Download as pdf or txt
Download as pdf or txt
You are on page 1of 13

European Journal of Medicinal Chemistry 120 (2016) 51e63

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


journal homepage: http://www.elsevier.com/locate/ejmech

Research paper

Anticancer properties of new synthetic hybrid molecules combining


naphtho[2,3-b]furan-4,9-dione or benzo[f]indole-4,9-dione motif
with phosphonate subunit
 ski c, Dorota Pomorska a,
Katarzyna Gach a, 1, Jakub Modranka b, 1, Jacek Szyman
Urszula Krajewska , Marek Mirowski , Tomasz Janecki b, **, Anna Janecka a, *
d d

a
Department of Biomolecular Chemistry, Faculty of Medicine, Medicinal University of Lodz, Mazowiecka 6/8, 92-215, Lodz, Poland
b
Institute of Organic Chemistry, Faculty of Chemistry, Lodz University of Technology, Zeromskiego 116, 90-924, Lodz, Poland
c
Central Scientific Laboratory, Division of Public Health, Faculty of Health Sciences, Medical University of Lodz, Mazowiecka 6/8, 92-215, Lodz, Poland
d
Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Medical University of Lodz, Muszynskiego 1, 90-151, Lodz, Poland

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

Article history: In this paper we report an efficient and general synthesis of substituted 3-diethoxyphosphorylnaphtho
Received 13 February 2016 [2,3-b]furan-4,9-diones and 3-diethoxyphosphorylbenzo [f]indole-4,9-diones which integrate the nat-
Received in revised form ural 1,4-naphtalenedione scaffold, present in several anticancer agents with the phosphonate moiety.
26 April 2016
The cytotoxicity of such hybrid molecules was tested against two leukemia cell lines, HL-60 and NALM-6
Accepted 1 May 2016
Available online 3 May 2016
and against a breast adenocarcinoma MCF-7 cell line. Selected compounds were also tested on normal
human cells: HUVEC and MCF-10A. In general, naphthofuran-4,9-diones showed much higher cytotoxic
activity (IC50 values below 10 mM) than benzoindole-4,9-diones. The most promising 2-(2-chlorophenyl)-
Keywords:
Naphthofurandiones
3-diethoxyphosphorylnaphtho [2,3-b]furan-4,9-dione, with the highest cytotoxic activity in the MTT
Phosphonate moiety test, was chosen for further evaluation of its anticancer potential. This compound, tested on HL-60 and
Hybrid molecules MCF-7 cells inhibited cell proliferation, generated DNA damage and induced apoptosis. The suggested
Cytotoxicity mechanism of its cytotoxic activity was the generation of intracellular reactive oxygen species and the
Apoptosis induction of mitochondrial membrane potential dissipation.
© 2016 Elsevier Masson SAS. All rights reserved.

1. Introduction isolated from Streptomyces peucetius and its 2-hydroxyacetyl


analog, doxorubicin [10] 4 (Fig. 1), both containing a tetracene-
The 1,4-naphtalenedione scaffold, usually ortho-condensed with dione framework and both used as drugs in cancer chemotherapy
one or two planar rings is a well-known pharmacophoric unit for the treatment of various human neoplastic diseases including
present in many natural and synthetic compounds displaying leukaemias, lymphomas, sarcomas, carcinomas and breast cancers
diverse biological activities. Natural 1,4-naphthoquinones such as [11].
plumbagin 1 or lapachol 2 (Fig. 1) had been investigated during the The mechanism of antitumor activity of quinones has been
past years, mainly due to their antibacterial, antifungal, trypano- extensively investigated. Due to their structural properties, qui-
cidal and anticancer activities [1e3]. Many synthetic derivatives of nones participate in multiple biological oxidative processes. The
such compounds were obtained and several of them turned out to fundamental feature of quinones is their ability to act as oxidizing
be highly cytotoxic against various cancer cell lines [4e8]. Well or dehydrogenating agents, the driving force of this being the for-
known examples of 1,4-naphtalenediones are daunorubicin [9] 3 mation of a fully aromatic system [12]. The two major mechanisms
of quinone cytotoxicity are the stimulation of oxidative stress and/
or alkylation of cellular nucleophiles, which encompasses a large
range of biomolecules [7,12e14]. Quinones undergo either one- or
* Corresponding author. two-electron reduction, leading to the formation of reactive oxygen
** Corresponding author.
species (ROS), including superoxide, hydrogen peroxide, and ulti-
E-mail addresses: tomasz.janecki@p.lodz.pl (T. Janecki), anna.janecka@umed.
lodz.pl (A. Janecka).
mately the hydroxyl radicals. The production of ROS can cause se-
1
K.G. and J.M. contributed equally. vere oxidative stress within cells through the formation of oxidized

http://dx.doi.org/10.1016/j.ejmech.2016.05.002
0223-5234/© 2016 Elsevier Masson SAS. All rights reserved.
52 K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63

Fig. 1. Structures of Plumbagin, Lapachol, Daunorubicin, Doxorubicin and compounds containing naphtho [2,3-b]furan-4,9-dione or benzoindole-4,9-dione framework.

