SlideShare ist ein Scribd-Unternehmen logo
1 von 5
Downloaden Sie, um offline zu lesen
Identification of a series of substituted 2-piperazinyl-5-pyrimidylhydroxamic
acids as potent histone deacetylase inhibitors
Patrick Angibaud a,*, Kristof Van Emelen b
, Laurence Decrane a
, Sven van Brandt b
, Peter ten Holte b
,
Isabelle Pilatte a
, Bruno Roux a
, Virginie Poncelet a
, David Speybrouck a
, Laurence Queguiner a
,
Sandrine Gaurrand a
, Ann Mariën c
, Wim Floren c
, Lut Janssen c
, Marc Verdonck b
, Jacky van Dun c
,
Jacky van Gompel d
, Ron Gilissen d
, Claire Mackie d
, Marc Du Jardin d
, Jozef Peeters d
, Marc Noppe d
,
Luc Van Hijfte a
, Eddy Freyne c
, Martin Page c
, Michel Janicot c
, Janine Arts c
a
Ortho-Biotech Oncology Research & Development, Department of Medicinal Chemistry, campus de Maigremont BP615, 27106, Val de Reuil, France
b
Johnson & Johnson Pharmaceutical Research & Development (J&JPRD), Turnhoutseweg, 30, 2340 Beerse, Belgium
c
Ortho-Biotech Oncology R&D, Turnhoutseweg, 30, 2340 Beerse, Belgium
d
J&JPRD, ADME-Tox department, Turnhoutseweg, 30, 2340 Beerse, Belgium
a r t i c l e i n f o
Article history:
Received 17 July 2009
Revised 25 October 2009
Accepted 27 October 2009
Available online 30 October 2009
Keywords:
HDAC inhibitors
Histone deacetylase
Anti-cancer agents
Petasis
Pyrimidylhydroxamic acid
a b s t r a c t
Pursuing our efforts in designing 5-pyrimidylhydroxamic acid anti-cancer agents, we have identified a
new series of potent histone deacetylase (HDAC) inhibitors. These compounds exhibit enzymatic HDAC
inhibiting properties with IC50 values in the nanomolar range and inhibit tumor cell proliferation at sim-
ilar levels. Good solubility, moderate bioavailability, and promising in vivo activity in xenograft model
made this series of compounds interesting starting points to design new potent HDAC inhibitors.
Ó 2009 Elsevier Ltd. All rights reserved.
Two enzyme families play a crucial role in controlling the acet-
ylation status of histones, thereby altering chromatin structure and
impacting transcription processes: histones acetyl transferases
(HATs) which catalyze the transfer of an acetyl group to lysine res-
idues of chromatin and histone deacetylases (HDACs) which re-
move an acetyl group from those residues, thus leading to a
changed chromatin structure impacting transcriptional activity.1,2
Inhibiting the HDAC enzymes results in increasing the acetylation
status of chromatin which has been linked to cell cycle arrest and
apoptosis as well as down regulation of genes involved in tumor
progression, invasion, and angiogenesis.3–6
In the past decade,
HDAC inhibition has emerged as an attractive target for the devel-
opment of new anti-cancer agents,7
resulting in the approval of the
HDAC inhibitor (HDACi) vorinostat for Cutaneous T-Cell Lym-
phoma (CTCL),8,9
and in the development of several other hydroxa-
mic acids such as, for example, belinostat10
or panobinostat.11
In recent years, a number of additional nonhistone HDAC sub-
strates have also been identified adding growing evidence that
HDACi exert their pleiotropic effect not only through epigenetic
control.12
Recent data have also demonstrated that HDACi could
target novel therapeutic applications such as neurodegenerative
diseases and inflammation.13–15
We have previously identified R306465 (Fig. 1) as a highly po-
tent class I selective HDACi, showing oral anti-tumor activity in hu-
man xenograft-bearing athymic nude mice.16
In order to expand and optimize this first generation HDACi, we
have explored a series of analogues on the basis that replacing the
0960-894X/$ - see front matter Ó 2009 Elsevier Ltd. All rights reserved.
doi:10.1016/j.bmcl.2009.10.118
* Corresponding author.
E-mail address: pangibau@prdfr.jnj.com (P. Angibaud).
N
N
O
N
H
HO
N
N
2
N
N
O
N
H
HO
N
N R1
R
3
N
N
O
N
H
HO
N
N
S
1
O
O
R306465
Figure 1. Johnson & Johnson first generation of HDACi: R306465 1, alkyl analogue
2, and targeted compounds 3.
Bioorganic & Medicinal Chemistry Letters 20 (2010) 294–298
Contents lists available at ScienceDirect
Bioorganic & Medicinal Chemistry Letters
journal homepage: www.elsevier.com/locate/bmcl
sulfonamide moiety would remove one key element hampering
solubility. We first postulated that substituting the SO2 moiety
by a carbon atom would restore the basicity of the nitrogen atom
and re-introduce the possibility of making salts, thus impacting
compound solubility. Indeed replacement of the sulfonyl moiety
by a CH2 in 2 provided an HDACi of similar in vitro potency (see
Table 1) and improved solubility (0.3 mg/mL at pH 4)17
as com-
pared to R306465 (<0.1 mg/mL at pH 4.8).
In addition, the piperazine secondary amine gave us the oppor-
tunity to use the Petasis multicomponent reaction18
as an option to
further introduce chemical diversity on the cap-part of this pyrim-
idyl-based hydroxamic acid series. First attempts were conducted
using 2-phenyl-ethenylboronic acid 6a in order to test the chemis-
try route as 6a is known for its good reactivity in the Petasis reac-
tion. Furthermore we postulate that the resulting double bond in 3
(Fig. 1) would mimic the first phenyl group of the naphtyl moiety
in 2. Finally, it enabled us to introduce a hydroxyl group to allow
for further chemical modifications.
Reacting the 2-(piperazin-1-yl)pyrimidylethyl ester 4,19
with
the glycolaldehyde dimer 5 and various boronic acids 6a–f in eth-
anol at room temperature provided us with esters 7a–f. Introduc-
tion of the hydroxamic acid moiety was then realized in three steps
using O-(tetrahydro-2H-pyran-2yl)-hydroxylamine as hydroxyl-
amine source. The first step encompasses the cleavage of the ester
into acids or carboxylates, then a classical coupling reaction en-
abled us to introduce the protected hydroxyl amine. This allowed
purification of the amides 8a–f by chromatography on silica gel.
This synthetic scheme was preferred from direct reaction of the
hydroxylamine with the ester as reported previously for other
HDACi,20
as, in our hands, hydroxamic acids where difficult to sep-
arate from the corresponding acids in case of incomplete reaction.
Tetrahydropyranyl (THP) protection was smoothly removed in
acidic media using trifluoroacetic acid which allowed easy isola-
tion, upon crystallization, of compounds 3a–f as trifluoroacetates.
Enantiomeric separation of 3a by liquid chromatography on chi-
ral phase proved to be difficult. We therefore chose to separate the
two enantiomers of the corresponding esters 7a1, 7a2 and to intro-
duce the hydroxamic acid moiety exactly as described above.
Alternatively, THP-protected hydroxamate 11 (Scheme 2) was
prepared as a key intermediate to rapidly test piperazine substitu-
tion. In one pot, the 2-(piperazin-1-yl)pyrimidylethyl ester 4 was
first saponified into its acid which was reacted with 9 to provide
the Fmoc-protected piperazine acid 10 with a moderate yield of
40%. THP-hydroxylamine was then introduced as in Scheme 1
and the Fmoc protection removed by reaction with piperazine in
CH2Cl2 at room temperature to provide 11. Petasis multicompo-
nent reaction could then be performed on 11, however with low
yields, and removal of the THP by trifluoroacetic acid then gave
compounds 3g–i.
All compounds were tested for their ability to inhibit histone
deacetylase activity of a mixture of HDAC enzyme obtained from
a nuclear HeLa cell extract and to inhibit ovarian carcinoma
(A2780) cell proliferation.21,22
Table 1
Enzymatic and antiproliferative potency of compounds 3a–i
Compounds R HDAC Cell proliferation A2780
IC50, nMa
± SD IC50, nMa
± SD
1 NA 6 ± 2 39 ± 17
2 NA 1.5 ± 0.2 66 ± 8
3a H 2.0 ± 0.1 25 ± 8
3b –4–Ph 0.9 ± 0.1 29b
3c –4–OCH3 0.95 ± 0.1 18 ± 1
3d –4–Cl 1.2 ± 0.2 17 ± 6
3e –3–F 1.2 ± 0.3 36 ± 14
3f –2–F 2b
<30b
3g –4–CH3 1.0 ± 0.1 18 ± 2
3h –4–CF3 2.1 ± 0.3 65 ± 18
3i –4–F 1.2 ± 0.3 32 ± 12
3a1 Hc
1.0 ± 0.1 40 ± 5
3a2 Hd
1.8 ± 0.3 30 ± 7
3i1 –4–Fc
1.05 ± 0.1 27 ± 13
3i2 –4–Fd
1.2 ± 0.1 31 ± 12
