H), 3.76 (s, 3H), 3.74 (s, 3H), 2.25 (t, J = 7.4 Hz, 2H), 1.92 (t, J = 7.3 Hz, 2H), 1.85 (p, J = 7.1 Hz, 2H), 1.65 (p, J = 6.3 Hz, 2H), 1.53 (p, J = 6.9
M AN U
Hz, 2H), 1.47 (p, J = 7.4 Hz, 2H), 1.25 (m, 4H). 13C NMR (125 MHz, DMSO-d6): δ 171.8, 169.6, 159.3, 148.8, 141.0, 140.2, 138.5, 130.2, 129.9, 125.5, 123.2, 111.8, 109.4, 105.9, 101.2, 98.3, 67.5, 61.9, 61.2, 56.5, 48.0, 36.9, 32.8, 28.9, 28.6, 26.6,
EP
564.2689.
TE D
25.5. HR-ESI-MS m/z: (M+Na)+ caclcd for C29H39N3O7Na 564.2686, found
5.8
AC C
N-Hydroxy-7-(4-(2-(5,6,7-trimethoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptana mide (6g)
Following the procedure as described for 6a, reaction of 5g (250 mg, 0.50 mmol) in THF (20 mL) with ethyl chloroformate (0.098 mL, 1.00 mmol) and triethylamine (0.14 mL, 1.00 mmol) gave 6g (236 mg, 92%) as a solid. Mp 73-78 oC. 1H NMR (500
34
ACCEPTED MANUSCRIPT MHz, DMSO-d6): δ 10.30 (s, 1H), 9.66 (s, 1H), 8.62 (s, 1H), 7.44 (d, J = 9.0 Hz, 2H), 7.22 (d, J = 3.1Hz, 1H), 6.83 (s, 1H), 6.81 (d, J = 9.0 Hz, 2H), 6.29 (d, J = 3.0 Hz,
RI PT
1H), 4.60 (t, J = 5.5 Hz, 2H), 4.21 (t, J = 5.5 Hz, 2H), 3.93 (s, 3H), 3.77 (s, 3H), 3.75 (s, 3H), 2.22 (t, J = 7.3 Hz, 2H), 1.92 (t, J = 7.3 Hz, 2H), 1.54 (t, J = 7.0 Hz, 2H), 1.47 13
C NMR (125 MHz, DMSO-d6): δ 171.5, 169.6,
SC
(t, J = 7.0 Hz, 2H), 1.25 (m, 4H).
148.9, 148.4, 140.1, 138.6, 131.0, 128.0, 125.6, 124.5, 123.1, 121.7, 121.1, 112.4,
M AN U
101.4, 98.5, 68.9, 62.0, 61.3, 56.5, 47.6, 36.8, 32.8, 29.0, 25.6. HR-ESI-MS m/z: (M+Na)+ caclcd for C27H35N3O7Na 536.2373, found 536.2365.
TE D
5.9
N-Hydroxy-7-(4-(3-(5,6,7-trimethoxy-1H-indol-1-yl)propoxy)phenylcarbamoyl)hepta
EP
namide (6h)
Following the procedure as described for 6a, reaction of 5h (342 mg, 0.67 mmol)
AC C
in THF (20 mL) with ethyl chloroformate (0.13 mL, 1.34 mmol) and triethylamine (0.19 mL, 1.34 mmol) gave 6h (295 mg, 84%) as a solid. Mp 73-78 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.31 (s, 1H), 9.69 (s, 1H), 7.46 (d, J = 9.0 Hz, 2H), 7.12 (d, J = 3.9 Hz, 1H), 6.82 (d, J = 9.0 Hz, 1H), 6.83 (s, 1H), 6.25 (d, J = 3.0 Hz, 1H), 4.38 (t, J = 7.0 Hz, 1H), 3.93 (s, 3H), 3.84 (t, J = 6.1 Hz, 2H), 3.76 (s, 3H), 3.73 (s, 3H), 2.33 (t,
35
ACCEPTED MANUSCRIPT J = 7.4 Hz, 2H), 2.13 (p, J = 6.3 Hz, 2H), 1.93 (t, J = 7.3 Hz, 2H), 1.53 (p, J = 6.8 Hz, 2H), 1.47 (p, J = 7.1 Hz, 2H), 1.26 (m, 4H). 13C NMR (125 MHz, DMSO-d6): δ 171.7,
RI PT
169.6, 149.8, 148.8, 140.2, 138.5, 131.1, 128.0, 125.5, 125.0, 123.2, 123.1, 120.7, 115.1, 112.8, 101.2, 98.3, 68.4, 61.9, 61.2, 56.5, 47.9, 36.6, 32.8, 29.0, 26.5, 25.7,
SC
25.6. HR-ESI-MS m/z: (M+Na)+ caclcd for C28H37N3O7Na 550.2529, found 550.2527.
M AN U
5.10
N-Hydroxy-7-(4-(4-(5,6,7-trimethoxy-1H-indol-1-yl)butoxy)phenylcarbamoyl)heptana
TE D
mide (6i)
Following the procedure as described for 6a, reaction of 5i (261 mg, 0.49 mmol)
EP
in THF (20 mL) with ethyl chloroformat (0.10 mL, 0.98 mmol) and triethylamine (0.14 mL, 0.98 mmol) gave 6i (257 mg, 96%) as a solid. Mp 110-115 oC. 1H NMR
AC C
(500 MHz, DMSO-d6): δ 7.37 (d, J = 9.0 Hz, 2H), 7.04 (d, J = 3.0 Hz, 1H), 6.83 (s, 1H), 6.79 (d, J = 9.0 Hz, 2H), 6.28 (d, J = 3.0 Hz, 1H), 4.34 (t, J = 7.0 Hz, 2H), 3.96 (s, 3H), 3.89 (t, J = 6.3 Hz, 2H), 3.83 (s, 3H), 3.82 (s, 3H), 2.32 (t, J = 7.4 Hz, 2H), 2.08 (t, J = 7.4 Hz, 2H), 1.95 (p, J = 7.3 Hz, 2H), 1.70 (p, J = 8.0 Hz, 2H), 1.68 (p, J = 6.7 Hz, 2H), 1.62 (p, J = 7.1 Hz, 2H), 1.38 (m, 4H). 13C NMR (125 MHz, DMSO-d6):
36
ACCEPTED MANUSCRIPT δ 171.9, 169.8, 148.8, 140.2, 138.5, 130.4, 127.6, 125.6, 125.3, 123.4, 123.2, 120.7, 112.4, 101.3, 98.4, 65.7, 62.1, 61.2, 56.5, 45.2, 36.6, 32.7, 31.2, 28.9, 25.7, 25.5.