cellular macromolecules, including lipids, proteins and DNA, lead- possessed significant selectivity in antitumor activity compared to
ing to cell damage and apoptosis. Alternatively, quinones are normal cells [22e24].
Michael-type acceptors, and cellular damage can occur through Additionally, 2-substituted naphtho [2,3-b]furan-4,9-diones
alkylation of crucial cellular proteins and/or DNA [14]. Generating suppressed hyperproliferation of human keratinocytes and could
oxidative stress and cellular damage results in the inhibition of cell be potentially useful in the treatment of psoriasis, a common,
proliferation, cell cycle arrest and apoptosis [2,4,14,15]. ROS are immune-mediated inflammatory and scaling skin disease [25].
important participants in the regulation of normal cellular pro- Likewise, compounds with the benzoindole-4,9-dione frame-
cesses, such as gene expression, metabolism, cell differentiation, work were found in nature and show promising biological activity.
proliferation, and apoptosis. However, at higher concentrations, Thus, 3-methyl-1H-benzo [f]indole-4,9-dione 7a [26] and Utha-
ROS become toxic and induce cell death by various signaling mycin B 7b [27] (Fig. 1) were isolated from Goniothalamus tapis Miq.
pathways [15]. Compounds that are able to modulate the redox and Streptomyces albus, respectively. Furthermore, their synthetic
balance in cancer cells are potential candidates for the develop- analogs of general structure 8 (Fig. 1), containing ester functionality
ment of anticancer drugs. Therefore, various chemical modifica- in position 3 were extensively tested for their cytotoxicity against
tions of compounds containing the 1,4-naphtalenedione scaffold numerous human cancer cell lines and it turned out that several of
are sought in order to obtain analogs with enhanced bioactivity and these analogs are very potent anticancer agents [28e32]. For
reduced toxicity, thereby maximizing their therapeutic potential. example, 3-ethoxycarbonylbenzoindole-4,9-dione 8a (R1 ¼ Et,
Interesting groups of such analogs are compounds with naphtho R2 ¼ cyclohexyl) exhibited greater cytotoxic activity against a wide
[2,3-b]furan-4,9-dione or benzo [f]indole-4,9-dione framework variety of human tumor cell lines than etoposite [29] and ben-
which often display cytotoxic and other biological activities. For zoindole-4,9-dione 8b (R1 ¼ Et, R2 ¼ 3,4-dimethylphenyl) displayed
example, maturone 5 (Fig. 1) was isolated from a Mexican plant, selective cytotoxicity against lung cancer cells which was compa-
Cacalia decomposita, the root extract of which has been used for the rable to doxorubicin [30].
treatment of diabetes and other diseases [16e19]. Other natural Continuing our research on biologically important phosphory-
compounds with the naphthofuranodione framework are avice- lated oxa- and aza-heterocycles we decided to synthesize com-
quinones 6 (Fig. 1). These compounds were isolated from the stem pounds with naphtho [2,3-b]furan-4,9-dione and benzo [f]indole-
bark of Avicennia alba or root and stem extracts of Mendonica 4,9-dione skeletons containing phosphoryl functionality instead of
cowanii and display cytotoxic activity [20,21]. Also several synthetic a carboxyl group in position 3. It is well known, that the phosphoryl
naphtho [2,3-b]furan-4,9-diones containing alkyl substituents in group can mimic the tetrahedral intermediates formed in enzy-
position 2 and/or EWG groups, such as carboxyl, alkoxycarbonyl or matic reactions involved in the carboxyl group metabolism
acyl, in position 3 were prepared and some of these analogs [33e35]. This rationale has resulted in the design of some
K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63 53

important drugs, such as N-phosphonoacetyl-L-aspartic acid (PALA) Table 1


with anticancer activity or fosinopril which is a clinically useful Yields of the obtained 13a-f.

antihypertensive agent [36]. We reasoned that such replacement Compound 13 R Yielda [%]
can produce biologically active analogs which might inhibit key a Me 10
enzymes or interact with receptors which normally bind the cor- b Ph 61
responding original compounds. Also, it is well known that intro- c 55
ducing organophosphorus functionalities into organic molecules
can increase their biological activity [33,34].
Accordingly, in this paper we report on efficient and general
synthesis of substituted 3-diethoxyphosphorylnaphtho [2,3-b] d 63
furan-4,9-diones 13 and 3-diethoxyphosphorylbenzo [f]indole-4,9-
diones 18 which integrate the natural 1,4-naphtalenedione scaffold
and the phosphonate moiety. Such hybrid molecules could poten-
tially improve their precursors’ cytotoxic properties and/or help to e 76
overcome their limitations [37,38].