NA = not applicable.
a
Mean results from experiments performed in triplicate.
b
Single experiment.
c
(+) enantiomer.
d
(À) enantiomer.
a,b
4
EtO
O
N
N
N
NH
+
HO
O
N
N
N
N
9
Fmoc
N
H
O
N
N
N
N
H
O
c,d
N
H
O
N
N
N
N
OH
R
HO
10
11
3g: R=-4-CH3
3h: R=-4-CF3
3i: R=-4-F
O
O
O
N
O
O
e,f
3
O
Scheme 2. Reagents and conditions: (a) (i) NaOH 1 N, THF, rt, 16 h then (ii), Na2CO3,
H2O, rt, 5 h, crystallized in CH3CN, 40%; (c) H2N–O–THP, EDC, HOBT, NEt3, THF,
CH2Cl2, rt, 68%; (d) Piperidine, CH2Cl2, rt, 59%. (e) EtOH, rt, 15–29%; (f) CF3CO2H,
MeOH, rt, 64–81%.
a
4
EtO
O
N
N
N
NH
+
O
O OH
HO
+ (HO)2B
R
EtO
O
N
N
N
N
5 6a-f
OH
R
b, c
N
H
O
N
N
N
N
OH
R
OO
d
N
H
O
N
N
N
N
OH
R
HO
7a-f
8a-f
3a-f
3a: R=H
3b: R=-4-Ph
3c: R=-4-OCH3
3d: R=-4-Cl
3e: R=-3-F
3f: R=-2-F
Scheme 1. Reagents and conditions: (a) EtOH, rt, 42–64%; (b) LiOH or NaOH, THF,
H2O, rt, 89–96%; (c) H2N–O–THP, 1-ethyl-3-(30
-dimethylaminopropyl)carbodiimide
(EDC), 1-hydroxy-1,2,3-benzo-triazole (HOBT), NEt3, THF, CH2Cl2, rt, 36–90%; (d)
CF3CO2H, MeOH, rt, 72–84%.
P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298 295
Results from Table 1 above clearly show that replacement of the
naphtalene-2-sulfonyl moiety by phenylbutenol group provided
compounds of similar in vitro potency. Substitution of the phenyl
ring does not seem to have significant impact on enzymatic or
anti-proliferative potency despite introduction of electron-with-
drawing or electron-donating substituent. Since hydroxamic acids
are very efficient Zn2+
chelators, this preliminary results may indi-
cates that additional interactions generated by the phenyl ring,
probably orientated towards the solvent region of the tube-shaped
catalytic site described for a bacterial histone deacetylase by Finnin
et al.,23
are not key drivers of potency. It is also interesting to note
that stereochemistry of the carbon linked to the piperazinyl moiety
has virtually no influence on in vitro potency. Thermodynamic sol-
ubility of compound 3a (>10 mg/mL at pH 4.6 and 1.1 mg/mL at pH
7.4) was greatly increased as compared to R306465 (<0.1 mg/mL at
pH 4.8 and 7.7), thus indicating achievement of our first goal. This
property was also confirmed with compounds 3i1, 3i2, and 21, all
showing a solubility >5 mg/mL at pH 4. As introduction of the hy-
droxyl group was not disturbing interaction with the catalytic site
we were interested in further investigation structure–activity rela-
tionships (SAR) at that position and especially in testing introduc-
tion of solubility enhancing substituents. For the sake of
comparison we replaced the hydroxyl by an hydrogen (compound
3j) using reductive amination of 4 with commercially available
butanone 12, followed by introduction of the hydroxamic acid
moiety in three steps as described before (Scheme 3). Compound
3j showed a similar potency as 3a (Table 2) confirming that the hy-
droxyl moiety is not making key interactions with the catalytic
site.
Alkylation of ester 7a with methyliodide provided methoxy
intermediate 13 and further methoxy analogue 3k showing a
similar potency as 3a (Scheme 4). Bulkier aminogroups were intro-
duced (compounds 3l and 3m) by substitution of the methanesulf-
onic acid ester obtained by reaction of methanesulfonylchloride
onto 7a, followed by introduction of the hydroxamic acid moiety
on the resulting esters 14 and 15 (Scheme 4). Compounds 3l and
3m showed $10-fold drop in antiproliferative IC50 as compared
with 3a (Table 2).
Petasis multicomponent reaction is also a powerful tool to pre-
pare alpha-amino acids using glyoxalic acid and boronic acids. We
could therefore rapidly access amides 3n and 3o using the pathway
depicted in Scheme 5. However for these two compounds we also
noticed a $5-fold drop in antiproliferative activity. For compounds
3l–o, enzymatic potency in inhibiting a mixture of HDAC enzyme,
obtained from a nuclear HeLa cell extract, stayed comparable to 3a
with the exception of 3l being slightly weaker (Table 2) but still in
the nanomolar range.
As cyclic amines such as 3l and 3m or amides were not suitable,
we thought to replace the hydroxyl group of 3a by a small, proton-
able amino moiety. We therefore prepared the phtalimide precur-
sor 17 by Mitsunobu reaction on ester 7a (Scheme 6). However
reaction of 17 with hydrazine in the classical conditions did not
give us the awaited 4-phenyl-2-piperazinyl-but-3-enylamine 18
but rather the 3-phenyl-1-(piperazinylmethyl)-allylamine deriva-
tive 19. Formation of 19 might be the result of the rearrangement
of a transient aziridinium similarly as already proposed on piperi-
dines bearing beta-amino alcohol chain by Frizzle et al.24
We took
a,b,c,d
EtO
O
N
N
N
NH
+
Ph
O
N
N
N
N Ph
O
HOHN
4 12 3j
Scheme 3. Reagents and conditions: (a) 4-phenylbut-3-ene-2-one, Ti(OEt)4, 1,2-
dichloroethane, rt, 8%; (b) NaOH, EtOH, H2O, 80 °C; (c) H2N–O–THP, EDC, HOBT,
NEt3, THF, CH2Cl2, rt, 70%; (d) CF3CO2H, MeOH, rt, 44%.
Table 2
Evaluation of the importance of the hydroxyl moiety (R = H)
N
NO
NHHO
N N
R1
Ph
Compounds R1
HDAC Cell proliferation
A2780
IC50,
nMa
± SD
IC50, nMa
3j –CH3 1.7 ± 0.2 20b
3a –CH2OH 2 ± 0.1 25 ± 8
3k –CH2OCH3 1.5 ± 0.2 45 ± 11
3l –CH2–1-morpholino 11 ± 2 315 ± 154
3m –CH2–4-methylpiperazin–1yl 6.9 ± 0.4 392 ± 189
3n –C(O)-morpholine 7 ± 1 124 ± 26
3o –C(O)NHMe 3.3 ± 0.1 159 ± 11
21 NA 3.8 ± 0.7 281 ± 137
NA = not applicable.
a
See Table 1.
b
Single experiment.
b,c or
b,d
EtO2C
N
N
N
N
OH
Ph
EtO2C
N
N
N
N
OMe
Ph
a
EtO2C
N
N
N
N
N
Ph
X
14: X= O
15: X= N-CH3
N
N
N
N
Y
Ph
3k:Y=OCH3
3l:Y=
3m:Y=
O
HOHN
N O
N NCH3
e,f,g
e,f,g
137a
Scheme 4. Reagents and conditions: (a) NaH, CH3I, THF, 0 °C rt, 34%; (b) MsCl, NEt3,
CH3CN, 5 °C rt; (c) morpholine, K2CO3, CH3CN, 80 °C, 84%; (d) N-methylpiperazine,
K2CO3, CH3CN, 80 °C, 33%; (e) LiOH, THF, H2O, rt, 83%-quantitative; (f) H2N–O–THP,
EDC, HOBT, NEt3, THF, CH2Cl2, rt, 35–89%; (g) CF3CO2H, MeOH, rt, 44–57%.
aEtO
O
N
N
N
NH
+
H
O
OH
+ (HO)2B
Ph
O
EtO2C
N
N
N
N
OH
Ph
O
b or c
N
N
N
N
NR1
R2
Ph
O
N O NH
O
HOHN
3n: 3o:
4
16
d,e,f
N
R2
R1
Scheme 5. Reagents and conditions: (a) glyoxalic acid, 2-phenyl-ethenylboronic
acid, EtOH, 60 °C, 87%; (b) morpholine, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 38%; (c)
CH3NH2ÁHCl, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 57%; (d) LiOH or NaOH, THF, H2O, rt,
71–73%; (f) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 71–74%; (g) CF3CO2H,
MeOH, rt, 75–87% .
296 P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298
this opportunity to prepare hydroxamic acid 21, first protecting the
amino moiety by Fmoc and then introducing THP-protected
hydroxylamine after saponification of the ester function. Com-
pound 21 displayed similar HDAC inhibiting potency on enzyme
as 3a showing that variation of linker size could be tolerated. This
observation prompted us to examine the influence of longer dia-
mine linkers (Scheme 7 and Table 3).
Esters 25, 26, and 27 were, respectively, prepared by condensa-
tion of boc-protected 4-aminopiperidine, 4-(aminomethyl)piperi-
dine, and boc-protected-2-amino-methylmorpholine onto 2-meth-
anesulfonylpyrimidine-5-carboxylic acid ethyl ester 22.19
Then
Petasis reaction was performed onto 25–27 and hydroxamic acid
functionality was introduced as described in Scheme 1 (Scheme 7).
All ligands were found to be equipotent with regard to enzy-
matic activity whereas Linker B (4-aminopiperidine) and linker D
(2-methylamino-morpholine) were slightly less potent in the cel-
lular assay.
As demonstrated by the results from Tables 1–3, a wide range of
Petasis substitution on the 2-piperazinyl-5-pyrimidyl-hydroxamic
acid moiety gave birth to a novel series of potent in vitro HDAC
inhibitors. Compounds 3a and 3i2 showed an acceptable pharma-
cokinetic profile (see Table 4) with a bioavailability in rat of
19–22%, similar to the bioavailability of 1 (R306465).
Compound 3a, 3i1, and 3i2 were evaluated in an animal model
allowing noninvasive real-time evaluation of the response to HDAC
