Bioorganic & Medicinal Chemistry

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 11

Bioorganic & Medicinal Chemistry 23 (2015) 411–421

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry


journal homepage: www.elsevier.com/locate/bmc

Investigation of fatty acid conjugates of 3,5-bisarylmethylene-


4-piperidone derivatives as antitumor agents and human
topoisomerase-IIa inhibitors
Elizabeth Potter a, Mamta Jha a, Khushwant S. Bhullar b, H. P. Vasantha Rupasinghe b, Jan Balzarini c,
Amitabh Jha a,⇑
a
Department of Chemistry, Acadia University, Wolfville, Nova Scotia B4P 2R6, Canada
b
Department of Environmental Sciences, Faculty of Agriculture, Dalhousie University, Truro, Nova Scotia B2N 5E3, Canada
c
Rega Institute for Medical Research, Katholieke Universiteit Leuven, B-3000 Leuven, Belgium

a r t i c l e i n f o a b s t r a c t

Article history: A series of five 3,5-bisarylidene-4-piperidones designed as analogs of curcumin and their twenty five fatty
Received 22 September 2014 acid conjugates were synthesized as candidate anticancer agents. The fatty acid conjugates were designed
Revised 8 December 2014 for efficient delivery of these compounds at the targeted cancer sites. The cytostatic potential of these com-
Accepted 17 December 2014
pounds was evaluated against three representative cancer cell lines namely murine leukemic L1210 cells,
Available online 26 December 2014
and human T-lymphocyte CEM cells and cervical HeLa cells. Most compounds were found to exhibit signif-
icant anti-cancer activity in vitro. QSAR studies indicated electrophilicity of these compounds towards cel-
Keywords:
lular nucleophiles may have a key role to play in their cytostatic activity. Representative compounds were
Curcumin analogs
Fatty acid amides
also tested for topoisomerase IIa inhibitory potential, which indicated strong catalytic inhibition of the
3,5-Bisarylmethylene-4-piperidones enzyme in vitro. The data showed that the fatty acid conjugates also possessed robust antioxidant activity
Michael acceptor in multiple analyses. This study also indicated that these compounds prompted significantly lower cellular
Anti-oxidant damage in human fibroblasts than a currently used cancer drug sorafenib in vitro. The wide spectrum of
Cytostatic activity anticancer action, supplemented with antioxidant potential along with non-toxic manifestations, certainly
Topoisomerase IIa inhibitors augment the anticancer candidacy of the novel fatty acid conjugates.
QSAR Ó 2014 Elsevier Ltd. All rights reserved.

1. Introduction Our research group is actively working on making analogs of


curcumin in attempts to develop molecules with improved phar-
Curcumin (Fig. 1) is a dietary phytochemical isolated from Cur- macokinetic as well as pharmacodynamic profile than those of cur-
cuma longa, commonly known as turmeric, a widely used spice in cumin.15–27 We previously prepared and studied curcumin analogs
South East Asia.1 Biological activities of this compound have been where the diarylheptanoid framework of curcumin was retained
a subject of intensive research worldwide and over the years a (series 1–3, Fig. 1).15–18 These studies involved anticancer cyto-
number of its pharmacological characteristics such as antiinflam- static, anti-melanoma, antioxidant, anti-hypertensive, and/or
matory, antitumor, antiangiogentic, antioxidant, antibacterial, car- anti-HIV properties of these compounds. Overall, several com-
dioprotective and neuroprotective properties have been pounds from series 1 and 2, which retained most of the structural
unravelled.2–6 Curcumin is being/has been subjected to a large features of curcumin were found to be most active in terms of their
number of clinical trials,7 including for the treatment of metastatic anticancer and antioxidant potency in vitro. We have also worked
breast cancer,8 colon cancer,9 advanced pancreatic cancer,10 cystic considerably on simplified diarylpentanoid analogs of curcumin,
fibrosis,11 inflammatory bowel disease,12 and Alzheimer’s dis- viz. 3,5-bisarylmethylene-4-piperidones, where the central keto-
ease.13 As expected from a commonly used dietary phytochemical, enol moiety was replaced by a ketone functionality (series 4–
curcumin displays excellent safety profile but its clinical efficacy is 10).19–24 Curcumin as well as its various series of analogs (1–10)
marred with it poor bioavailability which has kept this molecule contain enone functional group(s) and therefore are Michael
from culminating into a successful clinical drug.14 acceptors with selective affinity for cellular thiols as opposed to
nucleophiles found on genetic materials.25,28 It should be noted
that analogs from series 5–10 contain side chains bearing addi-
⇑ Corresponding author. Tel.: +1 902 585 1515; fax: +1 902 585 1414. tional enone functionality for interaction with auxiliary binding
E-mail address: ajha@acadiau.ca (A. Jha).

http://dx.doi.org/10.1016/j.bmc.2014.12.042
0968-0896/Ó 2014 Elsevier Ltd. All rights reserved.
412 E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421

O OH O OH
O O
R R
HO Curcumin OH
n
Series 1, n=0
Series 2, n=1
O O

R
R N R
H
Series 3 Series 4
O O

R N R N

O O
5 R'=H
6 E, R' = Ar R' R'
7 E, R' = CONHAr
8 Z, R' = CONHAr 10
9 Z, R' = CONH-amino acid esters O

Figure 1. Molecular structures of curcumin and its previously studied analogs.

domain20 and/or protein thiolation.25,28 The parent 3,5-bisarylm- if topoisomerase poisoning occurs, the breaks in the DNA strands
ethylene-4-piperidones 4 and there derivatives (5–10) (Fig. 1) created by the enzyme cannot ligate back resulting in lethal lesions
were consistently found to possess higher level of antiproliferative in the DNA frame work, which then leads to apoptosis.29,30 Since
activity than diarylheptanoids 1–3. Of particular relevance to this the expression of topoisomerase IIa is heightened in rapidly divid-
report are series 9 compounds where micronutrient amino acids ing cancerous cells, targeting this enzyme by protein-thiolators
were conjugated to the parent 3,5-bisarylmethylene-4-piperidones offers a crucial pathway of selectivity targeting malignant cells to
via maleamic diamide moiety to selectively target cancer cells inhibit cancer growth.
where the constant cell division leads to increased requirement In the present investigation, we aimed to design and develop
of cellular micronutrients. Most of these compounds displayed fatty acid conjugates of the established pharmacophore 3,5-bisa-
cytotoxicity in sub-micromolar inhibitory concentrations against rylmethylene-4-piperidones (Fig. 2) as therapeutic agents with
cancer cell lines tested and were identified as potent human topo- dual mode of action: (a) protein thiolation by the enone function-
isomerase IIa inhibitors.22 alities, and (b) targeted delivery at the cancer site. The conjugation
As stated, human topoisomerase IIa has been identified as a of an essential fatty acid with the 3,5-bisarylmethylene-4-piperi-
potential molecular target for 3,5-bisarylmethylene-4-piperidone done core is hypothesized to both increase site specificity and
derivatives in a previous investigation in our laboratory.22 This the ability of the compounds to penetrate the extracellular mem-
sulfhydryl-rich enzyme is essential for cell proliferation and sur- brane. This may increase the bioavailability of the drug resulting
vival.29–31 It releases torsional strain in DNA double helix through in lower clinical dose requirements. Fatty acids are known to be
transient double stranded nicks facilitating segregation of the taken up more rapidly by tumour cell, as they are vital in the syn-
strands, allowing replication and transcription processes to thesis of biochemical precursors, as well as an energy source for
occur.29,30,32–34 Inhibition of these processes brings replication rapid biosynthesis.35–37 Furthermore, fatty acids are also readily
and transcription to halt, thus impeding cell growth. Alternatively, incorporated into the lipid bilayer, thus disrupting the fluidity

Ar = R= H 1
A
O
2
O O
B
EtO2 C Ar Ar 3
O 9
N
C 4
N R 11
O

N A1-A7 5
B1, B3, B6, B7 13
D C1, C3-C7 O
D1, D3-D7 6
E1-E7 15
O
E 7
O
6 6

Figure 2. Novel amides of 3,5-bisarylmethylene-4-piperidones synthesised and investigated.


E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421 413

and structure of the cancer cell membrane.35 It is suggested that acidic41 or basic conditions42 and the products were isolated as
this results in improved chemosensitivity facilitating antitumor acid salts or free bases, respectively. The subsequent conversion
targeting. Conjugates of paclitaxel, an antitumor agent, with essen- to fatty acid amides were carried out by converting the fatty acids
tial polyunsaturated fatty acids led to the development of Taxopr- to corresponding acid chlorides and their conjugation with 3,5-bis-
exinÒ, the conjugate formed with docosahexaenoic acid.35–37 This arylidenepiperidones in presence of appropriate amount of trieth-
conjugation leads to an increase in antitumor activity, as well as ylamine (Scheme 1). All fatty acid amides were purified by column
a decrease in toxicity relative to the parent molecule, paclitaxel.35 chromatography and were conclusively characterized by spectro-
Butyric, lauric, myristic, palmitic, stearic and oleic acids were cho- scopic means. It was interesting to note that the fatty acid conju-
sen to represent fatty acids (Fig. 2). Butyric acid is a short chain gates acquired non-symmetrical conformation in the 3,5-
fatty acid known to have strong anticancer properties.38,39 bisarylidenepiperidone moiety as evident from the peaks in the
The aryl rings in the designed compounds are represented by NMR spectra. All compounds, except A143 and C1,44 studied in this
phenyl, 4-carboethoxyphenyl, 2-furyl, 2-pyridyl and 4-pyridyl investigation are new to chemical literature.
rings. The rationales behind the selection of these aryl group are
as follows: (a) In terms of substitution, the unsubstituted phenyl 2.2. Biological evaluation
ring offers the reference point; (b) since protein thiolation is being
considered as the primary mode of action, electron withdrawing 2.2.1. Cytostatic activity
groups on the phenyl ring are expected increase the electrophilic- All compounds synthesized under this investigation were eval-
ity of dienone functionality; carboethoxy group at position 4 of the uated for cytostatic activity against murine L1210 leukemia cells,
phenyl was introduced as an electron withdrawing group; (c) 2- as well as human T-lymphocyte CEM and HeLa cells, following a
furyl group, which happens to be a polar electron-rich heteroaro- literature procedure.45 The choice of cell lines was determined on
matic ring, was chosen to understand its effect on bioactivity due the basis that the murine cell lines such as L1210 have been
to reduced electrophilicity of the dienone functionality and altered claimed to be good predictors of clinical anticancer agents46 while
solubility; and (d) the selection of 2-pyridyl and 4-pyridyl aromatic activity towards the T-lymphocyte CEM and HeLa cells would indi-
substituents was based on their ability to form salts, increasing the cate a capacity to inhibit the growth of human tumours. The results
solubility of the compound and facilitating drug delivery. Also, the are presented in Table 1.
pyridyl rings are electron-deficient and therefore make the die- Some general observations with regards to the structure and the
none system more electrophilic towards protein thiolation. This cytostatic activity will be made first. As evident from Table 1, nearly
coupled with the rigidity imparted to the system by the 4-piperi- 50% of the compounds tested displayed high cytostatic activity
done ring appear to be essential in achieving desirable antitumor against three representative cancer cell lines with IC50 values in
properties.40 The compounds in Figure 2 may be viewed as pro- low micromolar or submicromolar range. This provides strong evi-
drugs of 3,5-bisarylmethylene-4-piperidone molecules but they dence that these molecules are potent anti-tumour agents as well
can also be viewed as active compounds in their own right as they as promising lead molecules for further development. Precursor
bear the pharmacophore required for the activity. Also, as these 3,5-bisarylmethylene-4-piperidones A1–D1 showed potent cyto-
compounds are structurally divergent from known human topoiso- static activity against each of the three cell lines and, on an average,
merase-IIa inhibitors currently available, their development may were found to be more potent than the reference drugs melphalan
be of value in treating drug-resistant tumours. and curcumin in vitro. In general, the majority of the fatty acid con-
jugates of series A–D were found to be significantly cytostatic with
2. Results and discussion no or small loss in magnitude of the activity with increasing chain
length. Interestingly, conjugation of stearic acid (A6–E6) consis-
2.1. Chemistry tently led to significant or complete loss of activity in each series
of molecules tested. However, conjugation with the corresponding
The synthetic strategy followed to synthesize five 3,5-bisaryl- monounsaturated oleic acid (A7–E7), led to restoration of the cyto-
idenepiperidones and their twenty five fatty acid amides is shown static activity.
in Scheme 1. Aldol condensations between 2 equiv of aromatic/ Compound E1 and its fatty acid amides (E2–E7) were found to be
heteroaromatic aldehydes with 4-piperidone hydrochloride inactive or moderately active in the cytostatic assays. Interestingly,
hydrate were carried out according to literature procedures under the precursor E1 was completely inactive (IC50 >250 lM against