2. Results and discussion


f 88
2.1. Chemistry

The target compounds were synthesized applying modified


methodology originally reported for the synthesis of 3-
alkoxycarbonylnaphtho [2,3-b]furan-4,9-diones [39] or 3-
a
alkoxycarbonylbenzo [f]indole-4,9-diones [29], i.e. performing the Yield of pure, isolated products based on 9.
reaction of 2,3-dichloro-1,4-naphthoquinone with b-ketophosph-
onates or diethoxyphosphorylacetate which were used instead of
b-ketoesters or malonate, respectively. confirmed by MS and spectroscopic methods. Formation of 13 can
A series of 2-substituted 3-diethoxyphosphorylnaphtho [2,3-b] be easily rationalized by assuming the Michael addition of phos-
furan-4,9-diones 13a-f were synthesized as shown in Scheme 1. phonate 9 to naphthoquinone 10, followed by elimination of the
Starting diethyl acylmethylphosphonates 9a,b are commercially chlorine anion and formation of the substitution product 11 which
available and phosphonates 9c-f were synthesized applying is in equilibrium with the enol form 12. Intramolecular substitution
modified literature procedure [40], from diethyl methyl- of the second chlorine ion in 12, via addition/elimination mecha-
phosphonate and corresponding ester in the presence of LDA. nism, gave the target furandiones 13.
Phosphonates 9aef were next used in the reaction with commer- We also decided to prepare the monoester derivative 14 which
cially available 2,3-dichloro-1,4-naphthoquinone 10 and after better mimics tetrahedral intermediates involved in carboxyl group
standard work-up and purification by column chromatography metabolism in comparison to the diester [36]. In this respect,
expected 3-diethoxyphosphorylnaphtho [2,3-b]furan-4,9-diones naphthofurandione 13c was chemoselectively hydrolized to 3-
13aef were obtained. Reactions with aryloylmethylphosphonates hydroxyethoxyphosphoryl-2-(2-chlorophenyl)naphtho [2,3-b]
9bef proceeded effectively and final 13bef were obtained in furan-4,9-dione 14 in 93% yield by the treatment with lithium
satisfactory yields (Table 1). Only the reaction with acetylmethyl- bromide followed by hydrolysis with 1 M hydrochloric acid
phosphonate 9a was much less efficient and a complex mixture of (Scheme 2).
products was formed. Fortunately, it was possible to isolate the In turn, 3-diethoxyphosphorylbenzo [f]indole-4,9-diones 18aee
desired naphthofurandione 13a in low, 10% yield. Structures of the were synthesized in the reaction sequence shown in Scheme 3.
obtained naphthofurandiones 13aef were unambiguously Reaction of 2,3-dichloro-1,4-naphthoquinone 10 with ethyl

Scheme 1. Synthesis of 3-diethoxyphosphorylnaphtho [2,3-b]furan-4,9-diones 13a-f.


54 K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63

2.2. Biological evaluation

2.2.1. In vitro cytotoxicity of new compounds against cancer cell


lines
The obtained 3-diethoxyphosphorylnaphtho [2,3-b]furan-4,9-
diones 13aef and 3-hydroxyethoxyphosphoryl-2-(2-
chlorophenyl)naphtho [2,3-b]furan-4,9-dione 14 as well as 3-
diethoxyphosphorylbenzo [f]indole-4,9-diones 18aee were tested
in vitro against three human cancer cell lines: leukemia NALM-6
and HL-60, as well as breast cancer MCF-7, and the results are
Scheme 2. Synthesis of 3-hydroxyethoxyphosphoryl-2-(2-chlorophenyl)naphtho [2,3-
shown in Table 3. Doxorubicin was used as the positive control. In
b]furan-4,9-dione 14.

Scheme 3. Synthesis of 3-diethoxyphosphorylbenzo [f]indole-4,9-diones 18a-e.

diethoxyphosphorylacetate (15) in the presence of NaH furnished general, naphthofuran-4,9-diones, except for the monoester de-
2-chloro-3-(2-ethoxy-1-diethoxyphosphoryl-2-oxoethyl)naphtha- rivative 14, showed high cytotoxic activity with the IC50 values
lene-1,4-dione 16 in 96% yield, via an addition/elimination mech- below 10 mM (after 48 h incubation), in all three cancer cell lines. On
anism. This compound treated with various aliphatic amines gave, the other hand benzoindole-4,9-diones were moderately active,
after standard work up and purification by column chromatog- with IC50 values of 20e60 mM. It is worth noticing that cytotoxicity
raphy, target diethoxyphosphorylbenzoindole-4,9-diones 18aee in of naphthofurandiones was about 2e3-fold higher in MCF-7 cells
the yields given in Table 2. Structures of the obtained benzoindo- than in the tested leukemia cell lines. Selected naphthofurandiones
lediones 18aee were unambiguously confirmed by MS and spec- were also tested on human mammary gland/breast cell line
troscopic methods. Clearly, in the first step of the reaction, amine (MCF10A) and human umbilical vein endothelial cells (HUVEC) to
displaces the chlorine ion to form 17 which undergoes intra- evaluate their influence on normal cells. The toxicity of the tested
molecular nucleophilic substitution on acyl group. compounds against leukemia and HUVEC cells was similar. How-
ever, these compounds were 4-fold less toxic for MCF-10A as
compared with MCF-7 cells. Compound 13c was selected for more
detailed evaluation of its anticancer potential against HL-60 and
MCF-7 cells. The MTT assay was performed for 13c after 24 h in-
cubation. The IC50 values were 7.27 ± 0.08 and 3.40 ± 0.15 for HL-60
and MCF-7 cells, respectively, and these concentrations were used
Table 2 in all further experiments.
Yields of the obtained 18a-e.