inhibitors using whole-body imaging.25
Human A2780 ovarian
carcinoma cells were engineered to express ZsGreen fluorescent
protein under control of the p21waf1,cip1
promoter, sub-cutaneously
grafted to athymic nude mice, and induction of fluorescence
in vivo was measured after 3 days of treatment. Results are reported
as increased fluorescence intensity as compared to R306465 tested
in same conditions and normalized to 1 (Fig. 2). This allowed rapid
comparison of compounds 3a, 3i1, and 3i2 with R306465 as regards
to their anti-tumor potency. Preliminary results indicate that the
three compounds may possess an in vivo efficacy in that model supe-
rior to R306465. Compounds 3i1 and 3i2 showed also similar in-
crease in fluorescence intensity (3–4-fold compared to R305465)
confirming that their stereochemistry had no influence. 21 was cho-
sen as a representative of compounds possessing decreased antipro-
liferative activity as compared to R306465 and showed reduced
fluorescence than R306465.
Compound 3i2, has been tested towards a broad panel of re-
combinant HDAC enzymes in vitro and compared to compound 1
(R306465) (see Table 5).
With the exception of HDAC4, 10 and 11 where 3i2 showed a
10–50-fold drop in inhibiting potency compared to R306465, the
overall profile of both compounds are very similar, even if 3i2
seems slightly less potent (1–3-fold). Compound 3i2 showed
activity towards all HDAC enzyme tested with some specificity
for class I inhibition, a class of HDACs where activity is key for
uncontrolled proliferation of cancer cells.26
Lowest in vitro potency
was observed towards HDAC4, 6, and 7.
aEtO
O
N
N
N
N
OH
Ph
+ NH
O
O
EtO
O
N
N
N
N Ph
N
O
O
N
N
NH2
Ph
but N
N
NH2
Ph
HO
O
N
N
N
N
Ph
HN
Fmoc
c,d e,f,g
N
H
O
N
N
N
N
Ph
NH2
HO
7a 17
18
19
19
20
21
b
Scheme 6. Reagents and conditions: (a) 1H-isoindole-1,3(2H)-dione, P(nBu)3, DIAD, CH2Cl2, rt, 52%; (b) NH2NH2ÁHBr, EtOH, rt, 98%; (c) LiOH, THF, H2O, rt; (d) 1[[(9H-fluoren-
9-ylmethoxy)carbonyl]-oxy]-2,5-pyrrolidinedione, THF, rt, 82%; (e) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 76%; (f) piperidine, CH2Cl2, rt, 56%; (g) CF3CO2H, MeOH, rt,
87%.
Table 3
Evaluation of several linker moieties (R = H)
Compounds Linker HDAC Cell proliferation A2780
IC50, nMa
IC50, nMa
3a A 6 ± 2 25 ± 8
28 B 7 ± 5 219b
29 C 1.5 ± 0.2 47b
30 D 8b
295b
a
See Table 1.
b
Single experiment.
N
N
EtO2C
SO2Me
HN
H
N
Boc
HN
NH2
HN
O
H
N
Boc
N
N
EtO2C
N
NH2
N
N
EtO2C
N N
N
EtO2C
N
O
NH2
NH2
a, b a, b
c
d,e,f,g
N
NO
OH HN
Linker
OH
Ph
N N N NH N
HN
N O
NH
A B C D
22
25 26 27
3a 28 29 30
23 24
Scheme 7. Reagents and conditions: (a) 23 or 24, K2CO3, CH3CN, rt, 53–75%; (b)
CF3CO2H, CH2Cl2, rt, 35–96%; (c) 4-aminomethyl-piperidine, K2CO3, CH3CN, rt, 33%;
(d) 5 + 6a, EtOH, rt, 47–73%; (e) NaOH or LiOH, THF, H2O, rt; (f) H2N–O–THP, EDC,
HOBT, NEt3, THF, CH2Cl2, rt, 4–54%; (g) CF3CO2H, MeOH, rt, 72–85%.
P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298 297
In conclusion, we have identified a series of novel 2-piperazi-
nyl-5-pyrimidyl-hydroxamic acids HDACi that are characterized
by high potency on HDAC HeLa cellular extract and cellular anti-
proliferation assay and have demonstrated improved solubility as
compared to our starting molecule 1. First in vivo evaluation of
leads 3a and 3i2 indicates superior potential as compared to
R306465. These results warrants more in depth evaluation of this
series.
References and notes
1. Johnstone, R. W. Nat. Rev. Drug Disc. 2002, 1, 287.
2. Grant, S.; Easley, C.; Kirkpatrick, P. Nat. Rev. Drug Disc. 2007, 6, 21.
3. Ten Holte, P.; van Emelen, K.; Janicot, M.; Jong, P. C.; de Bono, J. S.; Arts, J.. In
Top. Med. Chem; Bradbury, R., Ed.; Springer: Berlin, Heidelberg, 2007; Vol. 1, pp
293–331.
4. Bolden, J. E.; Peart, M. J.; Johnstone, R. W. Nat. Rev. Drug Disc. 2006, 5, 769.
5. Monneret, C. Anticancer Drugs 2007, 18, 363.
6. Paris, M.; Porcelloni, M.; Binashi, M.; Fattori, D. J. Med. Chem. 2008, 51, 1505.
7. Prince, H. M.; Bishton, M. J.; Harrison, S. J. Clin. Cancer Res. 2009, 15, 3958.
8. Marks, P. A. Oncogene 2007, 26, 1351.
9. Marks, P. A.; Breslow, R. Nat. Biotechnol. 2007, 25, 84.
10. Gimsing, P. Expert Opin. Investig. Drugs 2009, 18, 501.
11. Atadja, P. Cancer Lett. 2009, 280, 233.
12. Budillon, A.; Di Genarro, E.; Bruzzese, F.; Rocco, M.; Manzo, G.; Caraglio, M. Rec.
Patents Anticancer Drug Disc. 2007, 2, 119.
13. Kazantsev, A. G.; Thompson, L. M. Nat. Rev. Drug Disc. 2008, 7, 854.
14. Sun, Y.; Reddy, P. Curr. Enz. Inh. 2007, 3, 207.
15. Glauben, R.; Sonnenberg, E.; Zeitz, M.; Siegmund, B. Cancer Lett. 2008, 280,
154.
16. Arts, J.; Angibaud, P.; Mariën, A.; Floren, W.; Janssens, B.; King, P.; van Dun, J.;
Janssen, L.; Geerts, T.; Tuman, R. W.; Johnson, D. L.; Andries, L.; Jung, M.;
Janicot, M.; Van Emelen, K. Br. J. Cancer 2007, 97, 1344.
17. Thermodynamic solubility was determined at different pHs by accurately
weighting around 2.5 mg of compound, attempting to dissolve it in
phosphate-citrate buffers and filtering (if necessary) over a filter disc. An
aliquot of the filtrate was diluted with an appropriate solvent (0.1 N HCl/
acetonitrile, 1/1) and submitted for concentration measurement using a
generic UPLC method.
18. Petasis, N. A.; Goodman, A.; Zavialov, I. A. Tetrahedron 1997, 53, 16463.
19. Angibaud, P.; Arts, J.; Van Emelen, K.; Poncelet, V.; Pilatte, I.; Roux, B.; Van
Brandt, S.; Verdonck, M.; DeWinter, H.; Ten Holte, P.; Marien, A.; Floren, W.;
Janssens, B.; Van Dun, J.; Aerts, A.; Van Gompel, J.; Gaurrand, S.; Queguiner, L.;
Argoullon, J. M.; Van Hijfte, L.; Freyne, E.; Janicot, M. Eur. J. Med. Chem. 2005, 40,
597.
20. Mai, A.; Esposito, M.; Sbardella, G.; Massa, S. Org. Prep. Proced. Int. 2001, 33,
391.
21. Histone Deacetylase assays on a pool of HDAC enzymes obtained from a
nuclear HeLa cell extract or on HDAC isoforms were performed at Reaction
Biology Corporation (www.reactionbiology.com, Malvern PA) using the
‘Spectrum’ assay platform.
22. Van Brandt, S.F.A.; Van Emelen, K.; Angibaud, P.R.; Marconnet-Decrane, L.F.B.
WO2006/010749, 2006. Enzymatic test conditions reported page 49 and
cellular antiproliferative test conditions described pages 49–50 were slightly
modified to adapt to 384 well plates.
23. Finnin, M. S.; Donigian, J. R.; Cohen, A.; Richon, V. M.; Rifkind, R. A.; Marks, P.
A.; Breslow, R.; Pavletich, N. P. Nature 1999, 401, 188.
24. Frizzle, M. J.; Caille, S.; Marshall, T. L.; McRae, K.; Nadeau, K.; Guo, G.; Wu, S.;
Martinelli, M. J.; Moniz, G. A. Org. Process. Res. Dev. 2007, 11, 215.
25. Belien, A.; De Schepper, S.; Floren, W.; Janssens, B.; Marien, A.; King, P.; Van
Dun, J. Mol. Cancer Ther. 2006, 5, 2317.
26. Glaser, K. B.; Li, J.; Staver, M. J.; Wei, R. Q.; Albert, D. H.; Davidsen, . K. Biochem.
Biophys. Res. Commun. 2003, 310, 529.
Table 4
Rat DMPK properties of compounds 3a and 3i2
Compounds AUC0-inf
a
(ng h/mL) (IV) CLa
(L/h/kg) (IV) Vdss
a
(L/kg) (IV) T1/2 (h) 2 h to 8 ha
(IV) AUC0-inf
b
(ng h/mL) (po) F (%)
1 2654 0.8 0.6 1.5 3381 20
3a 413 5.9 5.8 2.6 316 19
3i2 521 4.6 1.6 0.8c
466 22
a
Study in male Sprague-Dawley rats dosed at 2.5 mg/kg in water at pH 4.4.
b
Study in male Sprague-Dawley rats dosed at 10 mg/kg in water at pH 4.5.
c
Half life determined from 1 h till 4 h post dose.
Figure 2. Folds increase of fluorescence for compounds 3a, 3i1 and 3i2 as compared
to R306465 (normalized at 1) following 3 days dosing po at 40 mg/kg in athymic
nude mice bearing s.c. A2780-p21waf1,cip1
ZsGreen ovarian xenograft.
Table 5
Comparison of activity towards all HDAC enzymes21
Class HDAC R306465 3i2
IC50, nMa
± SD IC50, nMa
± SD
I 1 2.5 ± 0.4 7.1 ± 05
I 2 9 ± 3 17.9 ± 2.5
I 3 9 ± 3 30 ± 3
I 8 27 ± 13 30 ± 2
IIa 4 4.6 ± 0.35 207 ± 23
IIa 5 7.1 ± 2.3 21.8 ± 2.6
IIa 7 46 ± 8 102 ± 11.5
IIa 9 28 ± 1 40.7 ± 3.2
IIb 6 66 ± 4 224 ± 33
IIb 10 4.3 ± 1.1 42.6 ± 1.5
IV 11 2.1 ± 0.6 20.7 ± 1.4
a
Mean data from experiments performed in triplicate.
298 P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298