RI PT
HR-ESI-MS m/z: (M+Na)+ caclcd for C29H39N3O7Na 564.2686, found 564.2682. 5.11
SC
N-Hydroxy-7-(3-(2-(5,6-dimethoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptanami de (6j)
M AN U
Following the procedure as described for 6a, reaction of 5j (84 mg, 0.18 mmol) in THF (10 mL) with ethyl chloroformate (0.03 mL, 0.36 mmol) and triethylamine
TE D
(0.05 mL, 0.36 mmol) gave 6j (61 mg, 71%) as a solid. Mp 55-62 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.31 (s, 1H), 9.80 (s, 1H), 8.64 (s, 1H), 7.30 (s, 1H), 7.21 (d, J =
EP
3.1 Hz, 1H), 7.14 (t, J = 9.9 Hz, 2H ), 7.04 (d, J = 8.1 Hz, 2H), 6.55 (dd, J = 2.2, 8.0 Hz, 1H), 6.28 (t, J = 3.0 Hz, 1H), 4.48 (t, J = 5.1 Hz, 2H), 4.20 (t, J = 5.2 Hz, 2H),
AC C
3.79 (s, 3H), 3.72 (s, 3H), 2.24 (t, J = 7.4 Hz, 2H), 1.92 (t, J = 7.3 Hz, 2H), 1.52 (p, J = 6.8 Hz, 2H), 1.47 (p, J = 7.0 Hz, 2H), 1.24 (m, 4H).
13
C NMR (125 MHz,
DMSO-d6): δ 171.8, 169.6, 158.8, 146.9, 145.0, 141.0, 131.1, 130.0, 127.6, 121.1, 112.0, 109.1, 106.0, 103.3, 100.9, 94.6, 67.5, 56.4, 56.3, 45.5, 36.9, 32.7, 28.9, 25.5, 25.4. HR-ESI-MS m/z: (M+Na)+ caclcd for C26H33N3O6Na 506.2267, found
37
ACCEPTED MANUSCRIPT 506.2262. 5.12
RI PT
N-Hydroxy-7-(3-(2-(5-methoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptanamide (6k)
SC
Following the procedure as described for 6a, reaction of 5k (218 mg, 0.50 mmol) in THF (20 mL) with ethyl chloroformat (0.09 mL, 1.00 mmol) and triethylamine
M AN U
(0.14 mL, 1.00 mmol) gave 6k (160 mg, 71%) as a solid. Mp 125-128 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.30 (s, 1H), 9.77 (s, 1H), 8.62 (s, 1H), 7.43 (d, J = 8.9 Hz,
TE D
2H), 7.34 (d, J = 3.0 Hz, 1H), 7.26 (s, 1H), 7.12 (t, J = 8.2 Hz, 1H), 7.05 (d, J = 8.2 Hz, 1H), 7.02 (d, J = 2.3 Hz, 1H), 6.76 (dd, J = 2.3, 8.8 Hz, 1H), 6.53 (dd, J = 1.8, 8.1
EP
Hz, 1H), 6.33 (d, J = 3.0 Hz, 1H), 4.49 (t, J = 5.2 Hz, 2H), 4.19 (t, J = 5.2 Hz, 2H), 3.73 (s, 3H), 2.23 (t, J = 7.4 Hz, 2H), 1.91 (t, J = 7.4 Hz, 2H), 1.53 (p, J = 6.7 Hz, 2H),
AC C
1.46 (p, J = 7.1 Hz, 2H), 1.24 (m, 4H).
13
C NMR (125 MHz, DMSO-d6): δ 171.7,
169.6, 158.8, 153.9, 141.0, 131.7, 129.9, 129.0, 112.1, 111.6, 111.1, 109.2, 105.9, 102.6, 100.9, 67.3, 55.8, 45.6, 36.9, 32.7, 28.9, 25.5, 25.4. HR-ESI-MS m/z: (M+Na)+ caclcd for C25H31N3O5Na 476.2161, found 476.2157. 5.13
38
ACCEPTED MANUSCRIPT N-Hydroxy-7-(3-(2-(6-methoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptanamide (6l)
RI PT
Following the procedure as described for 6a, reaction of 5l (83 mg, 0.19 mmol) in THF (10 mL) with ethyl chloroformate (0.02 mL, 0.38 mmol) and triethylamine (0.05
SC
mL, 0.38 mmol) gave 6l (86 mg, 99%) as a solid. Mp 130-134 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.31 (s, 1H), 9.80 (s, 1H), 8.64 (s, 1H), 7.38 (d, J=8.6 Hz, 1H),
M AN U
7.30 (s, 1H), 7.26 (d, J = 3.2 Hz, 1H), 7.14 (t, J = 8.1 Hz, 1H), 7.06 (t, J = 8.1 Hz, 2H), 6.65 (dd, J = 2.0, 8.6 Hz, 1H), 6.55 (dd, J = 2.0, 8.2 Hz, 1H), 6.34 (d, J = 3.1 Hz, 1H),
TE D
4.50 (t, J = 5.2 Hz, 2H), 4.21 (t, J = 5.2 Hz, 2H), 3.78 (s, 3H), 2.24 (t, J = 7.4 Hz, 2H), 1.92 (t, J = 7.3 Hz, 2H), 1.52 (p, J = 7.1 Hz, 2H), 1.47 (t, J = 7.1 Hz, 2H), 1.24 (m, 13
C NMR (125 MHz, DMSO-d6): δ 171.8, 169.6, 158.8, 156.1, 141.0, 137.2,
EP
4H).
129.9, 128.3, 122.6, 121.3, 112.1, 109.8, 109.1, 106.0, 101.2, 93.9, 67.3, 55.8, 45.4,
AC C
36.9, 32.7, 28.9, 25.5, 25.4. HR-ESI-MS m/z: (M+Na)+ caclcd for C25H31N3O5Na 476.2161, found 476.2168. 5.14 N-Hydroxy-7-(3-(2-(7-methoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptanamide (6m)
39
ACCEPTED MANUSCRIPT Following the procedure as described for 6a, reaction of 5m (223 mg, 0.51 mmol) in THF (20 mL) with ethyl chloroformate (0.09 mL, 1.02 mmol) and triethylamine
RI PT
(0.14 mL, 1.02 mmol) gave 6m (231 mg, 84%) as a solid. Mp 105-111 oC. 1H NMR (500 MHz, DMSO-d6): δ 10.31 (s, 1H), 9.79 (s, 1H), 8.64 (s, 1H), 7.29 (d, J = 3.0 Hz,
SC
1H), 7.25 (s, 1H), 7.13 (t, J = 8.0 Hz, 1H), 7.10 (d, J = 8.4 Hz, 1H), 7.07 (d, J = 8.2 Hz, 1H), 6.90 (t, J = 7.9 Hz, 1H), 6.66 (t, J = 7.7 Hz, 1H), 6.55 (dd, J = 2.0, 8.0 Hz,
M AN U
1H), 6.37 (d, J = 3.0 Hz, 1H), 4.71 (t, J = 5.5 Hz, 2H), 4.20 (t, J = 5.5 Hz, 2H), 3.87 (s, 3H), 2.24 (t, J = 7.3 Hz, 2H), 1.92 (t, J = 7.3 Hz, 2H), 1.52 (p, J = 6.9 Hz, 2H), 1.47 13
C NMR (125 MHz, DMSO-d6): δ 171.7, 169.6,
TE D
(p, J = 6.9 Hz, 2H), 1.24 (m, 4H).
158.8, 147.6, 141.0, 131.1, 130.7, 129.9, 125.4, 120.2, 113.9, 112.0, 109.2, 106.0,
EP
103.1, 101.5, 68.3, 56.0, 48.2, 36.9, 32.7, 28.9, 25.5, 25.4. HR-ESI-MS m/z: (M+Na)+ caclcd for C25H31N3O5Na 476.2161, found 476.2159.