Compound 18 R Yielda [%]


2.2.2. Inhibition of cell proliferation, induction of DNA damage and
a Me 94 apoptosis
b n-Pr 88
c c-Pr 81
The ability of 13c to inhibit cell proliferation, generate DNA
d n-Hex 98 damage and induce apoptotic cell death was examined by flow
e 52 cytometry using the ‘Apoptosis, DNA Damage, and Cell Proliferation
Kit’ (BD Bioscience). After 24 h treatment with compound 13c at
IC50 concentration, HL-60 and MCF-7 were incubated with bro-
modeoxyuridine (BrdU) for additional 8 h. Cells were simulta-
a
Yield of pure, isolated products based on 16. neously stained with fluorochrome-labeled anti-BrdU, H2AX
K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63 55

Table 3
In vitro cytotoxic activity of naphthofurandiones 13a-f, monoacid 14 and benzo [f]indolediones 18a-e tested on three cancer cell lines and two normal cell lines (HUVEC and
MCF-10A).

Compound IC50a (mM)

HL-60 NALM-6 MCF-7 HUVEC MCF-10A

13a 8.13 ± 0.16 5.85 ± 0.55 5.70 ± 0.21


13b 6.01 ± 0.18 6.06 ± 0.40 2.40 ± 0.30
13c 6.35 ± 0.46 5.07 ± 0.58 2.34 ± 0.18 3.73 ± 0.18 10.03 ± 0.43
13d 5.44 ± 0.25 4.82 ± 0.26 2.90 ± 0.12 4.06 ± 0.25 11.31 ± 0.71
13e 8.66 ± 0.88 6.65 ± 0.35 2.55 ± 0.05
13f 5.94 ± 0.26 5.32 ± 0.53 2.85 ± 0.85 4.85 ± 0.49 9.79 ± 0.48
14 486.72 ± 27.7 478.64 ± 45.68 150 ± 10.08
18a 59.66 ± 4.23 61.74 ± 4.7 50 ± 6.01
18b 56.20 ± 3.84 60.03 ± 2.02 43.93 ± 5.18
18c 65.05 ± 2.21 51.30 ± 3.99 102.0 ± 6.98
18d 30.74 ± 3.55 44.3 ± 3.2 19.5 ± 4.4
18e 42.35 ± 4.47 60.07 ± 2.16 65.0 ± 7.2
Doxorubicin 0.11 ± 0.01 e 0.89 ± 0.12 1.4 ± 0.02 1.2 ± 0.05
a
Compound concentration required to inhibit metabolic activity by 50%. Data are expressed as the mean ± SD from the concentrationeresponse curves of at least three
experiments.

(pS139), and cleaved PARP (Asp214) antibodies. BrdU incorporation 2.2.3. Induction of apoptosis
into DNA was used as an index of cell proliferation. The induction of Pro-apoptotic activity of 13c was also tested using APC-Annexin
apoptosis was assessed using PE Anti-Cleaved PARP (Asp214) con- V and PI staining. The results of this experiment are shown in Fig. 4.
jugated antibodies recognizing 89 kDa-cleaved fragment of PARP Compound 13c did not significantly influence the number of early
that can serve as a marker of cellular apoptosis. The DNA damage apoptotic cells in the tested cell lines but increased the number of
was evaluated using Alexa Fluor ® 647 Anti-H2AX (pS139) anti- late apoptotic cells (27.8 ± 1.3% and 19 ± 1.5% compared with
bodies, directed against the phosphorylated form of human H2AX control 2.7 ± 0.2% and 6.1 ± 1.0%, in HL-60 and MCF-7 cells,
protein at the pS139 residue that is considered a biomarker for DNA respectively).
double-strand breaks (DSB).
The obtained results demonstrated that 13c significantly 2.2.4. ROS generation
inhibited cell proliferation in both tested cell lines. In MCF-7 cells, One of the possible mechanisms of the cytotoxic activity of
inhibition of cell proliferation was observed in 34%, whereas in HL- compounds with 1,4-naphtalenedione scaffold can be associated
60 in about 91% of cell population (Fig. 2A). Similar tendency was with generation of ROS in cancer cells. To test the ability of 13c to
observed on the generation of DNA damage. As shown in Fig. 2B, induce ROS generation, HL-60 and MCF-7 cells treated for 24 h with
13c induced DNA damage in about 40.7 ± 1.65% and 11.03 ± 0.5% of the tested compound at IC50 concentration, were incubated with
HL-60 and MCF-7 cell populations, respectively. The tested com- CellROX® Green Oxidative Stress Reagent (Molecular Probes, Life
pound increased also the number of cleaved PARP positive Technologies). As shown in Fig. 5 13c increased intracellular ROS
(apoptotic) cells in both tested cell lines. In MCF-7 cells only very levels about 10 times in both tested cell lines, as compared with
slight effect on apoptosis induction was observed. On the contrary, control. Interestingly, the effect induced by this compound was
in HL-60 cells 13c enhanced the number of cleaved PARP positive much stronger than that of redox-cycling agent e menadione, used
cells up to 82.8 ± 5.47%, compared with control 1.2 ± 0.2% (Fig. 2C). as a positive control.
The representative multiparameter flow cytometry analysis of the
effects of 13c on cell proliferation, induction of DNA damage and
2.2.5. Dissipation of mitochondrial membrane potential
apoptosis is shown in Fig. 3.
Mitochondrial dysfunction has been shown to participate in the