O O O
AcOH 10% Aq KOH
H HCl (g) H
Ar Ar EtOH
Ar Ar
O N N N O
2 eq. H H H 2 eq.
.H 2O .HCl A1 .HCl (Ar=Phenyl) .H 2O .HCl
Ar = Phenyl B1 .AcOH (Ar=4-(CO2 Et)Ph) Ar = 2-Pyridyl
C1 .3HCl (Ar=4-Pyridyl) 2-Furyl
4-(CO2 Et)Ph
D1 (Ar=2-Pyridyl)
4-Pyridyl E1 (Ar=2-Furyl)
O
O Cl(CH 2) 2Cl, SOCl2 O A1-E1, Cl(CH 2) 2Cl
Ar Ar
R OH 0o C to reflux, 3h R Cl Et3N (2-5 eq), RT, 17 h A2-A7
N
B3, B6, B7
O R C3-C7
D3-D7
E2-E7

Scheme 1. Synthetic strategy followed to synthesize 3,5-bisarylmethylene-4-piperidone fatty acid amides in the present investigation. Please refer to Figure 2 for the
structure of various R-groups.
414 E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421

Table 1 each of the three cell lines) but five of its fatty acid conjugates (E2–
Evaluation of cytostatic potential of compounds of series A–D against human CEM T- E5, E7) were found to be moderately active (average IC50 51 lM) in
lymphocytes, HeLa and murine L1210 leukemic cells
the same assay system.
Compound IC50 (lM) SRa Poor cytostatic activity of compounds of series E was expected
CEM HeLa L1210 due to electron-rich nature of furan ring which renders the corre-
A1 0.9 ± 0.0 2.6 ± 0.7 7.0 ± 2.3 7.7
sponding bisarylmethylene-4-piperidone Michael acceptor system
A2 3.7 ± 2.3 3.9 ± 0.6 14.0 ± 7.0 3.8 much less electrophilic. This in turn adversely impacts the protein
A3 5.7 ± 0.7 2.6 ± 1.8 5.6 ± 0.2 2.2 thiolating capacity of these compounds retarding anticancer
A4 7.2 ± 3.4 4.8 ± 0.4 18.0 ± 6.0 3.8 effect by this mechanism. We have consistently observed this in
A5 140.0 ± 43.0 32.0 ± 6.0 109.0 ± 4.0 4.4
past19–25,27,28,47,48 and have confirmed the thiolation capacity of
A6 >250 >250 >250 —
A7 70.0 ± 47.0 25.0 ± 3.0 58.0 ± 25.0 2.8 the enone system.25,28 By the same token, series B–D were
B1 1.0 ± 0.7 0.8 ± 0.0 6.9 ± 0.5 8.7 expected to show superior cytostatic activity than series A (or
B3 13.0 ± 1.0 12.0 ± 1.0 3.5 ± 2.1 3.7 E). Series B–D bear electron deficient aromatic rings which make
B6 >125 >125 >125 —
the corresponding dienone systems more thiophilic. The manifes-
B7 29.0 ± 9.0 26.0 ± 2.0 29.0 ± 2.0 1.1
C1 1.7 ± 0.0 0.8 ± 0.1 8.9 ± 0.4 10.9
tation of this characteristic in the cytostatic activities of the cor-
C3 1.1 ± 0.3 2.6 ± 1.8 2.8 ± 0.7 2.5 responding analogs (barring the stearic acid conjugates, vide
C4 1.2 ± 0.0 0.9 ± 0.3 3.0 ± 0.3 3.3 supra) was indeed observed (Table 1). This effect was dramatic
C5 2.1 ± 1.0 3.0 ± 0.5 7.0 ± 1.1 3.3 in series C and D where nearly all compounds were found to pos-
C6 9.7 ± 6.3 2.2 ± 1.8 14.0 ± 9.0 6.4
sess potent cytostatic activity with low or sub-micromolar IC50
C7 8.5 ± 4.2 4.7 ± 0.2 6.6 ± 2.2 1.8
D1 1.9 ± 0.2 0.2 ± 0.0 1.0 ± 0.7 11.2 values against each of the three cell lines tested. All compounds
D3 2.5 ± 1.6 0.9 ± 0.1 2.8 ± 0.4 3.1 of series C were found to be more potent than curcumin while
D4 0.9 ± 0.1 0.7 ± 0.0 0.6 ± 0.0 1.5 most of the series D analogs (D1, D3–D5) were more potent than
D5 1.4 ± 0.6 0.7 ± 0.0 0.9 ± 0.1 2.0 both melphalan and curcumin in vitro.
D6 63.0 ± 5.6 100.0 ± 14.0 26.0 ± 6.0 3.9
D7 6.2 ± 0.6 15.0 ± 7.0 7.6 ± 5.1 2.4
Analysis of selective toxicity of these compounds towards the
E1 >500 >250 >250 — cell lines tested revealed moderate selectivity in the parent bisa-
E2 47.0 ± 40.0 95.0 ± 19.0 65.0 ± 40.0 2.0 rylmethylene-4-piperidones A1–E1 (SR range 4.7–11.2, Table 1);
E3 19.0 ± 9.0 84.0 ± 21.0 21.0 ± 2.0 4.4 the fatty acid conjugate, albeit potent, generally showed poor
E4 24.0 ± 7.0 71.0 ± 2.0 26.0 ± 10.0 3.0
selectivity (SR range 1.1–6.4, Table 1).
E5 26.0 ± 1.0 71.0 ± 35.0 34.0 ± 20.0 2.7
E6 >250 >250 >250 —
E7 53.0 ± 8.0 74.0 ± 35.0 62.0 ± 20.0 1.4 2.2.1.1. Quantitative structure–activity relationship stud-
Melphalan 2.6 ± 1.2 2.7 ± 2.4 5.8 ± 0.3 2.2 ies. In an attempt to investigate the effect of structural and
Curcumin 15.0 ± 5.0 18.0 ± 3.0 22.0 ± 1.0 1.5 electronic parameters on the cytostatic activity of the five series
The SR values in bold highlight compounds that displayed significant selective of compounds investigated, a quantitative structure–activity rela-
cytostatic activity against the tested cell lines. tionship (QSAR) study was undertaken using certain physicochem-
a
SR indicates the selectivity ratio, i.e., the ratio between the highest and lowest ical parameters of the bioactivity. The extent to which the
IC50 values of either the T-lymphocytes or murine leukemic cells.
electrophilicity of the dienone system affected the bioactivity
was evaluated by comparing the electronic charge at the b-position
Table 2 (calculated using a commercial software)49 of the symmetrical
Correlation constants between IC50 values of compounds under investigation and
enone structure with the IC50 values against the three cell lines.
their physicochemical parameters
Likewise, the calculated partition coefficient (log P) and molar
Independent Dependent Type Correlation coefficient ra p valueb refractivity (MR) values of the test compounds were also obtained
Charge b L1210 IC50 Semi-Log 0.479 0.0077 using a commercial software46 and utilized as a means of assessing
CEM IC50 Semi-Log 0.473 0.0084 the effect of the lipophilicity and steric properties of the com-
HeLa IC50 Semi-Log 0.638 0.0003 pounds with respect to their bioactivity. Linear and semilogarith-
a
The magnitude of coefficient r shows the extent (the closer the values to 1, the mic correlations were calculated using statistical analysis50 for
better) and the nature (sign positive or negative) of the correlation. the IC50 of each tumor cell line and the aforementioned physico-
b
The value of p <0.01 suggest significant correlation. chemical parameters. Only the significant correlations obtained
are included in Table 3.
Table 3 No significant correlation (p <0.05) was observed between log P
Antioxidant capacity measured by FRAP, ORAC and DPPH radical assays for 3,5-
and molar refractivities of the molecule (MR) with any of the bio-
bisarylidene-4-piperidones and curcumin
logical activities. Negative and significant correlations (p <0.05)
Compound FRAP ORAC DPPH ROS between charge b and antiproliferative activity against each of
(lmol TE/L) (lmol TE/L) (% inhibition) (% inhibition)
the three cell lines indicated that an increase in charge b led to a
A1 34.3 ± 2.2h 92.77 ± 1.9f 52.5 ± 2.6e 63.6 ± 2.5e decrease in the inhibitory concentration (desirable) on the cell
A3 91.1 ± 6.0g 288.4 ± 7.2b 83.5 ± 1.1b 84.6 ± 1.2b,c lines assayed. This was anticipated (vide supra) as the molecules
B1 160.8 ± 2.9e 67.07 ± 5.8f 68.6 ± 1.8c,d 73.9 ± 5.9d
B3 206.9 ± 18.8d 256.5 ± 2.7c 85.7 ± 1.1b 84.0 ± 1.9b,c
were expected to exert their cytostatic effect via protein thiolation.
C1 28.0 ± 2.7h 204.6 ± 16.5d 67.5 ± 1.6c,d 59.9 ± 3.6e This result also suggest that analogs with higher charge b than the
C4 127.0 ± 2.6f 170.2 ± 3.1e 83.5 ± 1.0b 86.4 ± 2.1b,c current compounds (more electrophilic) should be made and
D1 209.2 ± 9.2d 88.1 ± 5.6f 73.2 ± 1.2c 57.9 ± 1.1e tested to fine-tune the anticancer potential of these molecules.
D4 651.9 ± 9.5b 205.7 ± 4.2d 74.0 ± 6.4c 89.5 ± 0.5b
E1 38.2 ± 0.3h 245.4 ± 3.0c 7.2 ± 2.1f 61.2 ± 3.1e
E3 621.2 ± 19.3c 234.3 ± 1.1c 63.7 ± 4.2d 81.0 ± 1.5c,d 2.2.2. Topoisomerase inhibitory activity
Curcumin 1201.4 ± 0.2a 1581.9 ± 28.4a 96.8 ± 1.3a 98.7 ± 0.4a Based on the cytostatic activity displayed by the compounds
(Table 1), the precursor 3,5-bisarylidenepiperidones A1–E1 and
Antioxidant capacity measured by FRAP, ORAC, DPPH and ROS radical assays for the
test compounds 6–17 at 100 lM concentrations. Means ± standard deviation, fol- one of their potent fatty acid (dodecanoic or tetradecanoic acid)
lowed by the different letters within column is significantly different (Tukey’s analogues A3, B3, C4, D4 and E3 were selected for detailed biolog-
multiple means comparison test, p <0.05), ND: Not detected. ical activity analysis to discern possible mechanism of action.
E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421 415

Nicked Open

Linear band
Supercoiled

Figure 3. Topoisomerase IIa inhibitory activities of of 3,5-bisarylidene-4-piperidones derivatives. All the compounds were examined at a final concentration of 10 lM,
respectively, as designated. Lane K: pHOT1 DNA, supercoiled, Lane L: Marker DNA, Linear pHOT1, Lanes A1–E3: pHOT1 DNA + 4 U topoisomerase IIa + test compounds, Lane
SOR: pHOT1 DNA + 4 U topoisomerase IIa + Sorafenib.