AC C
5.15 N-Hydroxy-7-(3-(2-(1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptanamide (6n) Following the procedure as described for 6a, reaction of 5n (150 mg, 0.37 mmol)
in THF (20 mL) with ethyl chloroformate (0.07 mL, 0.74 mmol) and triethylamine (0.10 mL, 0.74 mmol) gave 6n (120 mg, 77%) as a solid. Mp 105-111 oC. 1H NMR (400 MHz, DMSO-d6): δ 10.32 (s, 1H), 9.80 (s, 1H), 8.65 (s, 1H), 7.56 (t, J = 8.5 Hz,
40
ACCEPTED MANUSCRIPT 2H), 7.43 (d, J = 3.0 Hz, 1H), 7.29 (s, 1H), 7.15 (t, J = 7.8 Hz, 2H), 7.08 (d, J = 8.0 Hz, 1H), 7.03 (t, J = 7.5 Hz, 1H), 6.56 (d, J = 6.9 Hz, 1H), 6.45 (d, J = 2.9 Hz, 1H),
RI PT
4.57 (t, J = 5.1 Hz, 2H), 4.24 (t, J = 5.0 Hz, 2H), 2.26 (t, J = 7.2 Hz, 2H), 1.94 (t, J = 7.2 Hz, 2H), 1.57 (t, J = 6.8 Hz, 2H), 1.49 (t, J = 6.8 Hz, 2H), 1.27 (m, 4H). 13C NMR
SC
(100 MHz, DMSO-d6): δ 171.7, 169.6, 158.8, 140.9, 136.4, 129.9, 129.5, 128.6, 121.5, 120.8, 119.5, 112.1, 110.4, 109.2, 105.9, 101.2, 67.3, 45.5, 36.9, 32.7, 28.9, 25.5, 25.4.
M AN U
HR-ESI-MS m/z: (M+Na)+ caclcd for C24H29N3O4Na 446.2056, found 446.2050. 5.16
(6o)
TE D
N-Hydroxy-7-(3-(2-(5-bromo-1H-indol-1-yl)ethoxy)phenylcarbamoyl)heptanamide
EP
Following the procedure as described for 6a, reaction of 5o (100 mg, 0.20 mmol) in THF (10 mL) with ethyl chloroformate (0.04 mL, 0.40 mmol) and triethylamine
AC C
(0.05 mL, 0.40 mmol) gave 6o (92 mg, 87%) as a solid. Mp 141-143 oC. 1H NMR (400 MHz, DMSO-d6): δ 10.32 (s, 1H), 9.79 (s, 1H), 8.65 (s, 1H), 7.73 (s, 1H), 7.57 (d, J = 8.7 Hz, 1H), 7.49 (d, J = 3.0 Hz, 1H), 7.26 (m, 2H), 7.14 (t, J = 8.1 Hz, 1H), 7.08 (d, J = 8.0 Hz, 1H), 6.54 (d, J = 8.1 Hz, 1H), 6.45 (d, J = 2.9 Hz, 1H), 4.57 (t, J = 5.0 Hz, 2H), 4.23 (t, J = 5.0 Hz, 2H), 2.26 (t, J = 7.3 Hz, 2H), 1.94 (t, J = 7.3 Hz, 2H),
41
ACCEPTED MANUSCRIPT 1.56 (p, J = 6.6 Hz, 2H), 1.49 (p, J = 6.6 Hz, 2H), 1.27 (m, 4H). 13C NMR (100 MHz, DMSO-d6): δ171.7, 169.5, 158.7, 140.9, 135.2, 131.0, 130.4, 129.9, 123.9, 122.9,
RI PT
112.5, 112.1, 109.1, 105.9, 100.9, 67.3, 45.7, 36.8, 32.7, 28.8, 25.5, 25.4. HR-ESI-MS m/z: (M+H)+ caclcd for C24H29O4N3Br 502.1336, found 502.1336.
SC
5.17
N-Hydroxy-5-(3-(2-(5-methoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)pentanamide
M AN U
(6p)
Following the procedure as described for 6a, reaction of 5p (100 mg, 0.24 mmol) in THF (20 mL) with ethyl chloroformate (0.04 mL, 0.48 mmol) and triethylamine
TE D
(0.07 mL, 0.48 mmol) gave 6p (89 mg, 79%) as a solid. Mp 90-93 oC. 1H NMR (400
EP
MHz, DMSO-d6): δ 10.35 (s, 1H), 9.82 (s, 1H), 8.67 (s, 1H), 7.46 (d, J = 8.9 Hz, 1H), 7.37 (d, J = 2.4 Hz, 1H), 7.28 (s, 1H), 7.15 (t, J = 8.0 Hz, 1H), 7.06 (m, 2H), 6.79 (d,
AC C
J = 8.9 Hz, 1H), 6.55 (d, J = 8.2 Hz, 1H), 6.35 (d, J = 2.3 Hz, 1H), 4.52 (t, J = 5.0 Hz, 2H), 4.21 (t, J = 5.0 Hz, 2H), 3.75 (s, 3H), 2.27 (t, J = 6.6 Hz, 2H), 1.97 (t, J = 6.6 Hz, 2H), 1.53 (m, 4H).
13
C NMR (100 MHz, DMSO-d6): δ 171.6, 169.4, 158.8, 153.9,
140.9, 131.7, 129.9, 128.9, 112.1, 111.6, 111.1, 109.2, 105.9, 102.6, 100.9, 67.4, 55.8, 45.6, 36.7, 32.6, 25.3, 25.2. HR-ESI-MS m/z: (M+H)+ caclcd for C23H28O5N3
42
ACCEPTED MANUSCRIPT 426.2023, found 426.2024. 5.18
RI PT
N-Hydroxy-8-(3-(2-(5-methoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)octanamide (6q)
SC
Following the procedure as described for 6a, reaction of 5q (100 mg, 0.22 mmol) in THF (10 mL) with ethyl chloroformate (0.04 mL, 0.44 mmol) and triethylamine
M AN U
(0.06 mL, 0.44 mmol) gave 6q (91 mg, 89%) as a solid. Mp 102-104 oC. 1H NMR (400 MHz, DMSO-d6): δ 10.32 (s, 1H), 9.82 (s, 1H), 8.64 (s, 1H), 7.46 (d, J = 8.9 Hz,
TE D
1H), 7.37 (d, J = 2.8 Hz, 1H), 7.29 (s, 1H), 7.14 (t, J = 8.0 Hz, 1H), 7.07 (m, 2H), 6.79 (d, J = 8.9 Hz, 1H), 6.55 (d, J = 8.1 Hz, 1H), 6.35 (d, J = 2.6 Hz, 1H), 4.52 (t, J =
EP
5.0 Hz, 2H), 4.21 (t, J = 5.0 Hz, 2H), 3.75 (s, 3H), 2.26 (t, J = 7.4 Hz, 2H), 1.94 (t, J = 7.4 Hz, 2H), 1.56 (p, J = 6.9 Hz, 2H), 1.48 (p, J = 6.9 Hz, 2H), 1.26 (m, 6H).