Weitere ähnliche Inhalte

Was ist angesagt?

tboc fmoc protocol in solid phase peptide synthesis
tboc fmoc protocol in solid phase peptide synthesistboc fmoc protocol in solid phase peptide synthesis
tboc fmoc protocol in solid phase peptide synthesisSANTOSH KUMAR SAHOO
 
Synthesis and screening of some new fluorinated
Synthesis and screening of some new fluorinatedSynthesis and screening of some new fluorinated
Synthesis and screening of some new fluorinatedAdel Abdelrahim, PhD
 
ASM Poster Kristen Bennett
ASM Poster Kristen BennettASM Poster Kristen Bennett
ASM Poster Kristen BennettKristen Bennett
 
T boc fmoc protocols in peptide synthesis
T boc fmoc protocols in peptide synthesisT boc fmoc protocols in peptide synthesis
T boc fmoc protocols in peptide synthesisSANTOSH KUMAR SAHOO
 
Solution And Solid Phase Synthesis Publication
Solution  And Solid Phase Synthesis PublicationSolution  And Solid Phase Synthesis Publication
Solution And Solid Phase Synthesis Publicationadotse
 
BMCL P1Bicyclic Arg Surrogates
BMCL  P1Bicyclic Arg SurrogatesBMCL  P1Bicyclic Arg Surrogates
BMCL P1Bicyclic Arg SurrogatesJ. Edward Semple
 
solid phase synthesis Presentation by komal
solid phase synthesis Presentation by komalsolid phase synthesis Presentation by komal
solid phase synthesis Presentation by komalKomal Rajgire
 
Synthesis, spectral characterization and bioactivity studies of some S-substi...
Synthesis, spectral characterization and bioactivity studies of some S-substi...Synthesis, spectral characterization and bioactivity studies of some S-substi...
Synthesis, spectral characterization and bioactivity studies of some S-substi...Jing Zang
 
plain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLET
plain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLETplain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLET
plain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLETAndrew Apals
 
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)Shikha Popali
 
2008 molecular mechanism of enzymatic allene oxide cyclization in plants
2008 molecular mechanism of enzymatic allene oxide cyclization in plants2008 molecular mechanism of enzymatic allene oxide cyclization in plants
2008 molecular mechanism of enzymatic allene oxide cyclization in plantsAgrin Life
 
Htos Presentation
Htos PresentationHtos Presentation
Htos Presentationbenmz101
 
URECA 2013-2014_azhari zain
URECA 2013-2014_azhari zainURECA 2013-2014_azhari zain
URECA 2013-2014_azhari zainAzhari Zain
 

Was ist angesagt? (20)

tboc fmoc protocol in solid phase peptide synthesis
tboc fmoc protocol in solid phase peptide synthesistboc fmoc protocol in solid phase peptide synthesis
tboc fmoc protocol in solid phase peptide synthesis
 
Synthesis and screening of some new fluorinated
Synthesis and screening of some new fluorinatedSynthesis and screening of some new fluorinated
Synthesis and screening of some new fluorinated
 
ASM Poster Kristen Bennett
ASM Poster Kristen BennettASM Poster Kristen Bennett
ASM Poster Kristen Bennett
 
Navab hdl
Navab hdlNavab hdl
Navab hdl
 
197 reducing atherosclerosis in mice
197 reducing atherosclerosis in mice197 reducing atherosclerosis in mice
197 reducing atherosclerosis in mice
 
From pneumonia to influenza
From pneumonia to influenzaFrom pneumonia to influenza
From pneumonia to influenza
 
Navab's an oral hdl
Navab's an oral hdlNavab's an oral hdl
Navab's an oral hdl
 
Rsc advance -2014
Rsc advance -2014Rsc advance -2014
Rsc advance -2014
 
T boc fmoc protocols in peptide synthesis
T boc fmoc protocols in peptide synthesisT boc fmoc protocols in peptide synthesis
T boc fmoc protocols in peptide synthesis
 
Solution And Solid Phase Synthesis Publication
Solution  And Solid Phase Synthesis PublicationSolution  And Solid Phase Synthesis Publication
Solution And Solid Phase Synthesis Publication
 
BMCL P1Bicyclic Arg Surrogates
BMCL  P1Bicyclic Arg SurrogatesBMCL  P1Bicyclic Arg Surrogates
BMCL P1Bicyclic Arg Surrogates
 
solid phase synthesis Presentation by komal
solid phase synthesis Presentation by komalsolid phase synthesis Presentation by komal
solid phase synthesis Presentation by komal
 
Synthesis, spectral characterization and bioactivity studies of some S-substi...
Synthesis, spectral characterization and bioactivity studies of some S-substi...Synthesis, spectral characterization and bioactivity studies of some S-substi...
Synthesis, spectral characterization and bioactivity studies of some S-substi...
 
plain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLET
plain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLETplain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLET
plain 4 SYNTHESIS, PURIFICATION AND STABILITY STUDY OF ISLET
 
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
Chemistry of peptide (BPHARM,MPHARM,MSC,BSC)
 
Kate_Poster
Kate_PosterKate_Poster
Kate_Poster
 
2008 molecular mechanism of enzymatic allene oxide cyclization in plants
2008 molecular mechanism of enzymatic allene oxide cyclization in plants2008 molecular mechanism of enzymatic allene oxide cyclization in plants
2008 molecular mechanism of enzymatic allene oxide cyclization in plants
 
Htos Presentation
Htos PresentationHtos Presentation
Htos Presentation
 
GPR40 BMCL Paper
GPR40 BMCL PaperGPR40 BMCL Paper
GPR40 BMCL Paper
 
URECA 2013-2014_azhari zain
URECA 2013-2014_azhari zainURECA 2013-2014_azhari zain
URECA 2013-2014_azhari zain
 

Ähnlich wie Potent HDAC inhibitors identified from substituted 2-piperazinyl-5-pyrimidylhydroxamic acids

JMC 1996 39 4531LactamSulf
JMC 1996 39 4531LactamSulfJMC 1996 39 4531LactamSulf
JMC 1996 39 4531LactamSulfJ. Edward Semple
 
C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...
C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...
C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...Keiji Takamoto
 
BioorganicMedChem Letters
BioorganicMedChem LettersBioorganicMedChem Letters
BioorganicMedChem LettersBrian Myers
 