Sorafenib, an orally bioavailable multi-target kinase inhibitor, also 50


known to inhibit topoisomerase IIa in vitro51 and in vivo,52 was a
used as the reference drug. The topoisomerase IIa inhibitory activ-

Percentage LDH Release


40
ity of these compounds (10 lM) is shown in Figure 3. From the
results of Figure 3, it is clear that the parent compounds and their 30
analogs (except E1) showed strong catalytic inhibitory action
towards topoisomerase IIa like the reference drug, sorafenib. 20
b
Decreased topoisomerase IIa activity indicates the fewer DNA b
b
strand breaks in gel, indicating potentially strong cytotoxicity in 10 cd
c
cd cd d
c
d
cancer cells. The results were in agreement with the cytostatic e

activity (IC50 values, Table 1) as compounds A1 and A3, C1 and 0


C3 exhibited similar topoisomerase IIa inhibition to their cyto- A1 A3 B1 B3 C1 C4 D1 D4 E1 E3 CUR SOR
static activity in vitro. These groups of compounds displayed sim- Compounds

ilar linear bands between nicked open DNA and supercoiled DNA,
Figure 5. In vitro toxicity assessment of of 3,5-bisarylidene-4-piperidones deriv-
indicating active catalytic inhibitory activity against topoisomer- atives using WI-38 cells (human fibroblasts) as measured by LDH release assay.
ase IIa action in vitro. The gel analysis showed that the compound Different letters (a through d) on columns designate a statistical significant
B3 displayed weaker topoisomerase IIa activity than its parent difference of p <0.05 between the assayed compounds using a Tukey’s test (n = 3),
CUR: Curcumin; SOR: Sorafenib.
molecule B1, as indicated by their band intensity. This observation
was also in agreement with IC50 values of both compounds, which
indicated weaker anti-cancer potential of compound B3 in vitro
logues, A3, B3, C4, D4 and E3 showed a more active antioxidant
(p <0.05). Compound E1, with presence of massive relaxed bands,
activity than their precursor compounds, as indicated by their
indicating weakest topoisomerase IIa activity among all com-
FRAP values (p <0.05). Especially, compound D4, with fatty acid
pounds examined in vitro. Its weak topoisomerase IIa activity
moiety, showed highest antioxidant activity among all tested
was also in agreement with its weak cytostatic activity, as its
derivatives (p <0.05). Moreover, compound E3, with 16 times
IC50 values range between 250 and 500 lM in vitro. However, its
higher antioxidant potential than its precursor molecule, closely
analog E3 exhibited potent topoisomerase IIa inhibitory activity
followed the leading compound D4 in vitro (p <0.05). Furthermore,
via catalytic inhibition of topoisomerase enzyme in vitro. Its activ-
among precursor compounds, A1, C1 and E1 exhibited the weakest
ity was comparable to analogs A1, A3, B1 and C1. Overall the
antioxidant activity, as measured by FRAP assay (p <0.05). Conse-
results were consistent with the anticancer drug, sorafenib, at least
quently, the structural modification emerged as a potent factor
in part, as potent topoisomerase IIa inhibitor.
towards change in antioxidant activity as compared to parent com-
pounds in vitro. The current study also revealed the peroxy radical
2.2.3. Antioxidant activity
scavenging activity of compounds using ORAC analysis. Interest-
The antioxidant properties of precursor 3,5-bisarylidene-4-pip-
ingly, compounds A3, B3, and D4, altered by structural modifica-
eridones and their selected fatty acid analogs an were investigated
tions exhibited better antioxidant activities than their
using multiple assays and compared with that of known antioxi-
corresponding parent compounds, as indicated by ORAC values
dant, curcumin. Among all the compounds, curcumin exhibited
(p <0.05). On the other hand, the antioxidant activity of compounds
the highest antioxidant activity in all the four antioxidants assays
of E1 and E3 had no statistical difference (p <0.05). Likewise, par-
conducted in vitro (p <0.05). As shown on Table 3, all novel ana-
ent compounds A1, B1 and D1 exhibited weakest peroxy radical
scavenging activity, possibly due to the absence any functionality
that could enable such effect. The antioxidant activity of these
120
compounds can also be associated with their ability to release elec-
a tron or hydrogen radical, resulting in stable DPPH in vitro. The
100 a a
Percentage Inhibition

DPPH assay confirmed the FRAP and ORAC based in vitro antioxi-
80 dant activity findings. The fatty acid amides, A3, B3, C4, and E3
b
exhibited relatively stronger anti-radical activity in vitro
60 b
bc (p <0.05). Among all modified compounds, A3, B3 and C4 exhibited
40 strongest activity while compound E1 was weakest anti-radical
c
d compound in vitro (p <0.05). These widespread variations in anti-
20 oxidant activities may be related to the variations in the electron
e e
f or hydrogen transfer abilities of these compounds, possibly due
0
A1 A3 B1 B3 C1 C4 D1 D4 E1 E3 KA to hydrolysis of amides resulting in in situ formation of fatty acids.
Compounds To get a more quantitative understanding for the possible effect of
these compounds on the cellular ROS, WI-38 cells cultured in 96-
Figure 4. Anti-tyrosinase activity of 3,5-bisarylidene-4-piperidones derivatives in
well plates were pre-exposed to all compounds, followed by per-
comparison to Kojic Acid (KA). Different letters (a through d) on columns designate
a statistical significant difference of p <0.05 between the assayed compounds using
oxy radical insult (AAPH induced) before measuring DCF fluores-
a Tukey’s test (n = 3), KA: Kojic acid. cence. All the modified compounds displayed higher ROS
416 E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421

1
inhibition and induced significant changes in DCF fluorescence as H NMR and 13C NMR were recorded on a Brucker AV300 spectro-
compared to parent compounds and assay controls (p <0.05). As photometer at 300 MHz and 75 MHz, respectively. Melting points
indicated in earlier assays, all analogs A3, B3, C4, D4 and E3 were recorded on a MEL-TEMP II apparatus and are uncorrected.
showed strong inhibition of cellular ROS in vitro (p <0.05). Regard- UV–Vis and IR spectra were recorded on Pharmacia Biotech Ultro-
ing the low ROS inhibitory activity, all parent compounds followed spec 4000 and Nicolet Avatar 330FT-IT spectrophotometers respec-
the earlier trend as compounds A1, C1, D1 and E1 exhibited statis- tively. HR-MS spectra were recorded at the Department of
tically weak inhibition of cellular ROS in vitro (p <0.05). Chemistry, Dalhousie University by Mr Xiao Feng.

2.2.4. Tyrosinase inhibition 4.1.1. Synthesis of (3E,5E)-3,5-diarylidenepiperidin-4-ones (A1–


Tyrosinase is a crucial enzyme involved in the initial stages of E1)
melanin biosynthesis and its overproduction is observed in mela- Method A: To the appropriate aromatic aldehyde (20 mmol),
noma cells.53 Investigations were undertaken to examine the inhib- acetic acid (125 mL) was added and the reaction mixture cooled
itory potential of the studied compounds against tyrosinase activity on ice. 4-Piperidone hydrochloride monohydrate (10 mmol) was
in vitro with kojic acid (as positive control). All of the ten compounds then added and dry HCl(g) passed briskly through the solution for
exhibited wide range of inhibitory effects against tyrosinase activity 3 h. The reaction mixture was then sealed and allowed to stir at
in vitro. The dramatic decrease in enzyme activities was observed room temperature overnight, or until the product crystallized from
with the structural modification of parent compounds increasing. solution. The resultant precipitate was filtered and washed with
All tested analogs (A3, B3, C4, D4 and E3) showed strong inhibition ice-cold methanol.
of tyrosinase in vitro (p <0.05) compared to parent molecules (A1–
E1). Among these, E3 and D4 displayed very high inhibition of tyros- 4.1.1.1. (3E,5E)-3,5-Dibenzylidenepiperidin-4-one hydrochlo-
inase while C1 was the weakest tyrosinase inhibitor (p <0.05). For ride (A1). Yellow powder; yield: 67%; mp 174–176 °C (lit.43
1
the parent molecules the tyrosinase inhibitory effects followed the 177–178 °C). H NMR (300 MHz; DMSO-d6): d 2.79 (s, 1H, NH),
sequence: C1 < A1 < B1 < E1 < D1 and the inhibitory effects were 4.01 (s, 4H, NCH2), 7.45–7.49 (m, 10H, ArH), 7.60 (s, 2H, ArH).
amplified with the structural modification as fatty acid conjugates. The spectral data were found to be in agreement with those
reported.43
2.2.5. In vitro-toxicity analysis
The in vitro cytotoxicity of the selected novel analogues was 4.1.1.2. Diethyl 4,40 -(1E,10 E)-(4-oxopiperidine-3,5-diylidene)bis
measured using normal fibroblasts cell line, WI-38. Toxicity was (methan-1-yl-1-ylidene)dibenzoate acetic acid salt (B1). Yellow
evaluated by LDH release assay with sorafenib and curcumin as powder; yield: 78%; mp 230–232 °C. 1H NMR (300 MHz; DMSO-d6): d
the positive controls, after exposure of cells to the tested com- 1.35 (t, J = 4.2 Hz, 6H, 2  CH3), 1.92 (s, 3H, CH3CO), 4.36 (q, 4H,
pounds for 24 h. As shown in Figure 5, these compounds exhibited J = 4.2 Hz, 2  OCH2), 4.50 (s, 4H, 2  NCH2), 7.68 (d, J = 5.1 Hz, 2H,
low toxic effects towards normal cells and were significantly safer ArH), 7.92 (s, 2H, 2  vinylic-H), 8.08 (d, J = 5.1 Hz, 2H, ArH), 10.03
as compared to sorafenib in vitro (p <0.05). Most of compounds (br s, 2H, N+H2). 13C NMR (75 MHz; DMSO-d6): d 14.09, 21.01,
exhibited <10% LDH release as compared to vehicle and curcumin 43.75, 61.00, 129.42, 129.67, 130.57, 130.62, 137.88, 138.09, 165.13,
emerged as safest compound in vitro (p <0.05). The membrane 171.91, 182.24. ESI-MS (amu): calcd C25H26NO5 [M+H]: 420.1811;
damage due to curcumin was 8 times lower than sorafenib found [M+H]: 420.1824.
in vitro (p <0.05). Among the newly synthesized derivatives, com-
pound A1, B3 and E3 exhibited highest membrane damage while 4.1.1.3. (3E,5E)-3,5-Bis(pyridin-4-ylmethylene)piperidin-4-one
C4 and D4 emerged as least cytotoxic towards normal cells trihydrochloride (C1). Yellow powder; yield: 80%; mp 202–
in vitro (p 6 0.05). 204 °C. 1H NMR (300 MHz; DMSO-d6): d 4.00 (s, 4H, 2  NCH2),
7.44 (d, J = 5.1 Hz, 4H, ArH), 7.51 (s, 2H, 2  vinylic-H), 8.65 (d,
3. Conclusion J = 5.1 Hz, 4H, ArH). 13C NMR (75 MHz; CDCl3): d 48.18, 124.10,
124.43, 133.72, 138.22, 142.64, 150.58, 150.84, 187.35. ESI-HRMS
In conclusion, 25 novel derivatives conjugating fatty acids of (amu): calcd C17H15N3O [M+]: 277.1215; found [M+]: 277.1221.
varying chain lengths to 3,5-bisarylmethylene-4-piperidones
through the amide bond were synthesised and their preliminary Method B: To an ice-cold ethanolic solution of KOH (10 g in
bioevaluations were performed. C-4, C-12, C-14 and C-16 fatty acid 30 mL), the appropriate aromatic aldehyde (20 mmol) was added.
amides of highly electron deficient pyridine ring-containing 3,5- While maintaining a reaction temperature of approximately 0 °C,
bisarylmethylene-4-piperidones were found to possess significant 4-piperidone hydrochloride monohydrate (10 mmol) was added
cytostatic activity. The compounds A3, C3, D4 and E3 emerged as slowly in portions over 5 min. The reaction mixture was allowed
potent catalytic inhibitors of topoisomerase IIa in vitro. The data to stir for 4 h at room temperature and then cooled on ice. The pre-
also indicated that most analogues exhibited potent antioxidants cipitated product was filtered and washed with ice-cold ethanol
and exhibited tyrosinase inhibitory activities, indicating anti-mel- yielding the product as a yellow or orange powder.
anoma potential. Furthermore, the results indicated that all the
analogs were less toxic towards human lung fibroblasts as com- 4.1.1.4. (3E,5E)-3,5-Bis(pyridin-2-ylmethylene)piperidin-4-one
pared to a widely used anticancer drug, sorafenib. Overall these (D1). Yellow powder; yield: 77%; mp 193–194 °C. 1H NMR
results show that these compounds were effective non-toxic anti- (300 MHz; DMSO-d6): d 4.32 (s, 4H, 2  NCH2), 7.16 (br s, 2H,
cancer agents with strong antioxidant ability, which may act as ArH), 7.35 (br s, 2H, ArH), 7.66 (d, J = 7.7 Hz, 2H, ArH), 7.84 (br
auxiliary therapy in cancer. s, 2H, 2  vinylic-H), 8.70 (br s, 2H, ArH). 13C NMR (75 MHz;
DMSO-d6): d 49.00, 123.97, 128.45, 132.01, 137.72, 140.20,
4. Experimental
150.50, 155.19, 189.91. ESI-HRMS (amu): calcd C17H15N3O
4.1. Chemistry [M+H]: 278.1293; found [M+H]: 278.1287.