13
C
AC C
NMR (100 MHz, DMSO-d6): δ171.7, 169.5, 158.7, 153.9, 140.9, 131.6, 129.9, 128.9, 112.1, 111.5, 111.0, 109.1, 105.9, 102.6, 100.8, 67.3, 55.8, 45.6, 36.9, 32.7, 29.0, 28.9, 25.5, 25.4. HR-ESI-MS m/z: (M+H)+ caclcd for C26H34O5N3 468.2493, found 468.2494. 5.19
43
ACCEPTED MANUSCRIPT N-Hydroxy-9-(3-(2-(5-methoxy-1H-indol-1-yl)ethoxy)phenylcarbamoyl)nonanamide (6r)
RI PT
Following the procedure as described for 6a, reaction of 5r (100 mg, 0.21 mmol) in THF (10 mL) with ethyl chloroformate (0.04 mL, 0.42 mmol) and triethylamine
SC
(0.06 mL, 0.42 mmol) gave 6r (84 mg, 83%) as a solid. Mp 135-137 oC. 1H NMR (400 MHz, DMSO-d6): δ 10.31 (s, 1H), 9.79 (s, 1H), 8.64 (s, 1H), 7.46 (d, J = 8.8 Hz,
M AN U
1H), 7.36 (m, 1H), 7.29 (s, 1H), 7.14 (t, J = 8.1 Hz, 1H), 7.07 (m, 2H), 6.79 (d, J = 8.9 Hz, 1H), 6.55 (d, J = 8.1 Hz, 1H), 6.35 (m, 1H), 4.52 (t, J = 5.0 Hz, 2H), 4.21 (t, J
TE D
= 5.0 Hz, 2H), 3.75 (s, 3H), 2.26 (t, J = 7.4 Hz, 2H), 1.93 (t, J = 7.4 Hz, 2H), 1.56 (p, J = 6.4 Hz, 2H), 1.48 (p, J = 6.4 Hz, 2H), 1.26 (m, 8H).
13
C NMR (100 MHz,
EP
DMSO-d6): δ171.7, 169.5, 158.7, 153.9, 140.9, 131.6, 129.9, 128.9, 112.1, 111.6, 111.0, 109.1, 105.9, 102.6, 100.8, 67.3, 55.8, 45.6, 36.9, 32.7, 29.1, 29.0, 25.5.
AC C
HR-ESI-MS m/z: (M+H)+ caclcd for C27H36O5N3 482.2649, found 482.2649. 5.20 Preparation of HDAC4 and HDAC8 Genes encoding HDAC4 (residues 648-1057) and HDAC8 (residues 1-377) flanked with NdeI and EcoRI sites at the 5ʹ- and 3ʹ - ends, respectively, were synthesized by GenScript Corporation (NJ, USA) and subcloned into expression
44
ACCEPTED MANUSCRIPT vectors pET-28a(+) and pET- 24b(+), respectively. Proteins were expressed in BL21(DE3) cells by overnight induction with IPTG (1 mM) at 20-25 ºC and purified
RI PT
from cleared cell lysates by sequential chromatography on Ni-Sepharose 6 fast flow, Mono Q 5/50 GL, and Superdex 75 10/ 300 GL columns (GE Healthcare). Protein
SC
concentrations were quantified with Bradford Reagent (Bio-Rad). 5.21 HDAC activity assay
M AN U
The HDAC activity assay was performed as described previously73. Enzyme, inhibitors and substrate were diluted with HDAC buffer (15 mM TriseHCl pH 8.1,
TE D
0.25 mM EDTA, 250 mM NaCl, 10% v:v glycerol). Briefly, 10 mL diluted HDAC, i.e., HeLa nuclear extract (Enzo), HDAC1 (BPS), HDAC 2 (BPS), HDAC3 (BPS),
EP
HDAC6 (BPS), HDAC4 and HDAC8 and 50 mL test compound solution at varying concentrations were added to each well of a 96-well microtiter plate and
AC C
pre-incubated at 30 ºC for 5 min. The enzymatic reaction was started by adding 40 mL substrate, i.e., Boc-Lys(Ac)-AMC (Bachem) for HeLa nuclear extract and HDAC6, Boc-Lys(TFA)-AMC (Bachem) for HDAC4, -8 and KI 177 (Enzo) for HDAC1, -2 and -3 in HDAC buffer. After incubation at 30 ºC for 30 min, the reaction was stopped by adding 100 mL trypsin solution (10 mg/mL trypsin in 50 mM
45
ACCEPTED MANUSCRIPT Tris-HCl pH 8, 100 mM NaCl, 2 mM SAHA). After incubation at 30 ºC for another 20 min, fluorescence was measured (excitation λ = 355 nm, emission λ = 460 nm). To
RI PT
calculate IC50 values, the fluorescence in wells without test compound (0.1% DMSO, negative control) was set as 100% enzymatic activity, and the fluorescence in wells
were performed in triplicate.
M AN U
5.22 Molecular docking and sequence analysis
SC
with 2µM SAHA (positive control) was set at 0% enzymatic activity. All experiments
The crystal structures of HDAC1 (PDB ID: 4BKX), HDAC2 (PDB ID: 4LXZ),
TE D
HDAC3 (PDB ID: 4A96), HDAC4 (PDB ID: 4CBT), HDAC6 (PDB ID: 5EEI), HDAC7 (PDB ID: 3C10), and HDAC8 (PDB ID: 5FUE) were obtained from RCSB
EP
protein data bank, with binding sites prepared in the co-crystallized ligand. The 3D structures of ligands generated by ACD/ChemSketch were docked into the protein
AC C
binding site of 4BKX and 4CBT. Compounds were docked in SYBYL [29] using default settings. The above HDAC structures were prepared within SYBYL, with the removal of any water molecules. AMBER7 FF99 and Gasteiger-Marsili charges were added to proteins and ligands. The docking mode of Surflex-Dock GeomX (SFXC) was apllied for docking. The charge of Zn ion was assigned with +2. The binding site
46
ACCEPTED MANUSCRIPT was defined by the bound ligands and prepares using SYBYL. Poses that coordinated with the Zn ion were selected for further analysis. Poses and interations were analysed
RI PT
with the Post-Screening Analysis function in iGEMDOCK. Mutliple sequence analysis of human HDACs were performed using Jalview [31] under T-Coffee
SC
parameters. Amino acid sequences were obtained through the Uniprot database [48] . 5.23 Cancer cell lines and cell culture
M AN U
Human prostate cancer PC-3 cells and human lung cancer A549 (American Type Culture Collection, Rockville, MD) were cultured in RPMI 1640 medium with 10%
TE D
FBS (v/v), penicillin (100 U/ml) and streptomycin (100 mg/ml). Cells were maintained in a humidified incubator at 37 ºC in an atmosphere of CO2: air (5: 95).
EP
5.24 The sulforhodamine B (SRB) growth inhibition assay Cells were seeded in 96-well plates in medium containing 5% FBS. After 24 h,
AC C
cells of T0 group were fixed with 10% trichloroacetic acid (TCA) to obtain a representative cell population at the time of hydroxamate addition (T0). After incubation of vehicle (0.1% DMSO) or hydroxamates (all dissolved in media without apparent precipitation) for an additional 48 h, cells were fixed with 10% TCA, and SRB at 0.4% (w/v) in 1% acetic acid was added to the stained cells. Unbound SRB
47
ACCEPTED MANUSCRIPT was washed out by 1% acetic acid, and SRB bound cells were solubilized with 10 mM Trizma base. The absorbance was read at a wavelength of 515 nm. The
RI PT
absorbance measurements obtained at time zero (T0) for each compound concentration level were control growth (C), cell growth in the presence of
SC
hydroxamates (Tx), and percentage growth. Percentage growth inhibition was calculated as [1-(Tx-T0)/(C-T0)] x 100% for concentrations for which Tx ≥ T0.