JOC-Liu-1989
JOC-Liu-1989JOC-Liu-1989
JOC-Liu-1989Paul Liu
 
A convenient new and efficient commercial synthetic route for dasatinib (Spry...
A convenient new and efficient commercial synthetic route for dasatinib (Spry...A convenient new and efficient commercial synthetic route for dasatinib (Spry...
A convenient new and efficient commercial synthetic route for dasatinib (Spry...Ratnakaram Venkata Nadh
 
PNAS-1998-Barawkar-11047-52
PNAS-1998-Barawkar-11047-52PNAS-1998-Barawkar-11047-52
PNAS-1998-Barawkar-11047-52Dinesh Barawkar
 
Tfa_N-methylation
Tfa_N-methylationTfa_N-methylation
Tfa_N-methylationJordan Gipe
 
A convenient synthesis of some diarylurea and thiourea derivatives as antimic...
A convenient synthesis of some diarylurea and thiourea derivatives as antimic...A convenient synthesis of some diarylurea and thiourea derivatives as antimic...
A convenient synthesis of some diarylurea and thiourea derivatives as antimic...Alexander Decker
 
N-alkylation methods, Characterization and Evaluation of antibacterial activi...
N-alkylation methods, Characterization and Evaluation of antibacterial activi...N-alkylation methods, Characterization and Evaluation of antibacterial activi...
N-alkylation methods, Characterization and Evaluation of antibacterial activi...IJERA Editor
 
Molecules 22-00357
Molecules 22-00357Molecules 22-00357
Molecules 22-00357elshimaa eid
 
SYNTHESIS OF DRUG CARRIER POLYACRYLIC
SYNTHESIS OF DRUG CARRIER POLYACRYLICSYNTHESIS OF DRUG CARRIER POLYACRYLIC
SYNTHESIS OF DRUG CARRIER POLYACRYLICTaghreed Al-Noor
 
Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...
Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...
Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...BRNSS Publication Hub
 
Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...
Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...
Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...Keiji Takamoto
 

Ähnlich wie Potent HDAC inhibitors identified from substituted 2-piperazinyl-5-pyrimidylhydroxamic acids (20)

JMC 1996 39 4531LactamSulf
JMC 1996 39 4531LactamSulfJMC 1996 39 4531LactamSulf
JMC 1996 39 4531LactamSulf
 
C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...
C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...
C-terminal Sequencing of Protein : Novel Partial Acid Hydrolysis & Analysis b...
 
BioorganicMedChem Letters
BioorganicMedChem LettersBioorganicMedChem Letters
BioorganicMedChem Letters
 
JOC-Liu-1989
JOC-Liu-1989JOC-Liu-1989
JOC-Liu-1989
 
A convenient new and efficient commercial synthetic route for dasatinib (Spry...
A convenient new and efficient commercial synthetic route for dasatinib (Spry...A convenient new and efficient commercial synthetic route for dasatinib (Spry...
A convenient new and efficient commercial synthetic route for dasatinib (Spry...
 
JOC '96 OH-Ester HPI
JOC '96 OH-Ester HPIJOC '96 OH-Ester HPI
JOC '96 OH-Ester HPI
 
paper 2 published
paper 2 publishedpaper 2 published
paper 2 published
 
PNAS-1998-Barawkar-11047-52
PNAS-1998-Barawkar-11047-52PNAS-1998-Barawkar-11047-52
PNAS-1998-Barawkar-11047-52
 
Tfa_N-methylation
Tfa_N-methylationTfa_N-methylation
Tfa_N-methylation
 
A convenient synthesis of some diarylurea and thiourea derivatives as antimic...
A convenient synthesis of some diarylurea and thiourea derivatives as antimic...A convenient synthesis of some diarylurea and thiourea derivatives as antimic...
A convenient synthesis of some diarylurea and thiourea derivatives as antimic...
 
N-alkylation methods, Characterization and Evaluation of antibacterial activi...
N-alkylation methods, Characterization and Evaluation of antibacterial activi...N-alkylation methods, Characterization and Evaluation of antibacterial activi...
N-alkylation methods, Characterization and Evaluation of antibacterial activi...
 
2004 hiv 1 inhibitory compounds from calophyllum brasiliense leaves
2004 hiv 1 inhibitory compounds from calophyllum brasiliense leaves2004 hiv 1 inhibitory compounds from calophyllum brasiliense leaves
2004 hiv 1 inhibitory compounds from calophyllum brasiliense leaves
 
Molecules 22-00357
Molecules 22-00357Molecules 22-00357
Molecules 22-00357
 
Tet. Lett. Eurystatin TS
Tet. Lett. Eurystatin TSTet. Lett. Eurystatin TS
Tet. Lett. Eurystatin TS
 
SYNTHESIS OF DRUG CARRIER POLYACRYLIC
SYNTHESIS OF DRUG CARRIER POLYACRYLICSYNTHESIS OF DRUG CARRIER POLYACRYLIC
SYNTHESIS OF DRUG CARRIER POLYACRYLIC
 
Synthesis of 2,3 o,o-dibenzyl-6-o-tosyl-l-ascorbic acid
Synthesis of 2,3 o,o-dibenzyl-6-o-tosyl-l-ascorbic acidSynthesis of 2,3 o,o-dibenzyl-6-o-tosyl-l-ascorbic acid
Synthesis of 2,3 o,o-dibenzyl-6-o-tosyl-l-ascorbic acid
 
paper 4
paper 4paper 4
paper 4
 
HCAM Org Lett 2000 2 19
HCAM Org Lett 2000 2 19HCAM Org Lett 2000 2 19
HCAM Org Lett 2000 2 19
 
Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...
Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...
Design, Synthesis, Biological Evaluation, and In Silico ADMET Studies of 1,8-...
 
Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...
Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...
Carboxy-terminal Degradation of Peptides using Perfluoroacyl Anhydrides : C-T...
 

Mehr von Peter ten Holte

Library Enhancement through the Wisdom of Crowds
Library Enhancement through the Wisdom of CrowdsLibrary Enhancement through the Wisdom of Crowds
Library Enhancement through the Wisdom of CrowdsPeter ten Holte
 
Assemblies of aziridinemethanols
Assemblies of aziridinemethanolsAssemblies of aziridinemethanols
Assemblies of aziridinemethanolsPeter ten Holte
 
Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...
Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...
Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...Peter ten Holte
 
The Synthetic Potential of Three-Membered Ring Aza-Heterocycles - Springer
The Synthetic Potential of Three-Membered Ring Aza-Heterocycles - SpringerThe Synthetic Potential of Three-Membered Ring Aza-Heterocycles - Springer
The Synthetic Potential of Three-Membered Ring Aza-Heterocycles - SpringerPeter ten Holte
 
Synthesis of Oxazolidinones
Synthesis of OxazolidinonesSynthesis of Oxazolidinones
Synthesis of OxazolidinonesPeter ten Holte
 
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...Peter ten Holte
 
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...Peter ten Holte
 

Mehr von Peter ten Holte (8)

Library Enhancement through the Wisdom of Crowds
Library Enhancement through the Wisdom of CrowdsLibrary Enhancement through the Wisdom of Crowds
Library Enhancement through the Wisdom of Crowds
 
Assemblies of aziridinemethanols
Assemblies of aziridinemethanolsAssemblies of aziridinemethanols
Assemblies of aziridinemethanols
 
Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...
Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...
Solution- and solid-phase synthesis of 4-hydroxy-4,5-dihydroisoxazole derivat...
 
The Synthetic Potential of Three-Membered Ring Aza-Heterocycles - Springer
The Synthetic Potential of Three-Membered Ring Aza-Heterocycles - SpringerThe Synthetic Potential of Three-Membered Ring Aza-Heterocycles - Springer
The Synthetic Potential of Three-Membered Ring Aza-Heterocycles - Springer
 
Synthesis of Oxazolidinones
Synthesis of OxazolidinonesSynthesis of Oxazolidinones
Synthesis of Oxazolidinones
 
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
 
Org Let PASS
Org Let PASSOrg Let PASS
Org Let PASS
 
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
Polymer-Supported N-Tritylaziridinyl(diphenyl)methanol as an Effective Cataly...
 