All reagents were purchased from Aldrich Chemical Co and 4.1.1.5. (3E,5E)-3,5-Bis(furan-2-ylmethylene)piperidin-4-one
were used without further purification. Precoated fluorescent silica (E1). Orange powder; yield: 97%; mp 227–228 °C. 1H NMR
gel TLC plates were used to monitor the progress of the reactions. (300 MHz; CDCl3): d 4.30 (s, 4H, 2  NCH2), 6.55 (br s, 2H,
E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421 417

ArH), 6.68 (d, J = 3.30 Hz, 2H, ArH), 7.52 (s, 2H, 2  vinylic-H), (300 MHz; CDCl3): d 0.92 (t, J = 6.2 Hz, 3H, CH3), 1.06–1.48 (m,
7.69 (br s, 2H, ArH). 13C NMR (75 MHz; CDCl3): d 44.94, 47.47, 26H, 13  CH2), 2.14 (t, J = 7.5 Hz, 2H, COCH2), 4.72 and 4.95 (s,
113.87, 118.46, 121.30, 132.04, 147.17, 151.94, 152.15, 186.56. 2H each, 2  NCH2), 7.40–7.51 (m, 10H, Ar-H), 7.86 and 7.90 (s,
ESI-HRMS (amu): calcd C15H13NO3 [M+H]: 256.0974; found 1 h each, 2H, 2  vinylic-H). 13C NMR (75 MHz; CDCl3): d 14.10,
[M+H]: 256.0949. 22.68, 25.12, 29.19, 29.29, 29.35, 29.43, 29.56, 29.65, 29.68,
31.92, 33.15, 43.59, 46.26, 128.81, 129.55, 130.07, 130.64, 132.00,
4.1.2. General procedure for the synthesis of (3E,5E)-N-acyl-3,5- 134.63, 137.09, 138.49, 172.07, 186.83. IR (KBr; mmax): 2953,
diarylidenepiperidin-4-ones 2917, 2848, 1666, 1635, 1612, 1578, 1470, 1457, 1439, 1269,
To an ice-cold mixture of the appropriate fatty acid (1 mmol) in 1096, 985, 938, 769, 691 cm1. UV (CHCl3; kmax): 330 nm. ESI-
dry DCE (10 mL), SOCl2 (2.4 mmol) was added dropwise maintain- HRMS (amu): calcd C35H47NO2 [M+H]: 514.3685; found [M+H]:
ing a temperature of 5 °C. The reaction mixture was then brought 514.3641.
to room temperature and then gradually heated to the reflux tem-
perature of 85 °C and maintained there for 3 h. The remaining 4.1.2.5. (3E,5E)-3,5-Dibenzylidene-1-stearoylpiperidin-4-one
SOCl2 and DCE were removed by rotary evaporation, yielding the (A6). Waxy yellow powder; yield: 56%; mp 71–73 °C. 1H
acid chloride. The prepared acid chloride was dissolved in dry NMR (300 MHz; CDCl3): d 0.90 (t, J = 6.6 Hz, 3H, CH3), 1.06–
DCE (5 mL) and added dropwise to a mixture of the appropriate 1.49 (m, 30H, 15  CH2), 2.14 (t, J = 5.0 Hz, 2H, COCH2), 4.73
3,5-bisarylmethylene-4-pipridone (A1–E1, 1 mmol) and triethyl- and 4.96 (s, 2H each, 2  NCH2), 7.42–7.59 (m, 10H, ArH), 7.86
amine (3 mmol for A1 and B1, 5 mmol for C1 and 2 mmol for D1 and 7.90 (s, 1H each, 2H, 2  vinylic-H). 13C NMR (75 MHz;
and E1) in dry DCE. The reaction mixture was sealed and allowed CDCl3): d 14.13, 22.70, 25.12, 25.62, 29.21, 29.30, 29.37, 29.45,
to stir at room temperature overnight. An aqueous extraction 29.58, 29.67, 29.71, 31.93, 33.19, 43.57, 46.23, 128.10, 128.80,
was then carried out to remove triethylamine HCl. The crude com- 129.57, 130.05, 130.68, 134.53, 137.12, 138.52, 172.04, 186.91.
pound was then purified via column chromatography (12.5–25% IR (KBr; mmax): 2949, 2917, 1848, 1635, 1612, 1580, 1470,
ethyl acetate/hexanes) and dried on vacuum. 1456, 1438, 1269, 1169, 1095, 985, 768, 690 cm1. UV (CHCl3;
kmax): 330 nm. ESI-HRMS (amu): calcd C37H51NO 2 [M+Na]:
4.1.2.1. (3E,5E)-3,5-Dibenzylidene-1-butyrylpiperidin-4-one 564.3817; found [M+Na]: 564.3758.
(A2). Yellow powder; yield: 60%; mp 116–119 °C. 1H NMR
(300 MHz; CDCl3): d 0.77 (t, J = 7.4 Hz, 3H, CH3), 1.46–1.55 (m, 4.1.2.6. (3E,5E)-3,5-Dibenzylidene-1-oleoylpiperidin-4-one
2H, CH2), 2.12 (t, J = 7.5 Hz, 2H, COCH2), 4.73 and 4.96 (s, 2H (A7). Reddish-orange low melting solid; yield: 56%; 1 H
each, 2  NCH2), 7.47 (m, 10H, ArH), 7.89 (br s, 2H, 2  vinylic- NMR (300 MHz; CDCl 3): d 0.89–0.95 (dist. t, 3H, CH 3), 1.00–
H). 13C NMR (75 MHz; CDCl3): d 13.74, 18.49, 34.92, 43.55, 1.48 (m, 22H, 11  CH2 ), 2.01–2.14 (m, 6H, 3  CH 2), 4.72 and
46.23, 128.83, 129.59, 130.07, 130.68, 131.87, 132.03, 134.61, 4.95 (s, 2H each, 2  NCH2 ), 5.36 (br s, 2H, 2  Z-vinylic-H),
137.18, 138.49, 171.90, 186.88. IR (KBr; mmax): 2953, 2912, 7.43–7.47 (m, 10H, ArH), 7.85 and 7.90 (s, 1H each,
2835, 1655, 1603, 1578, 1445, 1427, 1174, 984, 772, 694 cm1. 2  vinylic-H). 13 C NMR (75 MHz; CDCl 3): d 14.13, 22.69,
UV (CHCl3; kmax): 330 nm. ESI-HRMS (amu): calcd C 23H23NO2 25.10, 27.19, 27.24, 28.99, 29.11, 29.10, 29.26, 29.33, 29.45,
[M+Na]: 368.1626; found [M+Na]: 368.1593. 29.53, 29.68, 29.78, 31.79, 31.91, 32.62, 33.16, 43.57, 46.25,
128.83, 129.58, 129.75, 129.98, 130.07, 130.69, 131.86,
4.1.2.2. (3E,5E)-3,5-Dibenzylidene-1-dodecanoylpiperidin-4-one 132.07, 134.57, 134.68, 137.10, 138.51, 172.01, 186.86. IR
(A3). Yellow powder; yield: 46%; mp 66–68 °C. 1H NMR (KBr; m max ): 3002, 2921, 2852, 1636, 1612, 1579, 1446, 1272,
(300 MHz; CDCl3): d 0.90 (t, J = 6.6 Hz, 3H, CH3), 1.10–1.28 (m, 1170, 985, 769, 692 cm 1 . UV (CHCl 3; kmax): 332 nm. ESI-HRMS
16H, CH2), 1.44 (m, 2H, CH2), 2.14 (t, J = 7.6 Hz, 2H, COCH2), 4.72 (amu): calcd C37 H49 NO 2 [M+Na]: 562.3661; found [M+Na]:
and 4.95 (s, 2H each, 2  NCH2), 7.40–7.60 (m, 10H, ArH), 7.85 562.3609.
and 7.90 (s, 1H each, 2  vinylic-H). 13C NMR (75 MHz; CDCl3): d
14.09, 22.67, 25.11, 29.18, 29.28, 29.31, 29.42, 29.55, 29.58, 4.1.2.7. Diethyl 4,40 -(1E,10 E)-(1-dodecanoyl-4-oxopiperidine-3,5-
31.89, 33.17, 43.58, 46.26, 127.02, 127.98, 128.81, 129.09, 129.30, diylidene)bis(methan-1-yl-1-ylidene)dibenzoate (B3). Yellow
129.56, 129.92, 130.04, 130.61, 131.88, 134.54, 137.10, 138.49, powder; yield: 55%; mp 88–91 °C. 1H NMR (300 MHz; CDCl3): d
172.07, 186.89. IR (KBr; mmax): 2952, 1919, 1848, 1675, 1637, 0.89 (t, J = 6.2 Hz, 3H, CH3); 1.00–1.70 (m, 24H, 11  CH2;
1613, 1579, 1457, 1446, 1273, 1168, 984, 937, 769, 690 cm1. UV 2  CH3), 2.33 (t, J = 7.4 Hz, 2H, COCH2), 4.31 (dist. q, J = 6.6 Hz,
(CHCl3; kmax): 330 nm. ESI-HRMS (amu): calcd C31H39NO2 [M+H]: 4H, 2  OCH2), 4.73 and 4.94 (s, 2H each, 2  NCH2), 7.46 and
458.3059; found [M+H]: 458.3011. 7.56 (d, 2H each, J = 7.2 Hz, ArH), 7.86 and 7.91 (s, 2H,
2  vinylic-H), 8.11–8.14 (m, 4H, ArH). 13C NMR (75 MHz; CDCl3):
4.1.2.3. (3E,5E)-3,5-Dibenzylidene-1-tetradecanoylpiperidin-4- d 14.12, 14.32, 22.68, 24.75, 25.04, 29.09, 29.23, 29.26, 29.31, 29.41,
one (A4). Yellow powder; yield: 19%; mp 68–69 °C. 1H NMR 29.54, 29.59, 31.91, 33.05, 33.69, 43.47, 46.30, 61.26, 61.36, 129.47,
(300 MHz; CDCl3): d 0.91 (t, J = 6.2 Hz, 3H, CH3), 1.07–1.50 (m, 129.90, 130.35, 131.06, 131.24, 133.29, 136.20, 137.47, 138.63,
22H,11  CH2), 2.14 (t, J = 7.5 Hz, 2H, COCH2), 4.73 and 4.95 (s, 165.81, 165.94, 172.02. IR (KBr; mmax): 2955, 2925, 2851, 1715,
2H each, 2  NCH2), 7.40–7.51 (m, 10H, ArH), 7.85 and 7.90 (s, 1672, 1645, 1605, 1581, 1562, 1467, 1438, 1412, 1288, 1258,
1H each, 2  vinylic-H). 13C NMR (75 MHz; CDCl3): d 14.14, 1183, 1172, 1105, 1020, 990, 862, 774 cm1. UV (CHCl3; kmax):
22.70, 25.12, 29.21, 29.30, 29.37, 29.45, 29.5, 29.65, 29.69, 31.93, 334 nm. ESI-HRMS (amu): calcd C37H47NO6 [M+Na]: 624.3301;
33.19, 43.56, 46.24, 128.79, 129.58, 130.07, 130.69, 131.86, found [M+Na]: 624.3257.
132.06, 134.56, 137.12, 138.52, 172.04, 186.90. IR (KBr; mmax):
2956, 2918, 1847, 1674, 1634, 1611, 1578, 1469, 1456, 1438, 4.1.2.8. Diethyl 4,40 -(1E,10 E)-(4-oxo-1-stearoylpiperidine-3,
1276, 1266, 1169, 984, 937, 768, 690 cm1. UV (CHCl3; kmax): 5-diylidene)bis(methan-1-yl-1-ylidene)dibenzoate (B6). Yellow
333 nm. ESI-HRMS (amu): calcd C33H43NO2 [M+H]: 486.3372; powder; yield: 87%; mp 76–78 °C. 1H NMR (300 MHz; CDCl3): d
found [M+H]: 486.3323. 0.89 (t, J = 6.1 Hz, 3H, CH3); 1.05–1.48 (m, 30H, 13  CH2; 2  CH3),
2.12 (t, J = 7.3 Hz, 2H, COCH2), 4.43 (dist. q, J = 6.2 Hz, 4H, 2  OCH2),
4.1.2.4. (3E,5E)-3,5-Dibenzylidene-1-palmitoylpiperidin-4-one 4.73 and 4.94 (s, 2H each, 2  NCH2), 7.47–7.57 (m, 4H, ArH),
(A5). Yellow powder; yield: 57%; mp 76–78 °C. 1H NMR 7.85 and 7.91 (s, 1H each, 2  vinylic-H), 8.10–8.20 (m, 4H, ArH).
418 E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421