Fig. 2. Inhibition of cell proliferation (BrdU test) (A), induction of DNA damage (H2AX test) (B) and induction of apoptosis (PAPR test) (C) by 13c in MCF-7 cells. After 24 h incubation
with tested compounds, BrdU was added and cells were incubated for additional 8 h. The cells were stained with PerCP-Cy™5.5 Mouse Anti-BrdU, Alexa Fluor® 647 Mouse Anti-
H2AX (pS139), PE Mouse Anti-Cleaved PARP (Asp214) antibodies for 20 min at room temperature and analyzed by flow cytometry. Data represent mean ± SEM of three independent
experiments in triplicate. Statistical significance was assessed by the Student t-test. **p < 0.01, ***p < 0.001 was considered as significantly different from control.
56 K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63
K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63 57

induction of apoptosis. Opening of the mitochondrial permeability metabolic nature. This biochemical difference increases cancer cell
transition pore induces depolarization of the transmembrane po- susceptibility to oxidative stress-induced cell death and may be
tential (DJm). The DJm disruption is a critical step for the intrinsic exploited in the design of selective drugs [15,43]. Naphtho [2,3-b]
pathway of apoptosis. To investigate the changes in DJm, HL-60 furan-4,9-diones are highly redox active molecules that are able to
and MCF-7 cells treated for 24 h with 13c at IC50 concentrations, modulate the redox balance in cancer cells. Therefore, naph-
were stained with JC-10. The majority of untreated cells (control) thofurandiones increasing intracellular ROS levels over a critical
were within the population with high DJm. A mitochondrial un- threshold may induce different signaling pathways leading to
coupler e carbonyl cyanide-4-(trifluoromethoxy)phenylhydrazone apoptosis in cancer cells.
(FCCP) was used as a positive control and caused dissipation of
DJm in 67.1 ± 3.5% and 92.5 ± 2.0% of HL-60 and MCF-7 cells,
respectively. Compound 13c caused similar effects in both cell lines. 4. Experimental
In HL-60, 13c decreased DJm in about 44%, while in MCF-7 in 50%
of cell population (Fig. 6). 4.1. Synthesis