Potent HDAC inhibitors identified from substituted 2-piperazinyl-5-pyrimidylhydroxamic acids

  • 1. Identification of a series of substituted 2-piperazinyl-5-pyrimidylhydroxamic acids as potent histone deacetylase inhibitors Patrick Angibaud a,*, Kristof Van Emelen b , Laurence Decrane a , Sven van Brandt b , Peter ten Holte b , Isabelle Pilatte a , Bruno Roux a , Virginie Poncelet a , David Speybrouck a , Laurence Queguiner a , Sandrine Gaurrand a , Ann Mariën c , Wim Floren c , Lut Janssen c , Marc Verdonck b , Jacky van Dun c , Jacky van Gompel d , Ron Gilissen d , Claire Mackie d , Marc Du Jardin d , Jozef Peeters d , Marc Noppe d , Luc Van Hijfte a , Eddy Freyne c , Martin Page c , Michel Janicot c , Janine Arts c a Ortho-Biotech Oncology Research & Development, Department of Medicinal Chemistry, campus de Maigremont BP615, 27106, Val de Reuil, France b Johnson & Johnson Pharmaceutical Research & Development (J&JPRD), Turnhoutseweg, 30, 2340 Beerse, Belgium c Ortho-Biotech Oncology R&D, Turnhoutseweg, 30, 2340 Beerse, Belgium d J&JPRD, ADME-Tox department, Turnhoutseweg, 30, 2340 Beerse, Belgium a r t i c l e i n f o Article history: Received 17 July 2009 Revised 25 October 2009 Accepted 27 October 2009 Available online 30 October 2009 Keywords: HDAC inhibitors Histone deacetylase Anti-cancer agents Petasis Pyrimidylhydroxamic acid a b s t r a c t Pursuing our efforts in designing 5-pyrimidylhydroxamic acid anti-cancer agents, we have identified a new series of potent histone deacetylase (HDAC) inhibitors. These compounds exhibit enzymatic HDAC inhibiting properties with IC50 values in the nanomolar range and inhibit tumor cell proliferation at sim- ilar levels. Good solubility, moderate bioavailability, and promising in vivo activity in xenograft model made this series of compounds interesting starting points to design new potent HDAC inhibitors. Ó 2009 Elsevier Ltd. All rights reserved. Two enzyme families play a crucial role in controlling the acet- ylation status of histones, thereby altering chromatin structure and impacting transcription processes: histones acetyl transferases (HATs) which catalyze the transfer of an acetyl group to lysine res- idues of chromatin and histone deacetylases (HDACs) which re- move an acetyl group from those residues, thus leading to a changed chromatin structure impacting transcriptional activity.1,2 Inhibiting the HDAC enzymes results in increasing the acetylation status of chromatin which has been linked to cell cycle arrest and apoptosis as well as down regulation of genes involved in tumor progression, invasion, and angiogenesis.3–6 In the past decade, HDAC inhibition has emerged as an attractive target for the devel- opment of new anti-cancer agents,7 resulting in the approval of the HDAC inhibitor (HDACi) vorinostat for Cutaneous T-Cell Lym- phoma (CTCL),8,9 and in the development of several other hydroxa- mic acids such as, for example, belinostat10 or panobinostat.11 In recent years, a number of additional nonhistone HDAC sub- strates have also been identified adding growing evidence that HDACi exert their pleiotropic effect not only through epigenetic control.12 Recent data have also demonstrated that HDACi could target novel therapeutic applications such as neurodegenerative diseases and inflammation.13–15 We have previously identified R306465 (Fig. 1) as a highly po- tent class I selective HDACi, showing oral anti-tumor activity in hu- man xenograft-bearing athymic nude mice.16 In order to expand and optimize this first generation HDACi, we have explored a series of analogues on the basis that replacing the 0960-894X/$ - see front matter Ó 2009 Elsevier Ltd. All rights reserved. doi:10.1016/j.bmcl.2009.10.118 * Corresponding author. E-mail address: pangibau@prdfr.jnj.com (P. Angibaud). N N O N H HO N N 2 N N O N H HO N N R1 R 3 N N O N H HO N N S 1 O O R306465 Figure 1. Johnson & Johnson first generation of HDACi: R306465 1, alkyl analogue 2, and targeted compounds 3. Bioorganic & Medicinal Chemistry Letters 20 (2010) 294–298 Contents lists available at ScienceDirect Bioorganic & Medicinal Chemistry Letters journal homepage: www.elsevier.com/locate/bmcl
  • 2. sulfonamide moiety would remove one key element hampering solubility. We first postulated that substituting the SO2 moiety by a carbon atom would restore the basicity of the nitrogen atom and re-introduce the possibility of making salts, thus impacting compound solubility. Indeed replacement of the sulfonyl moiety by a CH2 in 2 provided an HDACi of similar in vitro potency (see Table 1) and improved solubility (0.3 mg/mL at pH 4)17 as com- pared to R306465 (<0.1 mg/mL at pH 4.8). In addition, the piperazine secondary amine gave us the oppor- tunity to use the Petasis multicomponent reaction18 as an option to further introduce chemical diversity on the cap-part of this pyrim- idyl-based hydroxamic acid series. First attempts were conducted using 2-phenyl-ethenylboronic acid 6a in order to test the chemis- try route as 6a is known for its good reactivity in the Petasis reac- tion. Furthermore we postulate that the resulting double bond in 3 (Fig. 1) would mimic the first phenyl group of the naphtyl moiety in 2. Finally, it enabled us to introduce a hydroxyl group to allow for further chemical modifications. Reacting the 2-(piperazin-1-yl)pyrimidylethyl ester 4,19 with the glycolaldehyde dimer 5 and various boronic acids 6a–f in eth- anol at room temperature provided us with esters 7a–f. Introduc- tion of the hydroxamic acid moiety was then realized in three steps using O-(tetrahydro-2H-pyran-2yl)-hydroxylamine as hydroxyl- amine source. The first step encompasses the cleavage of the ester into acids or carboxylates, then a classical coupling reaction en- abled us to introduce the protected hydroxyl amine. This allowed purification of the amides 8a–f by chromatography on silica gel. This synthetic scheme was preferred from direct reaction of the hydroxylamine with the ester as reported previously for other HDACi,20 as, in our hands, hydroxamic acids where difficult to sep- arate from the corresponding acids in case of incomplete reaction. Tetrahydropyranyl (THP) protection was smoothly removed in acidic media using trifluoroacetic acid which allowed easy isola- tion, upon crystallization, of compounds 3a–f as trifluoroacetates. Enantiomeric separation of 3a by liquid chromatography on chi- ral phase proved to be difficult. We therefore chose to separate the two enantiomers of the corresponding esters 7a1, 7a2 and to intro- duce the hydroxamic acid moiety exactly as described above. Alternatively, THP-protected hydroxamate 11 (Scheme 2) was prepared as a key intermediate to rapidly test piperazine substitu- tion. In one pot, the 2-(piperazin-1-yl)pyrimidylethyl ester 4 was first saponified into its acid which was reacted with 9 to provide the Fmoc-protected piperazine acid 10 with a moderate yield of 40%. THP-hydroxylamine was then introduced as in Scheme 1 and the Fmoc protection removed by reaction with piperazine in CH2Cl2 at room temperature to provide 11. Petasis multicompo- nent reaction could then be performed on 11, however with low yields, and removal of the THP by trifluoroacetic acid then gave compounds 3g–i. All compounds were tested for their ability to inhibit histone deacetylase activity of a mixture of HDAC enzyme obtained from a nuclear HeLa cell extract and to inhibit ovarian carcinoma (A2780) cell proliferation.21,22 Table 1 Enzymatic and antiproliferative potency of compounds 3a–i Compounds R HDAC Cell proliferation A2780 IC50, nMa ± SD IC50, nMa ± SD 1 NA 6 ± 2 39 ± 17 2 NA 1.5 ± 0.2 66 ± 8 3a H 2.0 ± 0.1 25 ± 8 3b –4–Ph 0.9 ± 0.1 29b 3c –4–OCH3 0.95 ± 0.1 18 ± 1 3d –4–Cl 1.2 ± 0.2 17 ± 6 3e –3–F 1.2 ± 0.3 36 ± 14 3f –2–F 2b <30b 3g –4–CH3 1.0 ± 0.1 18 ± 2 3h –4–CF3 2.1 ± 0.3 65 ± 18 3i –4–F 1.2 ± 0.3 32 ± 12 3a1 Hc 1.0 ± 0.1 40 ± 5 3a2 Hd 1.8 ± 0.3 30 ± 7 3i1 –4–Fc 1.05 ± 0.1 27 ± 13 3i2 –4–Fd 1.2 ± 0.1 31 ± 12 NA = not applicable. a Mean results from experiments performed in triplicate. b Single experiment. c (+) enantiomer. d (À) enantiomer. a,b 4 EtO O N N N NH + HO O N N N N 9 Fmoc N H O N N N N H O c,d N H O N N N N OH R HO 10 11 3g: R=-4-CH3 3h: R=-4-CF3 3i: R=-4-F O O O N O O e,f 3 O Scheme 2. Reagents and conditions: (a) (i) NaOH 1 N, THF, rt, 16 h then (ii), Na2CO3, H2O, rt, 5 h, crystallized in CH3CN, 40%; (c) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 68%; (d) Piperidine, CH2Cl2, rt, 59%. (e) EtOH, rt, 15–29%; (f) CF3CO2H, MeOH, rt, 64–81%. a 4 EtO O N N N NH + O O OH HO + (HO)2B R EtO O N N N N 5 6a-f OH R b, c N H O N N N N OH R OO d N H O N N N N OH R HO 7a-f 8a-f 3a-f 3a: R=H 3b: R=-4-Ph 3c: R=-4-OCH3 3d: R=-4-Cl 3e: R=-3-F 3f: R=-2-F Scheme 1. Reagents and conditions: (a) EtOH, rt, 42–64%; (b) LiOH or NaOH, THF, H2O, rt, 89–96%; (c) H2N–O–THP, 1-ethyl-3-(30 -dimethylaminopropyl)carbodiimide (EDC), 1-hydroxy-1,2,3-benzo-triazole (HOBT), NEt3, THF, CH2Cl2, rt, 36–90%; (d) CF3CO2H, MeOH, rt, 72–84%. P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298 295