13
C NMR (75 MHz; CDCl3): d 14.13, 14.32, 22.69, 25.03, 29.24, 29.27, ESI-HRMS (amu): calcd C33H45N3O2 [M+H]: 516.3590; found
29.36, 29.42, 29.56, 29.66, 29.70, 31.92, 33.04, 43.46, 46.30, 61.25, [M+H]: 516.3539.
129.90, 130.34, 131.23, 133.31, 136.17, 137.44, 138.63, 165.80,
171.96, 186.39. IR (KBr; mmax): 2979, 2922, 2851, 1708, 1674, 1629, 4.1.2.13. (3E,5E)-3,5-Bis(pyridin-4-ylmethylene)-1-stearoylpiperi-
1610, 1577, 1453, 1414, 1306, 1285, 1228, 1185, 1185, 1109, 1023, din-4-one (C6). Yellow powder; yield: 90%; mp 88–90 °C. 1H
943, 857, 771 cm1. UV (CHCl3; kmax): 336 nm. ESI-HRMS (amu): NMR (300 MHz; CDCl3): d 0.90 (t, J = 6.4 Hz, 3H, CH3); 1.14–.48 (m,
calcd C43H59NO6 [M+Na]: 708.4240; found [M+Na]: 708.4198. 30H, 15  CH2), 2.15 (t, J = 7.5 Hz, 2H, COCH2), 4.72 and 4.90 (s, 2H
each, 2  NCH2), 7.28–7.38 (m, 4H, ArH), 7.74 and 7.79 (s, 1H each,
4.1.2.9. Diethyl 4,40 -(1E,10 E)-(1-oleoyl-4-oxopiperidine-3,5-diy- 2  vinylic-H), 8.74–8.78 (m, 4H, ArH). 13C NMR (75 MHz; CDCl3): d
lidene)bis(methan-1-yl-1-ylidene)dibenzoate (B7). Yellow 14.13, 22.70, 24.96, 29.28, 29.37, 29.42, 29.48, 29.58, 29.70, 29.92,
low melting solid; yield: 55%; 1H NMR (300 MHz; CDCl3): d 0.89 31.93, 32.96, 33.97, 43.14, 46.22, 123.67, 123.94, 134.75, 141.75,
(t, J = 6.5 Hz, 3H, CH3); 1.10–1.27 (m, 22H, 11  CH2), 1.44 (t, 150.35, 150.35, 171.95, 185.78. IR (KBr; mmax): 3035, 3023, 2916,
J = 6.9 Hz, 6H, 2  CH3), 2.00 (m, 2H, CH2), 2.13 (t, J = 7.5 Hz, 2H, 2848, 1723, 1679, 1637, 1619, 1593, 1545, 1469, 1416, 1274, 1234,
COCH2), 4.43 (dist. q, J = 6.8 Hz, 4H, 2  OCH2), 4.73 and 4.94 (s, 1218, 1167, 996, 929, 840, 824 cm1. UV (CHCl3; kmax): 325 nm.
2H each, NCH2), 5.30–5.36 (m, 2H, 2  Z-vinylic-H), 7.46 and 7.56 ESI-HRMS (amu): calcd C35H49N3O2 [M+H]: 544.3903; found
(d, 2H each, J = 7.2 Hz, ArH), 7.86 and 7.91 (s, 2H, 2  vinylic-H), [M+H]: 544.3845.
8.11–8.18 (m, 4H, ArH). 13C NMR (75 MHz; CDCl3): d 14.12,
14.32, 22.68, 25.01, 27.15, 27.22, 28.98, 29.06, 29.15, 29.24, 4.1.2.14. (3E,5E)-1-Oleoyl-3,5-bis(pyridin-4-ylmethylene)piperi-
29.32, 29.52, 29.64, 29.76, 31.78, 31.90, 33.02, 43.47, 46.30, din-4-one (C7). Yellow low melting solid; yield: 36%; 1H NMR
61.35, 129.70, 129.91, 129.99, 130.34, 131.21, 133.29, 136.19, (300 MHz; CDCl3): d 0.89 (t, J = 6.1 Hz, 3H, CH3); 1.10–1.49 (m,
137.46, 138.69, 165.92, 171.94, 186.37. IR (KBr; mmax): 2923, 18H, 9  CH2), 2.00 (t, J = 5.3 Hz, 4H, 2  CH2), 2.14 (t, J = 7.5 Hz,
2851, 1710, 1626, 1610, 1577, 1456, 1414, 1366, 1305, 1284, 2H, COCH2), 4.71 and 4.90 (s, 2H each, 2  NCH2), 5.35 (m, 2H,
1185, 1108, 1021, 857, 771 cm1. UV (CHCl3; kmax): 335 nm. ESI- 2  Z-vinylic-H), 7.20–7.35 (m, 4H, ArH), 7.73 and 7.79 (s, 1H each,
HRMS (amu): calcd C43H57NO6 [M+Na]: 706.4084; found [M+Na]: 2  vinylic-H), 8.75 (d, J = 9.3 Hz, 4H, ArH). 13C NMR (75 MHz;
706.4013. CDCl3): d 14.13, 22.69, 24.93, 27.15, 27.22, 29.05, 29.20, 29.32,
29.52, 29.66, 29.76, 31.78, 31.90, 32.93, 43.13, 46.21, 123.63,
4.1.2.10. (3E,5E)-1-Dodecanoyl-3,5-bis(pyridin-4-ylmethylene) 123.93, 134.69, 134.83, 135.75, 141.63, 150.55, 171.88, 185.79. IR
piperidin-4-one (C3). Yellow powder; yield: 89%; mp 66– (KBr; mmax): 2923, 2852, 1680, 1634, 1619, 1592, 1544, 1457,
70 °C. 1H NMR (300 MHz; CDCl3): d 0.87 (br s, 3H, CH3); 1.12– 1429, 1416, 1259, 1232, 1167, 996, 929, 820 cm1. UV (CHCl3;
1.66 (m, 18H, 9  CH2), 2.13 (t, J = 6.9 Hz, 2H, COCH2), 4.70 and kmax): 302 nm. ESI-HRMS (amu): calcd C35H47N3O2 [M+H]:
4.88 (s, 2H each, 2  NCH2), 7.26–7.36 (m, 4H, ArH), 7.70 and 542.3747; found [M+H]: 542.3703.
7.76 (s, 1H each, 2  vinylic-H), 8.70–8.75 (m, 4H, ArH). 13C
NMR (75 MHz; CDCl3): d 14.10, 22.66, 24.93, 25.00, 29.21, 4.1.2.15. (3E,5E)-1-Dodecanoyl-3,5-bis(pyridin-2-ylmethylene)
29.24, 29.29, 29.32, 29.37, 29.47, 29.53, 29.60, 31.88, 32.91, piperidin-4-one (D3). Yellow powder; yield: 35%; mp 102–
34.30, 43.10, 46.22, 123.76, 124.03, 134.56, 134.95, 135.54, 104 °C. 1H NMR (300 MHz; CDCl3): d 0.90 (br s, 3H, CH3); 1.21–
141.97, 150.01, 150.16, 171.97, 177.19, 185.68. IR (KBr; mmax): 1.25 (m, 16H, 8  CH2), 1.58 (br s, 2H, CH2), 2.39 (br s, 2H, COCH2),
3019, 2920, 2849, 1723, 1678, 1639, 1619, 1593, 1465, 1417, 5.32 and 5.39 (s, 2H each, 2  NCH2), 7.29–7.77 (m, 8H, ArH,
1234, 1216, 1189, 1168, 995, 848, 824 cm1. UV (CHCl3; kmax): 2  vinylic-H), 8.76 (s, 2H, ArH). 13C NMR (75 MHz; CDCl3): d
304 nm. ESI-HRMS (amu): calcd C29H37N3O2 [M+H]: 460.2964; 14.09, 22.66, 25.39, 29.30, 29.45, 29.57, 31.89, 33.55, 44.66,
found [M+H]: 460.2939. 46.61, 123.08, 123.29, 127.59, 128.23, 132.68, 134.43, 135.77,
136.39, 136.64, 136.72, 149.54, 149.62, 154.17, 154.41, 172.52,
4.1.2.11. (3E,5E)-3,5-Bis(pyridin-4-ylmethylene)-1-tetradecanoyl- 188.01. IR (KBr; mmax): 2955, 2923, 2849, 1676, 1640, 1583, 1560,
piperidin-4-one (C4). Yellow powder; yield: 53%; mp 1466, 1428, 1304, 1273, 1162, 986, 939, 790 cm1. UV (CHCl3;
98–100 °C. 1H NMR (300 MHz; CDCl3): d 0.89 (br s, 3H, CH3); kmax): 333 nm. ESI-HRMS (amu): calcd C29H39N3O2 [M+H]:
1.14–1.50 (m, 22H, 11  CH2), 2.14 (br s, 2H, COCH2), 4.72 and 460.2964; found [M+H]: 460.2923.
4.90 (s, 2H each, 2  NCH2), 7.26–7.36 (m, 4H, ArH), 7.73 and 7.78
(s, 1H each, 2  vinylic-H), 8.75 (br s, 4H, ArH). 13C NMR (75 MHz; 4.1.2.16. (3E,5E)-3,5-Bis(pyridin-2-ylmethylene)-1-tetradecanoyl-
CDCl3): d 14.12, 22.68, 24.95, 25.07, 29.25, 29.34, 29.39, 29.55, piperidin-4-one (D4). Waxy yellow powder; yield: 30%; mp
29.63, 31.90, 32.94, 43.12, 46.22, 123.65, 123.92, 134.64, 134.87, 108–110 °C. 1H NMR (300 MHz; CDCl3): d 0.89 (t, J = 6.4 Hz, 3H,
135.69, 141.69, 150.42, 171.92, 176.41, 185.77. IR (KBr; mmax): CH3); 1.17–1.28 (m, 20H, 10  CH2), 1.56 (quintet, J = 7.6 Hz, 2H,
3035, 3019, 2917, 1848, 1676, 1637, 1619, 1592, 1544, 1458, 1430, CH2), 2.38 (t, J = 7.8 Hz, 2H, COCH2), 5.31 and 5.38 (s, 2H each,
1416, 1275, 1257, 1232, 1215, 1167, 996, 930, 831, 759, 717 cm1. 2  NCH2), 7.21–7.25 (m, 2H, ArH), 7.50 (t, J = 6.8 Hz, 2H, ArH),
UV (CHCl3; kmax): 304 nm. ESI-HRMS (amu): calcd C31H41N3O2 7.65–7.77 (m, 4H, ArH, 2  vinylic-H), 8.74 (br s, 2H, ArH). 13C
[M+H]: 488.3277; found [M+H]: 488.3286. NMR (75 MHz; CDCl3): d 14.10, 22.67, 24.87, 25.41, 29.15, 29.10,
29.15, 29.30, 29.33, 29.46, 29.59, 29.63, 29.66, 31.90, 33.56, 34.05,
4.1.2.12. (3E,5E)-1-Palmitoyl-3,5-bis(pyridin-4-ylmethylene) 44.72, 46.57, 122.99, 123.29, 127.57, 128.22, 132.63, 134.87,
piperidin-4-oneone (C5). Yellow powder; yield: 55%; mp 135.76, 136.10, 136.29, 136.71, 149.63, 149.83, 154.40, 172.60,
96–98 °C. 1H NMR (300 MHz; CDCl3): d 0.92–1.49 (m, 29H, 188.09. IR (KBr; mmax): 2956, 2920, 2850, 1639, 1582, 1469, 1427,
1  CH3; 13  CH3), 2.16 (br s, 2H, COCH2), 4.75 and 4.92 (s, 1269, 1163, 1098, 986, 790 cm1. UV (CHCl3; kmax): 334 nm. ESI-
2H each, 2  NCH2), 7.10–7.38 (m, 4H, ArH), 7.74 and 7.79 (s, HRMS (amu): calcd C31H41N3O2 [M+H]: 488.3277; found [M+H]:
1H each, 2  vinylic-H), 8.74–8.78 (br s, 4H, ArH). 13C NMR 488.3232.
(75 MHz; CDCl3): d 14.13, 22.69, 24.96, 29.27, 29.36, 29.67,
31.92, 32.95, 43.14, 46.22, 123.65, 123.91, 134.67, 134.87, 4.1.2.17. (3E,5E)-1-Palmitoyl-3,5-bis(pyridin-2-ylmethylene)
135.71, 141.66, 150.48, 171.92, 185.77. IR (KBr; mmax): 2917, piperidin-4-one (D5). Yellow powder; yield: 53%; mp
2849, 1635, 1619, 1592, 1465, 1416, 1260, 1226, 1167, 98–100 °C. 1H NMR (300 MHz; CDCl3): d 0.89 (br s, 3H, CH3);
996, 929, 841, 820, 755, 714 cm1. UV (CHCl3; kmax): 302 nm. 1.19–1.27 (m, 24H, 12  CH2), 1.57 (br s, 2H, CH2), 2.38 (t,
E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421 419