4.1.1. General methods


2.2.6. Cell cycle arrest
NMR spectra were recorded on a Bruker DPX 250 at 250.13 MHz
Cell cycle distribution in HL-60 and MCF-7 cells was studied
for 1H, 62.9 MHz for 13C, and 101.3 MHz for 31P NMR using tetra-
after 24 h exposure to 13c at IC50 concentration. DNA staining with
methylsilane as internal and 85% H3PO4 as external standard. 31P
DAPI (1mg/mL) was performed. In HL-60, 13c cased cell cycle arrest
NMR spectra were recorded using broadband proton decoupling. IR
in S phase, whereas in MCF-7 in G0/G1 phase (Fig. 7). A decrease in
spectra were recorded on a Bruker Alpha ATR spectrophotometer.
G2/M was observed in both tested cell lines.
Melting points were determined in open capillaries and are un-
corrected. Column chromatography was performed on Aldrich®
3. Conclusions
silica gel 60 (230e400 mesh). Thin-layer chromatography was
performed with precoated TLC sheets of silica gel 60 F254
We developed novel and efficient synthesis of substituted 3-
(Aldrich®). HRMS spectra were performed on Waters Acquity UPLC/
diethoxyphosphorylnaphtho [2,3-b]furan-4,9-diones 13 and 3-
Q-TOF LC-HRMS apparatus. Reagents and starting materials were
diethoxyphosphorylbenzo [f]indole-4,9-diones 18 which integrate
purchased from commercial vendors and used without further
the natural 1,4-naphtalenedione scaffold and the phosphonate
purification. All organic solvents were dried over appropriate dry-
moiety. The rational to obtain such hybrid molecules was to search
ing agents and distilled prior to use. Standard syringe techniques
for compounds with potentially improved cytotoxic properties.
were used for transferring dry solvents.
Screening of the synthesized 3-diethoxyphosphorylnaphtho
Diethyl (2-oxopropyl)phosphonate (9a), diethyl (2-oxo-2-
[2,3-b]furan-4,9-diones 13aef and 3-hydroxyethoxyphosphoryl-
phenylethyl)phosphonate (9b) and 2,3-dichloro-1,4-
2-(2-chlorophenyl)naphtho [2,3-b]furan-4,9-dione 14, as well as 3-
naphthoquinone (10) were purchased from SigmaeAldrich®.
diethoxyphosphorylbenzo [f]indole-4,9-diones 18aee was per-
formed in vitro against three human cancer cell lines: leukemia HL-
60 and NALM-6 and breast cancer MCF-7. Only naphtho [2,3-b] 4.1.2. General procedure for preparing compounds 13aef
furan-4,9-diones 13aef showed high cytotoxic activity with the A mixture of 2,3-dichloronaphthalene-1,4-dione (10) (1.14 g,
IC50 values below 10 mM. In the naphthodione moiety, the 5.0 mmol) K2CO3 (1.73 g, 12.5 mmol) and corresponding diethyl
replacement of the ortho-fused 3-diethoxyphosphoryl-2- acylmethylphosphonate (9aef) (5.0 mmol) in MeCN (50 mL) was
hydroxypirol by 3-diethoxyphosphoryl-2-arylfuran ring increased stirred at 60  C for 24 h. The reaction mixture was poured into sat.
the IC50 value about 10 fold. The activity of the most potent analog aq. NaCl solution (50 mL). Extraction with CHCl3 (3  50 mL), drying
13c was completely abolished by the hydrolysis of one ester group, (MgSO4) and evaporation of the solvent gave the crude product,
indicating that both ester groups were important for anticancer which was purified by column chromatography [petroleum ether-
activity of the hybrids. The toxicity of the tested compounds against eEtOAc (2:1)].
leukemia and HUVEC cells was similar. However, these compounds
were 4-fold less toxic to MCF-10A as compared with MCF-7 cells.
On the example of compound 13c we have shown that naphtho 4.1.2.1. Diethyl (2-methyl-4,9-dioxo-4,9-dihydronaphtho [2,3-b]
[2,3-b]furan-4,9-diones display a broad range of anticancer activ- furan-3-yl)phosphonate (13a). yellow solid, mp 148e150  C; 1H
ities including inhibition of cell proliferation, generation of DNA NMR (250 MHz, CDCl3) d 8.25e8.12 (m, 2H, HeAr), 7.81e7.69 (m,
damage and induction of apoptosis in two cancer cell lines. We 2H, HeAr), 4.39e4.13 (m, 4H, (CH3CH2O)2P(O)), 2.80 (d,
3
have shown that the underling mechanism of the anticancer ac- JHeP ¼ 2.1 Hz, 3H, CH3), 1.36 (t, 3JHeH ¼ 7.1 Hz, 6H,
tivity of this compound was the generation of intracellular ROS and (CH3CH2O)2P(O)); 13C NMR (63 MHz, CDCl3) d 178.79 (s, C(O)),
imbalance between ROS production and a cell's ability to readily 173.18(s, C(O)), 167.90 (d, 2JCeP ¼ 26.5 Hz, C-2), 151.79 (d,
2
detoxify the reactive intermediates or to repair the resulting JCeP ¼ 11.3 Hz, C-9a), 133.98 (s, CHeAr), 133.73 (s, CHeAr), 133.06
damage. Excess of ROS may induce lipid and protein oxidative (s, CeAr), 131.54 (s, CeAr), 130.41 (d, 2JCeP ¼ 10.1 Hz, C-3a), 127.13
modifications, DNA damage, and disruption of mitochondrial (s, CHeAr), 126.46 (s, CHeAr), 107.42 (d, 1JCeP ¼ 214.7 Hz, C-3),
membrane potential, resulting in cell damage or apoptosis [42]. 62.75 (d, 2JCeP ¼ 5.8 Hz, (CH3CH2O)2P(O)), 16.27 (d, 3JCeP ¼ 6.6 Hz,
It is well documented that most cancer cells contain higher (CH3CH2O)2P(O)), 14.36 (s, CH3); 31P NMR (101 MHz, CDCl3) d 9.69;
levels of endogenous ROS comparing with their non-cancerous IR n(cm1): 2984, 1685, 1360, 1287, 1235, 1020, 976; HRMS m/z,
counterparts, caused largely by the products of their highly calcd [MþH]þ 349.08355, observed 349.08377.

Fig. 3. Multiparameter analysis of cell proliferation, DNA damage and apoptosis in HL-60 and MCF-7 cells treated for 24 h with 13c, at IC50 concentration. Panel A: BrdU vs DAPI
staining profile. BrdU positive cells (proliferating cells) are in the inside frame. Panel B: H2AX (pS139) vs cleaved PARP (Asp214) staining profile. Quadrants Q1þQ2 represent H2AX
(pS139) positive cells (cells with DNA damage), quadrants Q2þQ4 represent cleaved PARP (Asp214) positive cells (apoptotic cells), whereas quadrant Q3 represents H2AX (pS139)
and cleaved PARP (Asp214) negative cells.
58 K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63

Fig. 4. Induction of apoptosis in HL-60 (A) and MCF-7 (B) cells treated for 24 h with 13c, at IC50 concentration. The cells were stained with APC-conjugated Annexin V in a buffer
containing propidium iodide and analyzed by flow cytometry. For each group of cells, the percentage of surviving cells is shown in quadrant Q3. Quadrant Q4 indicates the
percentage of early apoptotic cells (Annexin V-positive cells). The percentage of late apoptotic cells is shown in Q2 (Annexin V- and PI-positive cells). Data represent mean ± SEM of
three independent experiments in triplicate. Statistical significance was assessed by the Student t-test. **p < 0.01, ***p < 0.001 was considered as significantly different from control.