  • 3. Results from Table 1 above clearly show that replacement of the naphtalene-2-sulfonyl moiety by phenylbutenol group provided compounds of similar in vitro potency. Substitution of the phenyl ring does not seem to have significant impact on enzymatic or anti-proliferative potency despite introduction of electron-with- drawing or electron-donating substituent. Since hydroxamic acids are very efficient Zn2+ chelators, this preliminary results may indi- cates that additional interactions generated by the phenyl ring, probably orientated towards the solvent region of the tube-shaped catalytic site described for a bacterial histone deacetylase by Finnin et al.,23 are not key drivers of potency. It is also interesting to note that stereochemistry of the carbon linked to the piperazinyl moiety has virtually no influence on in vitro potency. Thermodynamic sol- ubility of compound 3a (>10 mg/mL at pH 4.6 and 1.1 mg/mL at pH 7.4) was greatly increased as compared to R306465 (<0.1 mg/mL at pH 4.8 and 7.7), thus indicating achievement of our first goal. This property was also confirmed with compounds 3i1, 3i2, and 21, all showing a solubility >5 mg/mL at pH 4. As introduction of the hy- droxyl group was not disturbing interaction with the catalytic site we were interested in further investigation structure–activity rela- tionships (SAR) at that position and especially in testing introduc- tion of solubility enhancing substituents. For the sake of comparison we replaced the hydroxyl by an hydrogen (compound 3j) using reductive amination of 4 with commercially available butanone 12, followed by introduction of the hydroxamic acid moiety in three steps as described before (Scheme 3). Compound 3j showed a similar potency as 3a (Table 2) confirming that the hy- droxyl moiety is not making key interactions with the catalytic site. Alkylation of ester 7a with methyliodide provided methoxy intermediate 13 and further methoxy analogue 3k showing a similar potency as 3a (Scheme 4). Bulkier aminogroups were intro- duced (compounds 3l and 3m) by substitution of the methanesulf- onic acid ester obtained by reaction of methanesulfonylchloride onto 7a, followed by introduction of the hydroxamic acid moiety on the resulting esters 14 and 15 (Scheme 4). Compounds 3l and 3m showed $10-fold drop in antiproliferative IC50 as compared with 3a (Table 2). Petasis multicomponent reaction is also a powerful tool to pre- pare alpha-amino acids using glyoxalic acid and boronic acids. We could therefore rapidly access amides 3n and 3o using the pathway depicted in Scheme 5. However for these two compounds we also noticed a $5-fold drop in antiproliferative activity. For compounds 3l–o, enzymatic potency in inhibiting a mixture of HDAC enzyme, obtained from a nuclear HeLa cell extract, stayed comparable to 3a with the exception of 3l being slightly weaker (Table 2) but still in the nanomolar range. As cyclic amines such as 3l and 3m or amides were not suitable, we thought to replace the hydroxyl group of 3a by a small, proton- able amino moiety. We therefore prepared the phtalimide precur- sor 17 by Mitsunobu reaction on ester 7a (Scheme 6). However reaction of 17 with hydrazine in the classical conditions did not give us the awaited 4-phenyl-2-piperazinyl-but-3-enylamine 18 but rather the 3-phenyl-1-(piperazinylmethyl)-allylamine deriva- tive 19. Formation of 19 might be the result of the rearrangement of a transient aziridinium similarly as already proposed on piperi- dines bearing beta-amino alcohol chain by Frizzle et al.24 We took a,b,c,d EtO O N N N NH + Ph O N N N N Ph O HOHN 4 12 3j Scheme 3. Reagents and conditions: (a) 4-phenylbut-3-ene-2-one, Ti(OEt)4, 1,2- dichloroethane, rt, 8%; (b) NaOH, EtOH, H2O, 80 °C; (c) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 70%; (d) CF3CO2H, MeOH, rt, 44%. Table 2 Evaluation of the importance of the hydroxyl moiety (R = H) N NO NHHO N N R1 Ph Compounds R1 HDAC Cell proliferation A2780 IC50, nMa ± SD IC50, nMa 3j –CH3 1.7 ± 0.2 20b 3a –CH2OH 2 ± 0.1 25 ± 8 3k –CH2OCH3 1.5 ± 0.2 45 ± 11 3l –CH2–1-morpholino 11 ± 2 315 ± 154 3m –CH2–4-methylpiperazin–1yl 6.9 ± 0.4 392 ± 189 3n –C(O)-morpholine 7 ± 1 124 ± 26 3o –C(O)NHMe 3.3 ± 0.1 159 ± 11 21 NA 3.8 ± 0.7 281 ± 137 NA = not applicable. a See Table 1. b Single experiment. b,c or b,d EtO2C N N N N OH Ph EtO2C N N N N OMe Ph a EtO2C N N N N N Ph X 14: X= O 15: X= N-CH3 N N N N Y Ph 3k:Y=OCH3 3l:Y= 3m:Y= O HOHN N O N NCH3 e,f,g e,f,g 137a Scheme 4. Reagents and conditions: (a) NaH, CH3I, THF, 0 °C rt, 34%; (b) MsCl, NEt3, CH3CN, 5 °C rt; (c) morpholine, K2CO3, CH3CN, 80 °C, 84%; (d) N-methylpiperazine, K2CO3, CH3CN, 80 °C, 33%; (e) LiOH, THF, H2O, rt, 83%-quantitative; (f) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 35–89%; (g) CF3CO2H, MeOH, rt, 44–57%. aEtO O N N N NH + H O OH + (HO)2B Ph O EtO2C N N N N OH Ph O b or c N N N N NR1 R2 Ph O N O NH O HOHN 3n: 3o: 4 16 d,e,f N R2 R1 Scheme 5. Reagents and conditions: (a) glyoxalic acid, 2-phenyl-ethenylboronic acid, EtOH, 60 °C, 87%; (b) morpholine, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 38%; (c) CH3NH2ÁHCl, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 57%; (d) LiOH or NaOH, THF, H2O, rt, 71–73%; (f) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 71–74%; (g) CF3CO2H, MeOH, rt, 75–87% . 296 P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298
  • 4. this opportunity to prepare hydroxamic acid 21, first protecting the amino moiety by Fmoc and then introducing THP-protected hydroxylamine after saponification of the ester function. Com- pound 21 displayed similar HDAC inhibiting potency on enzyme as 3a showing that variation of linker size could be tolerated. This observation prompted us to examine the influence of longer dia- mine linkers (Scheme 7 and Table 3). Esters 25, 26, and 27 were, respectively, prepared by condensa- tion of boc-protected 4-aminopiperidine, 4-(aminomethyl)piperi- dine, and boc-protected-2-amino-methylmorpholine onto 2-meth- anesulfonylpyrimidine-5-carboxylic acid ethyl ester 22.19 Then Petasis reaction was performed onto 25–27 and hydroxamic acid functionality was introduced as described in Scheme 1 (Scheme 7). All ligands were found to be equipotent with regard to enzy- matic activity whereas Linker B (4-aminopiperidine) and linker D (2-methylamino-morpholine) were slightly less potent in the cel- lular assay. As demonstrated by the results from Tables 1–3, a wide range of Petasis substitution on the 2-piperazinyl-5-pyrimidyl-hydroxamic acid moiety gave birth to a novel series of potent in vitro HDAC inhibitors. Compounds 3a and 3i2 showed an acceptable pharma- cokinetic profile (see Table 4) with a bioavailability in rat of 19–22%, similar to the bioavailability of 1 (R306465). Compound 3a, 3i1, and 3i2 were evaluated in an animal model allowing noninvasive real-time evaluation of the response to HDAC inhibitors using whole-body imaging.25 Human A2780 ovarian carcinoma cells were engineered to express ZsGreen fluorescent protein under control of the p21waf1,cip1 promoter, sub-cutaneously grafted to athymic nude mice, and induction of fluorescence in vivo was measured after 3 days of treatment. Results are reported as increased fluorescence intensity as compared to R306465 tested in same conditions and normalized to 1 (Fig. 2). This allowed rapid comparison of compounds 3a, 3i1, and 3i2 with R306465 as regards to their anti-tumor potency. Preliminary results indicate that the three compounds may possess an in vivo efficacy in that model supe- rior to R306465. Compounds 3i1 and 3i2 showed also similar in- crease in fluorescence intensity (3–4-fold compared to R305465) confirming that their stereochemistry had no influence. 