J = 7.1 Hz, 2H, COCH2), 5.32 and 5.39 (s, 2H each, 2  NCH2), 121.87, 123.58, 128.18, 145.58, 145.82, 151.76, 172.26, 185.86.
7.22–7.78 (m, 8H, ArH; 2  vinylic-H), 8.75 (s, 2H, ArH). 13C IR (KBr; mmax): 3133, 2921, 2848, 1647, 1613, 1567, 1546, 1466,
NMR (75 MHz; CDCl 3): d 14.10, 22.67, 24.94, 25.41, 29.34, 1246, 1168, 1092, 992, 882, 773, 743, 621 cm1. UV (CHCl3;
29.47, 29.68, 31.91, 33.57, 34.01, 44.73, 46.58, 122.98, 123.28, kmax): 382 nm. ESI-HRMS (amu): calcd C27H35NO4 [M+Na]:
127.58, 128.22, 132.60, 134.86, 135.79, 136.13, 136.27, 136.71, 460.2464; found [M+Na]: 460.2436.
149.64, 149.84, 154.43, 172.55, 188.11. IR (KBr; mmax): 3056,
2955, 2918, 1849, 1670, 1650, 1621, 1588, 1578, 1560, 1471, 4.1.2.22. (3E,5E)-3,5-Bis(furan-2-ylmethylene)-1-tetradecanoyl-
1427, 1274, 1167, 987, 787 cm1. UV (CHCl3; kmax): 334 nm. piperidin-4-one (E4). Yellow powder; yield: 74%; mp 74–76 °C.
1
ESI-HRMS (amu): calcd C33H45N3O 2 [M+H]: 516.3590; found H NMR (300 MHz; CDCl3): d 0.90 (br s, 3H, CH3); 1.10–1.40 (m,
[M+H]: 516.3560. 20H, 10  CH2), 1.62 (m, 2H, CH2), 2.44 (t, J = 6.9 Hz, 2H, COCH2),
4.98 and 5.06 (s, 2H each, 2  NCH2), 6.54–6.60 (m, 2H, ArH),
4.1.2.18. (3E,5E)-3,5-Bis(pyridin-2-ylmethylene)-1-stearoylpi- 6.77 (br s, 2H, ArH), 7.51 and 7.55 (s, 1H each, 2  vinylic-H),
peridin-4-one (D6). Yellow powder; yield: 25%; mp 102– 7.65 (br s, 2H, ArH). 13C NMR (75 MHz; CDCl3): d 14.12, 22.69,
104 °C. 1H NMR (300 MHz; CDCl3): d 0.90 (t, J = 6.3 Hz, 3H, CH3), 24.84, 25.34, 29.13, 29.36, 29.48, 29.64, 31.92, 33.42, 33.87,
1.19–1.40 (m, 28H, 14  CH2), 1.53–1.58 (m, 2H, CH2), 2.38 (t, 43.50, 46.14, 112.67, 112.79, 118.05, 118.46, 121.85, 123.57,
J = 7.5 Hz, 2H, COCH2), 5.33 and 5.39 (s, 2H each, 2  NCH2), 128.13, 128.44, 145.56, 145.87, 151.76, 172.26, 185.86. IR (KBr;
7.24–7.28 (m, 2H, ArH), 7.52 (t, J = 7.1 Hz, 2H, ArH), 7.66–7.79 mmax): 2949, 2921, 2947, 1643, 1617, 1575, 1542, 1464, 1443,
(m, 4H, ArH; 2  vinylic-H), 8.76 (br s, 2H, ArH). 13C NMR 1255, 1203, 1171, 982, 884, 757, 620 cm1. UV (CHCl3; kmax):
(75 MHz; CDCl3): d 14.13, 22.70, 25.42, 29.34, 29.27, 29.49, 29.62, 383 nm. ESI-HRMS (amu): calcd C29H39NO4 [M+Na]: 488.2777;
29.66, 29.71, 31.93, 33.59, 44.72, 46.59, 122.98, 123.29, 127.61, found [M+Na]: 488.2751.
128.25, 132.60, 134.88, 135.81, 136.15, 136.27, 136.72, 149.65,
149.87, 154.44, 172.49, 188.18. IR (KBr; v): 3056, 2955, 2918, 4.1.2.23. (3E,5E)-3,5-Bis(furan-2-ylmethylene)-1-palmitoylpi-
1849, 1650, 1622, 1588, 1578, 1560, 1471, 1427, 1275, 1167, peridin-4-one (E5). Yellow powder; yield: 51%; mp 84–
987, 787 cm1. UV (CHCl3; kmax): 334 nm. ESI-HRMS (amu): calcd 85 °C. 1H NMR (300 MHz; CDCl3): d 0.90 (t, J = 6.2 Hz, 3H, CH3);
C35H49N3O2 [M+H]: 544.3903; found [M+H]: 544.3860. 1.24–1.27 (m, 24H, 12  CH2), 1.62 (quintet, J = 6.2 Hz, 2H, CH2),
2.45 (t, J = 6.4 Hz, 2H, COCH2), 5.00 and 5.05 (s, 2H each, 2  NCH2),
6.58 (br s, 2H, ArH), 6.77–6.78 (m, 2H, ArH), 7.54 (br s, 2H,
4.1.2.19. (3E,5E)-1-Oleoyl-3,5-bis(pyridin-2-ylmethylene)piperi-
2  vinylic-H), 7.65 (br s, 2H, ArH). 13C NMR (75 MHz; CDCl3): d
din-4-one (D7). Orange solid; yield: 45%; mp 72–74 °C. 1H
14.09, 22.67, 25.34, 29.34, 29.47, 29.64, 29.67, 31.91, 33.38,
NMR (300 MHz; CDCl3): d 0.88 (dist. t, 3H, CH3), 1.18–1.40 (m,
43.54, 46.12, 112.71, 118.31, 121.89, 123.59, 128.33, 145.69,
22H, 11  CH2), 1.49–1.57 (m, 2H, CH2), 1.99–2.05 (m, 4H,
151.77, 172.28, 185.82. IR (KBr; mmax): 2949, 2920, 2848, 1647,
2  CH2), 2.38 (t, J = 7.7 Hz, 2H, COCH2), 5.31–5.38 (m, 6H,
1613, 1567, 1469, 1256, 1245, 1169, 1093, 992, 774, 744,
2  NCH2; 2  Z-vinylic), 7.24–7.28 (m, 2H, ArH), 7.51 (t,
622 cm1. UV (CHCl3; kmax): 385 nm. ESI-HRMS (amu): calcd
J = 6.4 Hz, 2H, ArH), 7.65–7.77 (m, 4H, ArH; 2  vinylic-H), 8.74
C31H43NO4 [M+Na]: 516.3090; found [M+Na]: 516.3047.
(br s, 2H, ArH). 13C NMR (75 MHz; CDCl3): d 14.17, 22.65, 25.38,
27.20, 28.96, 29.11, 29.21, 29.42, 29.46, 29.50, 29.68, 29.75, 31.88,
32.58, 33.53, 44.72, 46.57, 122.97, 123.27, 127.57, 128.21, 129.76, 4.1.2.24. (3E,5E)-3,5-Bis(furan-2-ylmethylene)-1-stearoylpiperi-
129.93, 132.60, 134.82, 135.79, 136.14, 136.29, 136.70, 149.63, din-4-one (E6). Yellow powder; yield: 86%; mp 82–84 °C. 1H
149.80, 154.41, 172.50, 188.08. IR (KBr; mmax): 2990, 2923, 2851, NMR (300 MHz; CDCl3): d 0.90 (t, J = 5.9 Hz, 3H, CH3); 1.25–1.28
1676, 1640, 1582, 1560, 1465, 1428, 1273, 1164, 986, 939, 790, (m, 28H, 14  CH2), 1.63 (quintet, J = 7.0 Hz, 2H, CH2), 2.44 (t,
746 cm1. UV (CHCl3; kmax): 334 nm. ESI-HRMS (amu): calcd J = 7.5 Hz, 2H, COCH2), 4.98 and 5.07 (s, 2H each, 2  NCH2),
C35H47N3O2 [M+H]: 542.3747; found [M+H]: 542.3703. 6.55–6.60 (m, 2H, ArH), 6.78 (br s, 2H, ArH), 7.52 and 7.56 (s, 1H
each, vinylic-H), 7.65 (br s, 2H, ArH). 13C NMR (75 MHz; CDCl3): d
4.1.2.20. (3E,5E)-1-Butyryl-3,5-bis(furan-2-ylmethylene)piperi- 14.12, 22.69, 25.34, 29.37, 29.49, 29.66, 29.70, 31.93, 33.43,
din-4-one (E2). Yellow powder; yield: 66%; mp 116–118 °C. 43.49, 46.14, 112.66, 112.79, 118.03, 118.44, 121.84, 123.57,
1
H NMR (300 MHz; CDCl3): d 0.95 (t, J = 7.4 Hz, 3H, CH3), 1.68 (sex- 128.17, 128.47, 145.55, 145.86, 151.78, 172.21, 185.91. IR (KBr;
tet, J = 7.4 Hz, 2H, CH2), 2.43 (t, J = 7.5 Hz, 2H, COCH2), 4.98 and 5.07 mmax): 2952, 2920, 2847, 1643, 1617, 1577, 1543, 1463, 1444,
(br s, 2H each, 2  NCH2), 6.54–6.60 (m, 2H, ArH), 6.78 (br s, 2H, 1254, 1198, 1171, 982, 757, 620 cm1. UV (CHCl3; kmax): 382 nm.
ArH), 7.52 and 7.55 (s, 1H each, 2  vinylic-H), 7.65 (br s, 2H, ESI-HRMS (amu): calcd C33H47NO4 [M+Na]: 544.3403; found
ArH). 13C NMR (75 MHz; CDCl3): d 13.95, 18.69, 35.20, 43.44, [M+Na]: 544.3359.
46.11, 112.66, 112.79, 118.05, 118.44, 121.87, 123.53, 128.13,
128.46, 145.59, 145.87, 151.77, 171.96, 185.92. IR (KBr; mmax): 4.1.2.25. (3E,5E)-3,5-Bis(furan-2-ylmethylene)-1-oleoylpiperi-
3109, 1962, 1921, 2872, 2847, 1645, 1609, 1576, 1540, 1464, din-4-one (E7). Brownish/yellow low melting solid; yield:
1440, 1263, 1244, 1200, 1174, 1024, 977, 753, 621 cm1. UV (CHCl3; 28%; 1H NMR (300 MHz; CDCl3): d 0.90 (t, J = 6.6 Hz, 3H, CH3),
kmax): 383 nm. ESI-HRMS (amu): calcd C19H19NO4 [M+Na]: 1.10–1.40 (m, 22H, 11  CH2), 1.63 (m, 2H, CH2), 2.02 (m, 4H,
348.1212; found [M+Na]: 348.1189. CH2), 2.44 (t, J = 7.6 Hz, 2H, COCH2), 4.98 and 5.07 (s, 2H each,
2  NCH2), 5.36 (m, 2H, Z vinylic-H), 6.58 (m, 2H, ArH), 6.78–6.79
4.1.2.21. (3E,5E)-1-Dodecanoyl-3,5-bis(furan-2-ylmethylene) (m, 2H, ArH), 7.52 and 7.56 (s, 1H each, 2  vinylic-H), 7.65 (br s,
piperidin-4-one (E3). Orange powder; yield: 57%; mp 78– 2H, ArH). 13C NMR (75 MHz; CDCl3): d 14.12, 22.68, 23.49, 27.22,
81 °C. 1H NMR (300 MHz; CDCl3): d 0.90 (t, J = 6.1 Hz, 3H, CH3), 27.79, 28.95, 29.00, 29.06, 29.33, 29.53, 29.65, 29.72, 29.77,
1.10–1.40 (m, 16H, 8  CH2), 1.62 (quintet, J = 6.7 Hz, 2H, CH2), 31.91, 39.69, 44.08, 46.33, 112.70, 112.91, 118.36, 118.66, 122.31,
2.44 (t, J = 7.6 Hz, 2H, COCH2), 4.98 and 5.06 (s, 2H each, 123.75, 127.40, 127.85, 129.77, 129.97, 145.82, 145.99, 151.48,
2  NCH2), 6.55–6.60 (m, 2H, ArH), 6.77–6.78 (m, 2H, ArH), 151.70, 168.48, 185.35. IR (KBr; mmax): 3105, 2924, 2852, 1644,
7.52 and 7.55 (s, 1H each, 2  vinylic-H), 7.65 (br s, 2H, ArH). 1611, 1569, 1541, 1465, 1283, 1258, 1166, 1020, 988, 758,
13
C NMR (75 MHz; CDCl3): d 14.10, 22.67, 25.34, 29.32, 29.35, 622 cm1. UV (CHCl3; kmax): 383 nm. ESI-HRMS (amu): calcd
29.47, 29.59, 31.90, 33.40, 43.52, 46.14, 112.72, 118.02, 118.43, C33H43NO4 [M+Na]: 542.3246; found [M+Na]: 542.3198.
420 E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421