13
C NMR (63 MHz, CDCl3) d 178.79 (s, C(O)), 173.36 (d,
3
JCeP ¼ 0.7 Hz, C(O)), 165.19 (d, 2JCeP ¼ 23.6 Hz, C-2), 152.19 (d,
2
JCeP ¼ 12.5 Hz, C-9a), 134.11 (s, CHeAr), 133.75 (s, CHeAr), 133.32
(s, CeAr), 131.80 (d, 2JCeP ¼ 10.3 Hz, C-3a), 131.63 (s, CeAr), 131.13 (s,
CeAr), 130.15 (s, CHeAr), 128.04 (s, CHeAr), 127.96 (s, CHeAr),
127.28 (s, CHeAr), 126.50 (s, CHeAr), 107.91 (d, 1JCeP ¼ 214.2 Hz, C-
3), 62.95 (d, 2JCeP ¼ 6.0 Hz, (CH3CH2O)2P(O)), 16.05 (d,
3
JCeP ¼ 6.8 Hz, (CH3CH2O)2P(O)). 31P NMR (101 MHz, CDCl3) d 7.18.
IR n(cm1): 2982, 1674, 1360, 1260, 1209, 1010, 936. HRMS m/z,
calcd [MþH]þ 411.09920, observed 411.09915.

4.1.2.3. Diethyl (2-(2-chlorophenyl)-4,9-dioxo-4,9-dihydronaphtho


[2,3-b]furan-3-yl)phosphonate (13c). yellow solid, mp 145e148  C;
1
H NMR (250 MHz, CDCl3) d 8.27e8.19 (m, 2H, HeAr), 7.81e7.74 (m,
Fig. 5. Effect of 13c on intracellular ROS generation in HL-60 and MCF-7 cells. Cells 2H, HeAr), 7.55e7.49 (m, 2H, HeAr), 7.49e7.43 (m, 1H, HeAr),
were treated for 24 h with 13c at IC50 concentration and stained with 5 mM CellROX® 7.42e7.33 (m, 1H, HeAr), 4.30e4.01 (m, 4H, (CH3CH2O)2P(O)), 1.16
Green Reagent in a 37  C, 5% CO2 incubator for 30 min. Menadione (200 mM, 1 h in-
(t, 3JHeH ¼ 7.1 Hz, 6H, (CH3CH2O)2P(O)); 13C NMR (63 MHz, CDCl3)
cubation) was used as a positive control. Data represent mean ± SEM of 3e4 inde-
pendent experiments in triplicate. Statistical significance was assessed using one-way d 178.65 (s, C(O)), 173.32 (d, 3JCeP ¼ 0.9 Hz, C(O)), 161.92 (d,
2
ANOVA and a post-hoc multiple comparison StudenteNewmaneKeuls test. **p < 0.01, JCeP ¼ 22.5 Hz, C-2), 152.54 (d, 2JCeP ¼ 12.7 Hz, C-9a), 134.43 (d,
2
***p < 0.001 was considered as significantly different from control. JCeP ¼ 0.9 Hz, C-3a), 134.09 (s, CCl), 133.74 (s, CHeAr), 133.00 (s,
CHeAr), 132.38 (s, CHeAr), 131.85 (s, CeAr), 131.53 (s, CeAr), 130.54
(s, CHeAr), 130.38 (s, CHeAr), 129.32 (s, CeAr), 128.20 (s, CHeAr),
4.1.2.2. Diethyl (4,9-dioxo-2-phenyl-4,9-dihydronaphtho [2,3-b]
127.16 (s, CHeAr), 126.49 (s, CHeAr), 126.16 (s, CHeAr), 111.40 (d,
furan-3-yl)phosphonate (13b). orange oil; 1H NMR (250 MHz, 1
JCeP ¼ 215.5 Hz, C-3), 62.70 (d, 2JCeP ¼ 6.1 Hz, (CH3CH2O)2P(O)),
CDCl3) d 8.29e8.10 (m, 2H, HeAr), 7.89e7.82 (m, 2H, HeAr),
15.88 (d, 3JCeP ¼ 7.3 Hz, (CH3CH2O)2P(O)); 31P NMR (101 MHz,
7.78e7.73 (m, 2H, HeAr), 7.56e7.43 (m, 3H, HeAr), 4.28e4.07 (m,
CDCl3) d 7.50; IR n(cm1): 2984, 1671, 1363, 1256, 1210, 1011, 940;
4H, (CH3CH2O)2P(O)), 1.19 (t, 3JHeH ¼ 7.0 Hz, 6H, (CH3CH2O)2P(O)).
HRMS m/z, calcd [MþH]þ 445.06023, observed 445.06033.
K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63 59

Fig. 6. Effect of 13c on dissipation of mitochondrial membrane potential (DJm) in HL-60 (A) and MCF-7 (B) cells. Cells were treated for 24 h with 13c at IC50 concentration and
stained with JC-10 (5 mL/mL) for 45 min in 37  C, 5% CO2. Changes in the fluoresence were analyzed by flow cytometry. FCCP (30 mM) was used as a positive control. Data represent
mean ± SEM of three independent experiments in triplicate. Statistical significance was assessed using one-way ANOVA and a post-hoc multiple comparison Stu-
denteNewmaneKeuls test. **p < 0.01, ***p < 0.001 was considered as significantly different from control.