M AN U
Growth inhibition of 50% (GI50) was defined as the drug concentration that resulted in a 50% reduction in the increase in total protein in control cells during incubation of
TE D
the compound. 5.25 Western blot analysis
EP
After 24 h treatment with compounds, the cells were rapidly washed with PBS then lysed in ice-cold lysis buffer (50 mM TriseHCl, pH 7.4, 1 mM EGTA, 150 mM
AC C
NaCl, 1% Triton X-100, 1 mM PMSF, 5 mg/mL leupeptin, 20 mg/mL aprotinin, 1mM NaF, and 1 mM Na3VO4). The lysates were subjected to SDS-PAGE by using 13% polyacrylamide gels; protein bands were transferred to a nitrocellulose membrane, and immunoblot analysis was performed as described in the literature. Briefly, the membrane was successively incubated with various antibodies at room
48
ACCEPTED MANUSCRIPT temperature for 1 h with 0.1% milk in TTBS. The antibodies included monoclonal antibody specific for β-actin (Santa Cruz biotechnology), polyclonal antibody specific
RI PT
for Ac-histone H3 (Upstate) and monoclonal antibody specific for Ac-α-tubulin (Sigma), and then with horseradish peroxidase-labeled anti-rabbit secondary antibody
SC
for 30 min. After each incubation, the membrane was thoroughly washed with TTBS. Immunoreactive bands were detected by enhanced chemiluminescence (ECL) and
5.26 Cell cycle distribution assay
M AN U
developed with Hyperfilm-ECL (GE Healthcare).
Cell cycle distribution was assayed by flow cytometry. After treatment of
TE D
compounds at indicated concentrations, the cells were harvested by trypsinization, fixed with 70% (v/v) alcohol at 4 ºC for 30 min, and washed with phosphate-buffered
EP
saline (PBS). Cells were centrifuged and incubated in 0.1 M of phosphate-citric acid
AC C
buffer (0.2 M NaHPO4, 0.1 M citric acid, pH 7.8) for 30 mins at room temperature. After another cycle of centrifugation, the cells were resuspended with 0.5 ml PI solution containing Triton X-100 (0.1% v/v), RNase (100 mg/ml), and PI (80 mg/ml). The DNA content was analyzed with the FACScan (Becton Dickinson, Mountain View, CA) and calculated using CellQuest software. Acknowledgements. We gratefully acknowledge the support from the Ministry of 49
ACCEPTED MANUSCRIPT Science
and
Technology
(MOST
105-2311-B-038-001
and
MOST105-2320-B-038-024-MY3) and Health and welfare surcharge of tobacco
partially
supported
by
the
Taiwan
Protein
RI PT
products (MOHW106-TDU-B-212-144001) in Taiwan. This research was also Project
(Grant
No.
SC
MOST105-0210-01-12-01 and Grant No. MOST106-0210-01-15-04). References
389 (1997) 349-352.
M AN U
[1] M. Grunstein, Histone acetylation in chromatin structure and transcription, Nature [2] P.A. Wade, Transcriptional control at regulatory checkpoints by histone deacetylases: molecular connections between cancer and chromatin, Human Mol. Genet. 10 (2001) 693-698.
[3] W.D. Cress, E. Seto, Histone deacetylases, transcriptional control, and cancer, J.
TE D
Cell Physiol. 184 (2000) 1-16.
[4] R.W. Johnstone, Histone-deacetylase inhibitors: novel drugs for the treatment of cancer, Nat. Rev. Drug Discov. 1 (2002) 287-299. [5] M. Jung, Inhibitors of histone deacetylase as new anticancer agents, Curr. Med.
EP
Chem. 8 (2001) 1505-1511.
[6] I.V. Gregoretti, Y.M. Lee, H.V. Goodson, Molecular evolution of the histone
AC C
deacetylase family: functional implications of phylogenetic analysis, J. Mol. Biol. 338 (2004) 17-31.
[7] M. Paris, M. Porcelloni, M. Binaschi, D. Fattori, Histone deacetylase inhibitors: from bench to clinic, J. Med. Chem. 51 (2008) 1505-1529. [8] A. Clocchiatti, C. Florean, C. Brancolini, Class IIa HDACs: from important roles in differentiation to possible implications in tumourigenesis, J. Cell Mol. Med. 15 (2011) 1833-1846. [9] M.S. Kim, M.W. Akhtar, M. Adachi, M. Mahgoub, R. Bassel-Duby, E.T. Kavalali, E.N. Olson, L.M. Monteggia, An Essential Role for Histone Deacetylase 4 in Synaptic Plasticity and Memory Formation, J. Neurosci. 32 (2012) 10879-10886. [10] R.B. Vega, K. Matsuda, J. Oh, A.C. Barbosa, X.L. Yang, E. Meadows, J. 50
ACCEPTED MANUSCRIPT McAnally, C. Pomajzl, J.M. Shelton, J.A. Richardson, G. Karsenty, E.N. Olson, Histone deacetylase 4 controls chondrocyte hypertrophy during skeletogenesis, Cell 119 (2004) 555-566. [11] S.R. Chang, B.D. Young, S.J. Li, X.X. Qi, J.A. Richardson, E.N. Olson, Histone deacetylase 7 maintains vascular integrity by repressing matrix metalloproteinase 10,
RI PT
Cell 126 (2006) 321-334. [12] E. Telles, E. Seto, Modulation of cell cycle regulators by HDACs, Front. Biosci. 4 (2012) 831-839.
[13] T. Eckschlager, J. Plch, M. Stiborova, J. Hrabeta, Histone Deacetylase Inhibitors as Anticancer Drugs, Int. J. Mol. Sci. 18 (2017) 1414.
SC
[14] K.M. VanderMolen, W. McCulloch, C.J. Pearce, N.H. Oberlies, Romidepsin (Istodax, NSC 630176, FR901228, FK228, depsipeptide): a natural product recently
M AN U
approved for cutaneous T-cell lymphoma, J. Antibiot. 64 (2011) 525-531. [15] H.Z. Lee, V.E. Kwitkowski, P.L. Del Valle, M.S. Ricci, H. Saber, B.A. Habtemariam, J. Bullock, E. Bloomquist, Y. Li Shen, X.H. Chen, J. Brown, N. Mehrotra, S. Dorff, R. Charlab, R.C. Kane, E. Kaminskas, R. Justice, A.T. Farrell, R. Pazdur, FDA Approval: Belinostat for the Treatment of Patients with Relapsed or Refractory Peripheral T-cell Lymphoma, Clin. Cancer Res. 21 (2015) 2666-2670.
TE D
[16] L.A. Raedler, Farydak (Panobinostat): First HDAC Inhibitor Approved for Patients with Relapsed Multiple Myeloma, Am. Health Drug Benefits 9 (2016) 84-87. [17] B.E. Gryder, Targeted cancer therapy: giving histone deacetylase inhibitors all they need to succeed, Future Med. Chem. 4 (2012) 1369-1370.