4.2. Biological evaluations a 96-well tissue culture plate. Cells were pretreated with the test
compounds for 24 h (100 lM) and placed in a humidified incubator
4.2.1. Cytotoxicity evaluations (VWR International, Mississauga, ON, Canada) for 24 h. Plates were
The Molt 4/C8, CEM and L1210 assays were conducted using a removed from incubator and LDH release assessment was per-
literature methodology.45 The data is reported in Table 1. formed according to the manufacturer’s instructions. The mem-
brane damage was expressed as the percentage release of the
4.2.2. Cell culture enzyme lactate dehydrogenase (LDH) with respect to the positive
Human diploid lung fibroblast WI-38 cells were obtained from control. The absorbance was measured using BioTek, Power wave
Cederlane Labs (Burlington, ON, Canada) and cultured according XS2 spectrophotometer (BioTek, Winooski, VT, USA) at emission
to the supplier’s instructions. The WI-38 cells were maintained in spectra of 490 nm. The data is reported in Figure 5.
75 cm2 culture flasks in a humidified incubator (VWR Interna-
tional, Mississauga, ON, Canada) at temperature of 37 °C. WI-38 4.2.9. Topoisomerase IIa activity assay
cells were supplied with DMEM medium, supplemented with Topoisomerase IIa inhibitory activity was performed using a
10% fetal bovine serum (FBS) and 100 mg/L of penicillin and strep- commercial kit, (Topogen Inc, Columbus, OH, USA). The com-
tomycin. The cells were used between passages 2–8, and the pounds were challenged against 4 units of topoisomerase enzyme
growth medium of cultured cells was changed every 48 h. using pHOT1 DNA as the substrate. All the preparatory and analysis
steps were conducted as per the manufacturer’s instructions and
4.2.3. Ferric reducing ability of plasma (FRAP) assay all samples (10 lM) were loaded directly onto a 2% agarose gel
The FRAP assay was performed according to the method in 1 TBE (Tris/Borate/EDTA) running buffer. In addition to the test
described earlier.15 The test compounds (20 lL) and FRAP working compounds, sorafenib was also used as a standard drug in the
reagent (180 lL) were added to 96-well microplates and absor- assay. The DNA bands were visualized by placing gels on a UV sam-
bance was read at 590 nm using the FLUOstar OPTIMA plate reader ple tray, followed by exposure to UV light using gel Doc™ EZ Sys-
(BMG Labtech, Durham, NC, USA). The antioxidant capacity results tem (Biorad, ON, Canada).
were expressed as lM TE L1 of solution. The data is reported in
Table 3. 4.2.10. Biostatistics
All the biological analysis was conducted using completely ran-
4.2.4. Oxygen radical absorbance capacity (ORAC) assay domized design (CRD) of experiments. One way analysis of vari-
The ORAC assay was performed according to the method ance (ANOVA) was performed to analyse all data from biological
described earlier with appropriate controls.15 The fluorescence sig- assays using a commercial software package.47 Data was consid-
nal was read at excitation and emission wavelength of 485 nm and ered significant at p60.05 and the means were compared using
510 nm using the FLUOstar OPTIMA plate reader (BMG Labtech, Tukey’s multiple means comparison test. All the analyses were per-
Durham, NC, USA). The antioxidant capacity results were expressed formed in triplicates.
as lM TE L1 of solution. The data is reported in Table 3.
4.2.11. Statistical analyses using electronic and physicochemical
4.2.5. The 2,2-diphenylpicrylhydrazyl (DPPH) assay parameters
The DPPH assay was performed according to the method Electronic and physicochemical properties such as charge b,
described earlier.15 The test compounds were challenged against partition coefficient (log P), and molar refractivity of the molecule
0.2 mM DPPH solutions in 1:1 ratio in 96 well plates and the absor- were calculated using a commercial software.46 The correlations
bance was measured using FLUOstar OPTIMA plate reader (BMG between these molecular descriptors and the IC50 values of the
Labtech, Durham, NC, USA) at 520 nm. The data is reported in three cell lines (Table 1) were generated using a commercial soft-
Table 3. ware package.47 All significant correlations are reported in Table 2.

4.2.6. Cellular reactive oxygen species (ROS) detection assay Acknowledgments


Cellular ROS was analyzed using Abcam’s cellular reactive oxy-
gen species detection assay kit (Abcam Inc, Toronto, ON, Canada). The authors thank the following agencies for financial support,
The assay uses the cell permeant reagent 20 ,70 -dichlorofluorescin namely the Nova Scotia Health Research Foundation (A.J., E.P.),
diacetate (DCFDA), a fluorogenic dye that measures hydroxyl, per- Natural Sciences and Engineering Research Council (NSERC) of
oxyl and other reactive oxygen species (ROS) activity within the Canada (A.J.), Fonds voor Wetenschappelijk Onderzoek-Vlaanderen
cell. The results were expressed as percentage inhibition of ROS (J.B.). Appreciation is extended to Mrs. Lizette van Berckelaer for
with respect to control cell using FLUOstar OPTIMA plate reader conducting the HeLa, CEM and L1210 assays. H.P.V.R. gratefully
(BMG Labtech, Durham, NC, USA) with maximum excitation and acknowledges funding from the Canada Research Chair program
emission spectra of 490 nm and 510 nm, respectively. The data is and the Discovery Grant program of the Natural Sciences and Engi-
reported in Table 3. neering Research Council (NSERC) of Canada.