21 was cho- sen as a representative of compounds possessing decreased antipro- liferative activity as compared to R306465 and showed reduced fluorescence than R306465. Compound 3i2, has been tested towards a broad panel of re- combinant HDAC enzymes in vitro and compared to compound 1 (R306465) (see Table 5). With the exception of HDAC4, 10 and 11 where 3i2 showed a 10–50-fold drop in inhibiting potency compared to R306465, the overall profile of both compounds are very similar, even if 3i2 seems slightly less potent (1–3-fold). Compound 3i2 showed activity towards all HDAC enzyme tested with some specificity for class I inhibition, a class of HDACs where activity is key for uncontrolled proliferation of cancer cells.26 Lowest in vitro potency was observed towards HDAC4, 6, and 7. aEtO O N N N N OH Ph + NH O O EtO O N N N N Ph N O O N N NH2 Ph but N N NH2 Ph HO O N N N N Ph HN Fmoc c,d e,f,g N H O N N N N Ph NH2 HO 7a 17 18 19 19 20 21 b Scheme 6. Reagents and conditions: (a) 1H-isoindole-1,3(2H)-dione, P(nBu)3, DIAD, CH2Cl2, rt, 52%; (b) NH2NH2ÁHBr, EtOH, rt, 98%; (c) LiOH, THF, H2O, rt; (d) 1[[(9H-fluoren- 9-ylmethoxy)carbonyl]-oxy]-2,5-pyrrolidinedione, THF, rt, 82%; (e) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 76%; (f) piperidine, CH2Cl2, rt, 56%; (g) CF3CO2H, MeOH, rt, 87%. Table 3 Evaluation of several linker moieties (R = H) Compounds Linker HDAC Cell proliferation A2780 IC50, nMa IC50, nMa 3a A 6 ± 2 25 ± 8 28 B 7 ± 5 219b 29 C 1.5 ± 0.2 47b 30 D 8b 295b a See Table 1. b Single experiment. N N EtO2C SO2Me HN H N Boc HN NH2 HN O H N Boc N N EtO2C N NH2 N N EtO2C N N N EtO2C N O NH2 NH2 a, b a, b c d,e,f,g N NO OH HN Linker OH Ph N N N NH N HN N O NH A B C D 22 25 26 27 3a 28 29 30 23 24 Scheme 7. Reagents and conditions: (a) 23 or 24, K2CO3, CH3CN, rt, 53–75%; (b) CF3CO2H, CH2Cl2, rt, 35–96%; (c) 4-aminomethyl-piperidine, K2CO3, CH3CN, rt, 33%; (d) 5 + 6a, EtOH, rt, 47–73%; (e) NaOH or LiOH, THF, H2O, rt; (f) H2N–O–THP, EDC, HOBT, NEt3, THF, CH2Cl2, rt, 4–54%; (g) CF3CO2H, MeOH, rt, 72–85%. P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298 297
  • 5. In conclusion, we have identified a series of novel 2-piperazi- nyl-5-pyrimidyl-hydroxamic acids HDACi that are characterized by high potency on HDAC HeLa cellular extract and cellular anti- proliferation assay and have demonstrated improved solubility as compared to our starting molecule 1. First in vivo evaluation of leads 3a and 3i2 indicates superior potential as compared to R306465. These results warrants more in depth evaluation of this series. References and notes 1. Johnstone, R. W. Nat. Rev. Drug Disc. 2002, 1, 287. 2. Grant, S.; Easley, C.; Kirkpatrick, P. Nat. Rev. Drug Disc. 2007, 6, 21. 3. Ten Holte, P.; van Emelen, K.; Janicot, M.; Jong, P. C.; de Bono, J. S.; Arts, J.. In Top. Med. Chem; Bradbury, R., Ed.; Springer: Berlin, Heidelberg, 2007; Vol. 1, pp 293–331. 4. Bolden, J. E.; Peart, M. J.; Johnstone, R. W. Nat. Rev. Drug Disc. 2006, 5, 769. 5. Monneret, C. Anticancer Drugs 2007, 18, 363. 6. Paris, M.; Porcelloni, M.; Binashi, M.; Fattori, D. J. Med. Chem. 2008, 51, 1505. 7. Prince, H. M.; Bishton, M. J.; Harrison, S. J. Clin. Cancer Res. 2009, 15, 3958. 8. Marks, P. A. Oncogene 2007, 26, 1351. 9. Marks, P. A.; Breslow, R. Nat. Biotechnol. 2007, 25, 84. 10. Gimsing, P. Expert Opin. Investig. Drugs 2009, 18, 501. 11. Atadja, P. Cancer Lett. 2009, 280, 233. 12. Budillon, A.; Di Genarro, E.; Bruzzese, F.; Rocco, M.; Manzo, G.; Caraglio, M. Rec. Patents Anticancer Drug Disc. 2007, 2, 119. 13. Kazantsev, A. G.; Thompson, L. M. Nat. Rev. Drug Disc. 2008, 7, 854. 14. Sun, Y.; Reddy, P. Curr. Enz. Inh. 2007, 3, 207. 15. Glauben, R.; Sonnenberg, E.; Zeitz, M.; Siegmund, B. Cancer Lett. 2008, 280, 154. 16. Arts, J.; Angibaud, P.; Mariën, A.; Floren, W.; Janssens, B.; King, P.; van Dun, J.; Janssen, L.; Geerts, T.; Tuman, R. W.; Johnson, D. L.; Andries, L.; Jung, M.; Janicot, M.; Van Emelen, K. Br. J. Cancer 2007, 97, 1344. 17. Thermodynamic solubility was determined at different pHs by accurately weighting around 2.5 mg of compound, attempting to dissolve it in phosphate-citrate buffers and filtering (if necessary) over a filter disc. An aliquot of the filtrate was diluted with an appropriate solvent (0.1 N HCl/ acetonitrile, 1/1) and submitted for concentration measurement using a generic UPLC method. 18. Petasis, N. A.; Goodman, A.; Zavialov, I. A. Tetrahedron 1997, 53, 16463. 19. Angibaud, P.; Arts, J.; Van Emelen, K.; Poncelet, V.; Pilatte, I.; Roux, B.; Van Brandt, S.; Verdonck, M.; DeWinter, H.; Ten Holte, P.; Marien, A.; Floren, W.; Janssens, B.; Van Dun, J.; Aerts, A.; Van Gompel, J.; Gaurrand, S.; Queguiner, L.; Argoullon, J. M.; Van Hijfte, L.; Freyne, E.; Janicot, M. Eur. J. Med. Chem. 2005, 40, 597. 20. Mai, A.; Esposito, M.; Sbardella, G.; Massa, S. Org. Prep. Proced. Int. 2001, 33, 391. 21. Histone Deacetylase assays on a pool of HDAC enzymes obtained from a nuclear HeLa cell extract or on HDAC isoforms were performed at Reaction Biology Corporation (www.reactionbiology.com, Malvern PA) using the ‘Spectrum’ assay platform. 22. Van Brandt, S.F.A.; Van Emelen, K.; Angibaud, P.R.; Marconnet-Decrane, L.F.B. WO2006/010749, 2006. Enzymatic test conditions reported page 49 and cellular antiproliferative test conditions described pages 49–50 were slightly modified to adapt to 384 well plates. 23. Finnin, M. S.; Donigian, J. R.; Cohen, A.; Richon, V. M.; Rifkind, R. A.; Marks, P. A.; Breslow, R.; Pavletich, N. P. Nature 1999, 401, 188. 24. Frizzle, M. J.; Caille, S.; Marshall, T. L.; McRae, K.; Nadeau, K.; Guo, G.; Wu, S.; Martinelli, M. J.; Moniz, G. A. Org. Process. Res. Dev. 2007, 11, 215. 25. Belien, A.; De Schepper, S.; Floren, W.; Janssens, B.; Marien, A.; King, P.; Van Dun, J. Mol. Cancer Ther. 2006, 5, 2317. 26. Glaser, K. B.; Li, J.; Staver, M. J.; Wei, R. Q.; Albert, D. H.; Davidsen, . K. Biochem. Biophys. Res. Commun. 2003, 310, 529. Table 4 Rat DMPK properties of compounds 3a and 3i2 Compounds AUC0-inf a (ng h/mL) (IV) CLa (L/h/kg) (IV) Vdss a (L/kg) (IV) T1/2 (h) 2 h to 8 ha (IV) AUC0-inf b (ng h/mL) (po) F (%) 1 2654 0.8 0.6 1.5 3381 20 3a 413 5.9 5.8 2.6 316 19 3i2 521 4.6 1.6 0.8c 466 22 a Study in male Sprague-Dawley rats dosed at 2.5 mg/kg in water at pH 4.4. b Study in male Sprague-Dawley rats dosed at 10 mg/kg in water at pH 4.5. c Half life determined from 1 h till 4 h post dose. Figure 2. Folds increase of fluorescence for compounds 3a, 3i1 and 3i2 as compared to R306465 (normalized at 1) following 3 days dosing po at 40 mg/kg in athymic nude mice bearing s.c. A2780-p21waf1,cip1 ZsGreen ovarian xenograft. Table 5 Comparison of activity towards all HDAC enzymes21 Class HDAC R306465 3i2 IC50, nMa ± SD IC50, nMa ± SD I 1 2.5 ± 0.4 7.1 ± 05 I 2 9 ± 3 17.9 ± 2.5 I 3 9 ± 3 30 ± 3 I 8 27 ± 13 30 ± 2 IIa 4 4.6 ± 0.35 207 ± 23 IIa 5 7.1 ± 2.3 21.8 ± 2.6 IIa 7 46 ± 8 102 ± 11.5 IIa 9 28 ± 1 40.7 ± 3.2 IIb 6 66 ± 4 224 ± 33 IIb 10 4.3 ± 1.1 42.6 ± 1.5 IV 11 2.1 ± 0.6 20.7 ± 1.4 a Mean data from experiments performed in triplicate. 298 P. Angibaud et al. / Bioorg. Med. Chem. Lett. 20 (2010) 294–298