EP
[18] K.J. Falkenberg, R.W. Johnstone, Histone deacetylases and their inhibitors in cancer, neurological diseases and immune disorders, Nat. Rev. Drug Discov. 13 (2014)
AC C
673-691.
[19] H.Y. Lee, J.Y. Chang, L.Y. Chang, W.Y. Lai, M.J. Lai, K.H. Shih, C.C. Kuo, C.Y. Chang, J.P. Liou, Concise syntheses of N-aryl-5,6,7-trimethoxyindoles as antimitotic and vascular disrupting agents: application of the copper-mediated Ullmann-type arylation, Org. Biomol. Chem. 9 (2011) 3154-3157. [20] D.G. Zhao, J. Chen, Y.R. Du, Y.Y. Ma, Y.X. Chen, K. Gao, B.R. Hu, Synthesis and structure-activity relationships of N-methyl-5,6,7-trimethoxylindoles as novel antimitotic and vascular disrupting agents, J. Med. Chem. 56 (2013) 1467-1477. [21] M. Arthuis, R. Pontikis, G.G. Chabot, L. Quentin, D. Scherman, J.C. Florent, Domino approach to 2-aroyltrimethoxyindoles as novel heterocyclic combretastatin A4 analogues, Eur. J. Med. Chem. 46 (2011) 95-100. 51
ACCEPTED MANUSCRIPT [22] M. Ichimura, T. Ogawa, S. Katsumata, K. Takahashi, I. Takahashi, H. Nakano, Duocarmycins, new antitumor antibiotics produced by Streptomyces; producing organisms and improved production, J. Antibio. 44 (1991) 1045-1053. [23] M. Ichimura, T. Ogawa, K. Takahashi, E. Kobayashi, I. Kawamoto, T. Yasuzawa, I. Takahashi, H. Nakano, Duocarmycin SA, a new antitumor antibiotic from
RI PT
Streptomyces sp, J. Antibio. 43 (1990) 1037-1038. [24] G.J. Atwell, J.J. Milbank, W.R. Wilson, A. Hogg, W.A. Denny, 5-Amino-1-(chloromethyl)-1,2-dihydro-3H-benz[e]indoles:
relationships
between
structure and cytotoxicity for analogues bearing different DNA minor groove binding subunits, J. Med. Chem. 42 (1999) 3400-3411.
SC
[25] W.J. Huang, C.C. Chen, S.W. Chao, S.S. Lee, F.L. Hsu, Y.L. Lu, M.F. Hung, C.I. Chang, Synthesis of N-hydroxycinnamides capped with a naturally occurring moiety
M AN U
as inhibitors of histone deacetylase, ChemMedChem 5 (2010) 598-607. [26] W.J. Huang, C.C. Chen, S.W. Chao, C.C. Yu, C.Y. Yang, J.H. Guh, Y.C. Lin, C.I. Kuo, P. Yang, C.I. Chang, Synthesis and evaluation of aliphatic-chain hydroxamates capped with osthole derivatives as histone deacetylase inhibitors, Eur. J. Med. Chem. 46 (2011) 4042-4049.
[27] Y.C. Chen, W.J. Huang, J.L. Hsu, C.C. Yu, W.T. Wang, J.H. Guh, A novel
TE D
hydroxysuberamide derivative potentiates MG132-mediated anticancer activity against human hormone refractory prostate cancersthe role of histone deacetylase and endoplasmic reticulum stress, Prostate 73 (2013) 1270-1280. [28] W.J. Huang, Y.A. Tang, M.Y. Chen, Y.J. Wang, F.H. Hu, T.W. Wang, S.W. Chao,
EP
H.W. Chiu, Y.L. Yeh, H.Y. Chang, H.F. Juan, P. Lin, Y.C. Wang, A histone deacetylase inhibitor YCW1 with antitumor and antimetastasis properties enhances cisplatin
AC C
activity against non-small cell lung cancer in preclinical studies, Cancer Lett. 346 (2014) 84-93.
[29] Certara L.P., SYBYL-X Suite Molecular Moedling Software, Version 2.0. Certara L.P., Princeton, NJ, 2012. https://www.certara.com. [30] K.C. Hsu, Y.F. Chen, S.R. Lin, J.M. Yang, iGEMDOCK: a graphical environment of enhancing GEMDOCK using pharmacological interactions and post-screening analysis, BMC Bioinform. 12 Suppl 1 (2011) S33. [31] A.M. Waterhouse, J.B. Procter, D.M.A. Martin, M. Clamp, G.J. Barton, Jalview Version 2-a multiple sequence alignment editor and analysis workbench, Bioinform. 25 (2009) 1189-1191. [32] R.W. Burli, C.A. Luckhurst, O. Aziz, K.L. Matthews, D. Yates, K.A. Lyons, M. 52
ACCEPTED MANUSCRIPT Beconi, G. McAllister, P. Breccia, A.J. Stott, S.D. Penrose, M. Wall, M. Lamers, P. Leonard, I. Muller, C.M. Richardson, R. Jarvis, L. Stones, S. Hughes, G. Wishart, A.F. Haughan, C. O'Connell, T. Mead, H. McNeil, J. Vann, J. Mangette, M. Maillard, V. Beaumont, I. Munoz-Sanjuan, C. Dominguez, Design, Synthesis, and Biological Evaluation of Potent and Selective Class IIa Histone Deacetylase (HDAC) Inhibitors
RI PT
as a Potential Therapy for Huntington's Disease, J. Med. Chem. 56 (2013) 9934-9954. [33] P.A. Marks, Discovery and development of SAHA as an anticancer agent, Oncogene 26 (2007) 1351-1356.
[34] L.A. Stivala, O. Cazzalini, E. Prosperi, The cyclin-dependent kinase inhibitor p21CDKN1A as a target of anti-cancer drugs, Curr. Cancer Drug Targ. 12 (2012)
SC
85-96.
[35] E.A. Musgrove, C.E. Caldon, J. Barraclough, A. Stone, R.L. Sutherland, Cyclin
M AN U
D as a therapeutic target in cancer, Nat. Rev. Cancer, 11 (2011) 558-572. [36] X.C. Xu, Tumor-suppressive activity of retinoic acid receptor-beta in cancer, Cancer Lett. 253 (2007) 14-24.
[37] S.Y. Sun, Retinoic acid receptor beta and colon cancer, Cancer Biol. Ther. 3 (2004) 87-88.
[38] G.T. Baxter, R.C. Kuo, O.J. Jupp, P. Vandenabeele, D.J. MacEwan, Tumor
TE D
necrosis factor-alpha mediates both apoptotic cell death and cell proliferation in a human hematopoietic cell line dependent on mitotic activity and receptor subtype expression, J. Biol. Chem. 274 (1999) 9539-9547. [39] T.E. Vaskivuo, F. Stenback, J.S. Tapanainen, Apoptosis and apoptosis-related
EP
factors Bcl-2, Bax, tumor necrosis factor-alpha, and NF-kappaB in human endometrial hyperplasia and carcinoma, Cancer 95 (2002) 1463-1471.
AC C
[40] P.N. Kelly, A. Strasser, The role of Bcl-2 and its pro-survival relatives in tumourigenesis and cancer therapy, Cell Death Differ. 18 (2011) 1414-1424. [41] A.N. Hata, J.A. Engelman, A.C. Faber, The BCL2 Family: Key Mediators of the Apoptotic Response to Targeted Anticancer Therapeutics, Cancer Discov. 5 (2015) 475-487.