4.2.7. Tyrosinase inhibitor assay Supplementary data


Tyrosinase inhibition potential was determined using the mod-
ified dopachrome method with L-DOPA as substrate.15 Assay was Supplementary data (spectroscopic data) associated with this
conducted in a 96-well microtitre plate using test compounds article can be found, in the online version, at http://dx.doi.org/
and kojic acid (100 lM) as the reference compound. The results 10.1016/j.bmc.2014.12.042.
were expressed as percentage inhibition of tyrosinase with respect
to the assay control. The data is reported in Figure 4.
References and notes

4.2.8. In vitro toxicity analysis 1. Aggarwal, B. B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian
In vitro toxicity analysis was conducted using the CytoTox- Solid Gold. In The molecular Targets and Therapeutic Uses of Curcumin in Health
and Disease; Sangi, C., Ed.; Springer: New York, 2007; pp 1–75.
ONE™ homogeneous membrane integrity assay (Promega Corpora-
2. Julie, S.; Jurenka, M. Altern. Med. Rev. 2009, 14, 141.
tion, Madison, WI, USA). Briefly, 2  104 WI-38 cells were seeded in
E. Potter et al. / Bioorg. Med. Chem. 23 (2015) 411–421 421

3. Perry, M.-C.; Demeule, M.; Régina, A.; Moumdjian, R.; Béliveau, R. Mol. Nutr. 28. Jha, A.; Mukherjee, C.; Prasad, A. K.; Parmar, V. S.; Vadaparti, M.; Das, U.; De
Food Res. 2010, 54, 1192. Clercq, E.; Balzarini, J.; Stables, J. P.; Shrivastav, A.; Sharma, R. K.; Dimmock, J. R.
4. Lee, Y.-K.; Park, S. Y.; Kim, Y.-M.; Park, O. J. Ann. N.Y. Acad. Sci. 2009, Bioorg. Med. Chem. Lett. 2010, 20, 1510.
1171, 489. 29. McClendon, A. K.; Osheroff, N. Mutat. Res. 2007, 623, 83.
5. Yang, C.-L.; Liu, Y.-Y.; Ma, Y.-G.; Xue, Y.-X.; Liu, D.-G.; Ren, Y.; Liu, X.-B.; Li, Y.; 30. Deweese, J. E.; Osheroff, N. Nucl. Acids Res. 2009, 37, 738.
Li, Z. PLoS One 2012, 7, e37960. 31. Burden, D. A.; Osheroff, N. BBA Gene Struct. Expr. 1998, 1400, 139.
6. Aggarwal, B. B.; Surh, Y.-J.; Shishodia, S. The molecular targets and therapeutic 32. Wang, J. C. Nat. Rev. Mol. Cell Biol. 2002, 3, 430.
uses of curcumin in health and disease; Springer, 2007. 33. Champoux, J. J. Annu. Rev. Biochem. 2001, 70, 369.
7. National Institutes of Health. http://clinicaltrials.gov/ct2/ 34. Chene, P.; Rudloff, J.; Schoepfer, J.; Furet, P.; Meier, P.; Qian, Z.; Schlaeppi, J. M.;
results?term=curcumin&pg=1 (last accessed July 11, 2014). Schmitz, R.; Radimerski, T. BMC Chem. Biol. 2009, 9, 1.
8. Bayet-Robert, M.; Kwiatkowski, F.; Leheurteur, M.; Gachon, F.; Planchat, E.; 35. Kuznetsova, L.; Chen, J.; Sun, L.; Wu, X.; Pepe, A.; Veith, J. M.; Pera, P.; Bernacki,
Abrial, C.; Mouret-Reynier, M. A.; Durando, X.; Barthomeuf, C.; Chollet, P. R. J.; Ojima, I. Bioorg. Med. Chem. Lett. 2006, 16, 974.
Cancer Biol. Ther. 2010, 9, 8. 36. Jaracz, S.; Chen, J.; Kuznetsova, L. V.; Ojima, I. Bioorg. Med. Chem. 2005, 13, 5043.
9. Sun, S. H.; Huang, H. C.; Huang, C.; Lin, J. K. Eur. J. Pharmacol. 2012, 690, 22. 37. Sparreboom, A.; Wolff, A. C.; Verweij, J.; Zabelina, Y.; van Zomeren, D. M.;
10. Epelbaum, R.; Schaffer, M.; Vizel, B.; Badmaev, V.; Bar-Sela, G. Nutr. Cancer McIntire, G. L.; Swindell, C. S.; Donehower, R. C.; Baker, S. D. Clin. Cancer Res.
2010, 62, 1137. 2003, 9, 151.
11. Goss, C. H.; Genatossio, A.; Rowbotham, R. K.; Hemblett, N.; McNamara, S.; 38. Blank-Porat, D.; Gruss-Fischer, T.; Tarasenko, N.; Malik, Z.; Nudelman, A.;
Knowles, M.; Brass-Ernst, L.; Aitken, M. L.; Zeitlin, P. L.; Boyle, M. P. Ped. Rephaeli, A. Cancer Lett. 2007, 256, 39.
Pulmonol. 2006, 41, 293. 39. Rephaeli, A.; Waks-Yona, S.; Nudelman, A.; Tarasenko, I.; Tarasenko, N.;
12. Taylor, R. A.; Leonard, M. C. Altern. Med. Rev. 2011, 16, 152. Phillips, D. R.; Cutts, S. M.; Kessler-Icekson, G. Br. J. Cancer 2007, 96, 1667.
13. Kelley, B. J.; Knopman, D. S. Neurologist 2008, 14, 299. 40. Pati, H. N.; Das, U.; Quail, J. W.; Kawase, M.; Sakagami, H.; Dimmock, J. R. Eur. J.
14. Sharma, R. A.; Steward, W. P.; Gescher, A. J. Pharmacokinetics and Med. Chem. 2008, 43, 1.
Pharmacodynamics of Curcumin. In The Molecular Targets and Therapeutic 41. Dimmock, J. R.; Arora, V. K.; Quail, J. W.; Pugazhenthi, U.; Allen, T. M.; Kao, G.
Uses of Curcumin in Health and Disease; Aggarwal, B., Surh, Y.-J., Shishodia, S., Y.; De Clercq, E. J. Pharm. Sci. 1994, 83, 1124.
Eds.; Springer: US, 2007; pp 453–470. 42. Kumar, R. R.; Perumal, S.; Senthilkumar, P.; Yogeeswari, P.; Sriram, D. J. Med.
15. Bhullar, K.; Jha, A.; Youssef, D.; Rupasinghe, H. P. V. Molecules 2013, 18, 5389. Chem. 2008, 51, 5731.
16. Jha, A.; Zhao, J.; Stanley, T.; Cameron, E.; De Clercq, J.; Balzarini, E. K.; 43. Dimmock, J. R.; Padmanilayam, M. P.; Puthucode, R. N.; Nazarali, A. J.;
Manavathu, J. P. Drug Des. Discov. 2006, 3, 304. Motaganahalli, N. L.; Zello, G. A.; Quail, J. W.; Oloo, E. O.; Kraatz, H.-B.;
17. Nichols, C. E.; Youssef, D.; Harris, R. G.; Jha, A. ARKIVOC 2006, 13, 64. Prisciak, J. S.; Allen, T. M.; Santos, C. L.; Balzarini, J.; De Clercq, E.; Manavathu, E.
18. Youssef, D.; Nichols, C. E.; Cameron, T. S.; Balzarini, J.; De Clercq, E.; Jha, A. K. J. Med. Chem. 2001, 44, 586.
Bioorg. Med. Chem. Lett. 2007, 17, 5624. 44. Leonova, E. S.; Makarov, M. V.; Rybalkina, E. Y.; Nayani, S. L.; Tongwa, P.; Fonari,
19. Dimmock, J. R.; Jha, A.; Zello, G. A.; Quail, J. W.; Oloo, E. O.; Nienaber, K. H.; A.; Timofeeva, T. V.; Odinets, I. L. Eur. J. Med. Chem. 2010, 45, 5926.
Kowalczyk, E. S.; Allen, T. M.; Santos, C. L.; De Clercq, E.; Balzarini, J.; 45. Balzarini, J.; De Clercq, E.; Mertes, M. P.; Shugar, D.; Torrence, P. F. Biochem.
Manavathu, E. K.; Stables, J. P. Eur. J. Med. Chem. 2002, 37, 961. Pharmacol. 1982, 31, 3673.
20. Jha, A.; Mukherjee, C.; Prasad, A. K.; Parmar, V. S.; Clercq, E. D.; Balzarini, J.; 46. Dykes, D. J.; Waud, W. R. Murine L1210 and P388 leukemias. In Tumor Models in
Stables, J. P.; Manavathu, E. K.; Shrivastav, A.; Sharma, R. K.; Nienaber, K. H.; Cancer Research; Teicher, B. A., Ed.; Humana Press: New York, 2011; pp 23–41.
Zello, G. A.; Dimmock, J. R. Bioorg. Med. Chem. 2007, 15, 5854. 47. Dimmock, J. R.; Jha, A.; Kumar, P.; Zello, G. A.; Quail, J. W.; Oloo, E. O.; Oucharek,
21. Dimmock, J. R.; Jha, A.; Zello, G. A.; Sharma, R. K.; Shrivastav, A.; Selvakumar, P.; J. J.; Pasha, M. K.; Seitz, D.; Sharma, R. K.; Allen, T. M.; Santos, C. L.; Manavathu,
Allen, T. M.; Santos, C. L.; Balzarini, J.; De Clercq, E.; Manavathu, E. K.; Stables, J. E. K.; De Clercq, E.; Balzarini, J.; Stables, J. P. Eur. J. Med. Chem. 2002, 37, 35.
P. J. Enzyme Inhib. Med. Chem. 2003, 18, 325. 48. Paul, N. K.; Jha, M.; Bhullar, K. S.; Vasantha Rupasinghe, H. P.; Balzarini, J.; Jha,
22. Youssef, D.; Potter, E.; Jha, M.; De Clercq, E.; Balzarini, J.; Stables, J. P.; Jha, A. A. Eur. J. Med. Chem. 2014, 87, 467.
Bioorg. Med. Chem. Lett. 2009, 19, 6364. 49. Charge b, partition coefficients (log P) and molar refractivity (MRmol) were
23. Jha, A.; Dimmock, J. R. Synth. Commun. 2003, 33, 1211. calculated by Chem3DÒ computer program (version 8.0) after energy
24. Jha, A.; Duffield, K. M. Indian J. Chem., Sec. B Org. Chem. Incl. Med. Chem. 2006, 45, minimization of the molecules using MOPAC. Chem3DÒ is obtainable from
2313. Cambridge Soft Corporation, Cambridge, MA, USA.
25. Jha, A.; Mukherjee, C.; Rolle, A. J.; De Clercq, E.; Balzarini, J.; Stables, J. P. Bioorg. 50. MinitabÒ Release 14 obtained from Minitab Inc., Quality Plaza, 1829 Pine Hall
Med. Chem. Lett. 2007, 17, 4545. Road, State College, PA 16801-3008, USA.
26. Parmar, V.; Jain, S.; Bisht, K.; Sharma, N. K.; Gupta, S.; Prasad, A. K.; Jha, A.; 51. Nair, S. V. G.; Ziaullah; Rupasinghe, H. P. V. PLoS One 2014, 9, e107149.
Malhotra, S.; Sharma, S. K.; Bracke, M. Indian J. Chem., Sec. B Org. Chem. Incl. 52. Shen, H. J.; Wang, Y. H.; Xu, J. Zhonghua zhong liu za zhi [Chin. J. Oncol.] 2013, 35,
Med. Chem. 1999, 628. 98.
27. Dimmock, J. R.; Jha, A.; Zello, G. A.; Allen, T. M.; Santos, C. L.; Balzarini, J.; De 53. Sanchez-Ferrer, A.; Rodriguez-Lopez, J. N.; Garcia-Canovas, F.; Garcia-Carmona,
Clercq, E.; Manavathu, E. K.; Stables, J. P. Pharmazie 2003, 58, 227. F. Biochim. Biophys. Acta 1995, 1247, 1.

You might also like