[42] S. Yamada, S. Nomoto, T. Fujii, S. Takeda, N. Kanazumi, H. Sugimoto, A. Nakao, Frequent promoter methylation of M-cadherin in hepatocellular carcinoma is associated with poor prognosis, Anticancer Res. 27 (2007) 2269-2274. [43] M. Cool, P. Jolicoeur, Elevated frequency of loss of heterozygosity in mammary tumors arising in mouse mammary tumor virus/neu transgenic mice, Cancer Res. 59 (1999) 2438-2444. 53
ACCEPTED MANUSCRIPT [44] M.C. Anania, M. Sensi, E. Radaelli, C. Miranda, M.G. Vizioli, S. Pagliardini, E. Favini, L. Cleris, R. Supino, F. Formelli, M.G. Borrello, M.A. Pierotti, A. Greco, TIMP3 regulates migration, invasion and in vivo tumorigenicity of thyroid tumor cells, Oncogene 30 (2011) 3011-3023. [45] M. Brait, M. Loyo, E. Rosenbaum, K.L. Ostrow, A. Markova, S. Papagerakis, M.
RI PT
Zahurak, S.M. Goodman, M. Zeiger, D. Sidransky, C.B. Umbricht, M.O. Hoque, Correlation between BRAF mutation and promoter methylation of TIMP3, RARbeta2 and RASSF1A in thyroid cancer, Epigenet. 7 (2012) 710-719.
[46] J.P. Kallio, S. Hopkins-Donaldson, A.H. Baker, V.M. Kahari, TIMP-3 promotes receptor pathway, Int. J. Cancer. 128 (2011) 991-996.
SC
apoptosis in nonadherent small cell lung carcinoma cells lacking functional death [47] F. van Roy, Beyond E-cadherin: roles of other cadherin superfamily members in
M AN U
cancer, Nat. Rev. Cancer 14 (2014) 121-134.
[48] R. Apweiler, M.J. Martin, C. O'Donovan, M. Magrane, Y. Alam-Faruque, R. Antunes, E.B. Casanova, B. Bely, M. Bingley, L. Bower, B. Bursteinas, W.M. Chan, G. Chavali, A. Da Silva, E. Dimmer, R. Eberhardt, F. Fazzini, A. Fedotov, J. Garavelli, L.G. Castro, M. Gardner, R. Hieta, R. Huntley, J. Jacobsen, D. Legge, W.D. Liu, J. Luo, S. Orchard, S. Patient, K. Pichler, D. Poggioli, N. Pontikos, S. Pundir, S.
TE D
Rosanoff, T. Sawford, H. Sehra, E. Turner, T. Wardell, X. Watkins, M. Corbett, M. Donnelly, P. van Rensburg, M. Goujon, H. McWilliam, R. Lopez, I. Xenarios, L. Bougueleret, A. Bridge, S. Poux, N. Redaschi, G. Argoud-Puy, A. Auchincloss, K. Axelsen, D. Baratin, M.C. Blatter, B. Boeckmann, J. Bolleman, L. Bollondi, E. Boutet,
EP
S.B. Quintaje, L. Breuza, E. deCastro, L. Cerutti, E. Coudert, B. Cuche, I. Cusin, M. Doche, D. Dornevil, S. Duvaud, A. Estreicher, L. Famiglietti, M. Feuermann, S.
AC C
Gehant, S. Ferro, E. Gasteiger, V. Gerritsen, A. Gos, N. Gruaz-Gumowski, U. Hinz, C. Hulo, N. Hulo, J. James, S. Jimenez, F. Jungo, T. Kappler, G. Keller, V. Lara, P. Lemercier, D. Lieberherr, X. Martin, P. Masson, M. Moinat, A. Morgat, S. Paesano, I. Pedruzzi, S. Pilbout, M. Pozzato, M. Pruess, C. Rivoire, B. Roechert, M. Schneider, C. Sigrist, K. Sonesson, S. Staehli, E. Stanley, A. Stutz, S. Sundaram, M. Tognolli, L. Verbregue, A.L. Veuthey, C.H. Wu, C.N. Arighi, L. Arminski, W.C. Barker, C.M. Chen, Y.X. Chen, P. Dubey, H.Z. Huang, A. Kukreja, K. Laiho, R. Mazumder, P. McGarvey, D.A. Natale, T.G. Natarajan, N.V. Roberts, B.E. Suzek, C.R. Vinayaka, Q.H. Wang, Y.Q. Wang, L.S. Yeh, J. Zhang, U. Consortium, Reorganizing the protein space at the Universal Protein Resource (UniProt), Nucleic Acids Res. 40 (2012) D71-D75. 54
ACCEPTED MANUSCRIPT
Figure legends
RI PT
Figure 1. Examples of pan and class I-selective HDAC inhibitors in clinical trials. Figure 2. Molecular docking analysis of inhibitors 6f and 6j in HDAC1. Compound
SC
6f and 6j docked into HDAC 1 (PDB ID: 4BKX) crystal structure. (A-B) Surface view of compound 6j (yellow) and compound 6f (purple) docked in HDAC 1. (C-D)
M AN U
Compounds 6j (yellow) and compound 6f (purple) docking pose stick view of the HDAC1 (grey) active site reveal interactions with residues. (E) Compounds 6f and 6j
TE D
were superimposed in HDAC 1 catalytic center. Residues contributed hydrogen bond are colored in green, residues contributed van der Waals interaction are colored in
EP
brown. H178 contain hydrogen donor and pi-donor is colored in yellow. (F) Heat map of corresponding residue interactions and compounds 6j and 6f. Hydrogen bonds are
AC C
represented in shades of green and van der Waals in shades of grey. Figure 3. Interaction analysis of inhibitors 6f and 6j. (A) Multiple Sequence Alignment (MSA) of HDAC family. Residues were aligned to HDAC1. Amino acids labeled using Crustal X color scheme. (B) The docking pose of inhibitor 6j in HDAC1 (yellow) was aligned with class I and class IIb HDAC6 and (C) class IIa
55
ACCEPTED MANUSCRIPT HDAC4 and HDAC7. (D) All HDAC isozyme structures used previously are aligned. Red circle highlights contrasting residues in alignment. HDACs color is labeled as
RI PT
shown. Figure 4. Surface residues interacts with the inhibitor cap. The surface residues of
SC
class I and HDAC6 (A) and class IIa (B) isozymes, as described in Figure 3, are compared. (C) Alignment of all HDAC structures used with Inhibitor 6j docked in
M AN U
HDAC1, were transposed. Red circle denotes contrasting histidine locations between isozymes. HDACs labeled as shown. The surface model of surface residues of HDAC
TE D
1 (D) and HDAC4 (E) reveal non-interacting pocket (red circle). Figure 5. Effect of compounds 6j and 6k on induction of histone acetylation and
EP
α-tubulin acetylation in cultured human prostate cancer PC-3 cells. Cells were incubated with the HDAC inhibitors at the indicated concentrations for 16 h. The
AC C
whole cell lysates were analyzed for histone and α-tubulin acetylation by SDS-PAGE and Western blot for either acetylated histone or α-tubulin. Figure 6. Effect of compounds 6j and 6k on cell cycle progression in cultured human prostate PC-3 cells. (A) Cells were treated without (ctrl) or with the HDAC inhibitors at the indicated concentrations for 24 h and were analyzed by flow cytometry for cell
56
ACCEPTED MANUSCRIPT cycle distribution. (B) Data were presented by mean value of three independent
AC C
EP
TE D
M AN U
SC
RI PT
experiments.
57
ACCEPTED MANUSCRIPT Table 1 IC50 value for the inhibition against HeLa nuclear HDACa (nM) and cytotoxicity against cancer cells (µM) by compounds 6a-m H N 6
N
O
O
OH
Substitution
Chain
Substitution
position
length (n)
(R)
6a
ortho
2
5,6,7-TriOMe
109.9±1.2
5.6
5.9
6b
ortho
3
5,6,7-TriOMe
111.0±5.2
3.9
5.0
6c
ortho
4
5,6,7-TriOMe
111.4±9.8
2.4
4.8
6d
meta
2
5,6,7-TriOMe
SC
R
nO
RI PT
H N
11.5±0.2
1.3
2.4
6e
meta
3
5,6,7-TriOMe
22.5±0.8
2.7
2.7
6f
meta
4
5,6,7-TriOMe
17.8±1.9
2.1
2.4
6g
para
2
5,6,7-TriOMe
57.2±3.3
4.4
4.9
6h
para
3
5,6,7-TriOMe
138.6±1.1
6.9
5.9
6i
para
4
5,6,7-TriOMe
46.4±0.6
7.7
9.7
6j
meta
2
5,6-DiOMe
11.2±1.3
1.2
6.4
6k
meta
2
5-OMe
8.6±0.7
1.3
1.7
6l
meta
2
6-OMe
14.9±0.1
1.8
4.2
6m
meta
2
7-OMe
13.5±0.4
1.4
1.9
51.6±3.6
1.5
1.7
M AN U
Data was obtained from three independent experiments.
AC C
a
HDAC
TE D
SAHA
EP
Compound
58
PC-3
A549
ACCEPTED MANUSCRIPT Table 2. IC50 value for the inhibition against HeLa nuclear HDACa (nM) by compounds 6n-6r H N
O
N
O
OH
Chain length (n)
Substitution (R)
6n
6
H
6o
6
5-Br
6p
4
5-OMe
185.8±10.3
6q
7
5-OMe
40.0±3.2
6r
8
5-OMe
298.0±14.2
6k
6
5-OMe
8.6±0.7
M AN U
SC
Compound
SAHA
EP
TE D
Data was obtained from three independent experiments.
AC C
a
n O
H N
RI PT
R
59
HDAC
23.1±0.3
41.3±0.9
51.6±3.6
ACCEPTED MANUSCRIPT
Table 3 IC50a (nM) value for the inhibition against class I and class II HDACs by compounds 6d, 6f, 6j and 6k Class I
Class II
Compound HDAC8
HDAC4 HDAC5 HDAC7 HDAC9 HDAC6
RI PT
HDAC1 HDAC2 HDAC3
11.1±1.7 111.2±0.2 72.5±0.5 420.9±3.6 >10000
>10000
>10000 >10000 9.6±0.2
6f
29.3±0.9 999.6±5.9 108.2±0.4 752.0±5.2 >10000
>10000
>10000 >10000 8.5±0.7
6j
3.9±0.3 16.6±1.4 29.7±0.7 398.3±2.3 >10000
>10000
>10000 >10000 7.1±1.0
6k
4.5±0.1 33.1±0.5 35.3±0.2 560.9±1.2 >10000
>10000
>10000 >10000 5.9±2.0
SAHA
63.5±1.9 214.0±0.1 72.8±1.8 3400.1±6.2 >10000
>10000
>10000 >10000 9.1±0.3
M AN U
EP
TE D
Each value is obtained by three independent experiments.
AC C
a
SC
6d
60
AC C
Figure 1
EP
TE D
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
61
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
Figure 2
62
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
Figure 3
63
M AN U
SC
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
Figure 4
64
RI PT
ACCEPTED MANUSCRIPT
AC C
EP
TE D
M AN U
SC
Figure 5
65
ACCEPTED MANUSCRIPT
SC
RI PT
(A)
AC C
EP
TE D
M AN U
(B)
Figure 6
66
ACCEPTED MANUSCRIPT H3CO H3CO
H3CO
OCH3 N
O 2O
N H
O 6
N H
OCH3
H3CO
OH
N
H N nO
H N
6
O
OH
O
n=2-4
6d
RI PT
ortho-, meta-, para-(6a-i)
O
R
N
2O
N H
O
N H n=4, 6-8 n
6j-r
AC C
EP
TE D
M AN U
SC
Scheme 1. Design of indole-containg aliphatic hydroxamates derived from compound 6d
67
OH
ACCEPTED MANUSCRIPT Scheme 2
HO
6
O
H N
a
O O
NH2
O
HO
H N
b
O
6
O
Br
Cl n
Cl
O
nO
H N
O
6
O
H N
d
O
R
O N
O
O
R
N
O
N
M AN U
O
N
Br N
O
nO
H N
6
OH
O
6a-o
O
O
H N
SC
:
e
5a-o
O R
6
O
nO
4a-o
OH
RI PT
n=2-4
1a: o-aminophenol 1b: m-aminophenol 1c: p-aminophenol
nO
O
3a-i
2a-c HO
R
c
O
6
N
N
O
AC C
EP
TE D
Reagents and conditions: (a) 1) (COCl)2, CH2Cl2, RT; 2) DMF, 60 oC, N2, 42-46%; (b) K2CO3, MeCN, ∆, N2, 79-99%; (c) substituted indole or indole, K2CO3, DMF, RT, N2, 25-95%; (d) LiOH, MeOH, 95-99%; (e) 1) ClCO2Et, Et3N, THF, RT; 2) NH2OH-HCl, KOH, MeOH, 56-99%
68
ACCEPTED MANUSCRIPT Scheme 3
O
H N
a
O
n
O
O
7a-c n=4, 7, 8
O
n O
N
Br
H N O
n O
N
n O
O
Cl
O
c
9a-c
d
O
O
N
H N
O
O
n O
OH
e
4p-r
4p-r
O
H N
O
Cl
8a-c
OH
O
b
O
RI PT
HO
H N
O
H N
OH
SC
O
n O
6p-r
AC C
EP
TE D
M AN U
Reagents and conditions: (a) 1) (COCl)2, CH2Cl2, RT; 2) 1b, DMF, 60 oC, N2, 65-87%; (b) K2CO3, MeCN, ∆, N2, 8897%; (c) 5-Methoxyindole, K2CO3, DMF, RT, N2, 80-97%; (d) LiOH, MeOH, 97-98%; (e) 1) ClCO2Et, Et3N, THF, RT; 2) NH2OH-HCl, KOH, MeOH, 79-89%
69
ACCEPTED MANUSCRIPT >Novel indole-containing aliphatic hydroxamates have been developed. >Several compounds had much greater enzyme-inhibiting than SAHA towards HDACs.
AC C
EP
TE D
M AN U
SC
RI PT
>Compounds 6j and 6k displayed potent inhibition for class I HDACs.