4 .1. 2 . 4 . D i e t h y l ( 2 - ( 2 , 4 - d i c h l o r o p h e n y l ) - 4 , 9 - d i o x o - 4 , 9 - CHeAr), 130.20 (s, CHeAr), 129.30 (s, CHeAr), 127.18 (s, CeAr),
dihydronaphtho [2,3-b]furan-3-yl)phosphonate (13d). yellow solid, 126.74 (s, CHeAr), 126.63 (s, CHeAr), 126.52 (s, CHeAr), 111.83 (d,
mp 122e124  C; 1H NMR (250 MHz, CDCl3) d 8.28e8.17 (m, 2H, 1
JCeP ¼ 214.4 Hz, C-3), 62.79 (d, 2JCeP ¼ 6.1 Hz, (CH3CH2O)2P(O)),
HeAr), 7.82e7.75 (m, 2H, HeAr), 7.54 (d, 4JHeH ¼ 1.9 Hz, 1H, HeAr), 15.97 (d, 3JCeP ¼ 6.7 Hz, (CH3CH2O)2P(O)). 31P NMR (101 MHz,
7.46 (d, 3JHeH ¼ 8.3 Hz, 1H, HeAr), 7.37 (dd, 3JHeH ¼ 8.3 Hz, CDCl3) d 7.18. IR n(cm1): 2986, 1674, 1357, 1238, 1209, 939. HRMS
4
JHeH ¼ 1.9 Hz, 1H, HeAr), 4.33e4.04 (m, 4H, (CH3CH2O)2P(O)), 1.21 m/z, calcd [MþH]þ 479.02126, observed 479.02118.
(t, 3JHeH ¼ 7.1 Hz, 6H, (CH3CH2O)2P(O)). 13C NMR (63 MHz, CDCl3)
d 178.51 (s, C(O)), 173.25 (d, 3JCeP ¼ 0.7 Hz, C(O)), 160.98 (d, 4 .1. 2 . 5 . D i e t hyl ( 2 - ( 2 - ch l o r o p y r i d i n - 3 - yl ) - 4 , 9 - d i o x o - 4 , 9 -
2
JCeP ¼ 22.3 Hz, C-2), 152.69 (d, 2JCeP ¼ 12.2 Hz, C-9a), 137.44 (s, dihydronaphtho [2,3-b]furan-3-yl)phosphonate (13e). yellow solid,
CCl), 135.35 (d, 2JCeP ¼ 0.9 Hz, C-3a), 134.15 (s, CHeAr), 133.79 (s, mp 204e206  C; 1H NMR (250 MHz, CDCl3) d 8.56 (dd,
CHeAr), 133.19 (s, CCl), 132.96 (s, CeAr), 131.48 (s, CeAr), 130.36 (s, 3
JHeH ¼ 4.9 Hz, 4JHeH ¼ 1.9 Hz, 1H, HeAr), 8.28e8.19 (m, 2H, HeAr),
60 K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63

NMR (63 MHz, DMSO-d6) d 178.43 (s, C(O)), 172.99 (s, C(O)), 161.01
(d, 2JC-P ¼ 21.6 Hz, C-2), 152.71 (d, 2JC-P ¼ 12.1 Hz, C-9a), 134.43 (s,
CHeAr), 134.20 (s, CHeAr), 133.37 (s, CCl), 132.76 (s, CHeAr), 132.76
(s, CHeAr), 132.55 (s, CeAr), 131.44 (s, CeAr), 129.56 (d, 2JC-
P ¼ 10.7 Hz, C-3a), 129.34 (s, CeAr), 128.13 (s, CHeAr), 126.96 (s,
CHeAr), 126.70 (s, CHeAr), 126.13 (s, CHeAr), 111,02 (d,
1
JCeP ¼ 212.6 Hz, C-3), 62.15 (d, 2JC-P ¼ 5.4 Hz, (CH3CH2O)P(O)),
15.78 (d, 3JC-P ¼ 6.8 Hz, (CH3CH2O)P(O)).31P NMR (101 MHz, DMSO-
d6) d 2.43; IR n(cm1): 2923, 1676, 1365, 1209, 1038, 998, 963, 937;
HRMS m/z, calcd [MþH]þ 417.02893, observed 417.02948.

4.1.4. Ethyl 2-(3-chloro-1,4-dioxo-1,4-dihydronaphthalen-2-yl)-2-


(diethoxyphosphoryl)acetate (14)
Fig. 7. Effect of 13c on cell cycle distribution in HL-60 and MCF-7 cells. Cells were To a stirred solution of 2,3-dichloronaphthalene-1,4-dione (10)
treated for 24 h with 13c at IC50 concentration. The percentage of cells at each stage of
the cycle was analyzed by flow cytometry, following DNA staining with DAPI and
(4.5 g, 20.0 mmol) and ethyl 2-(diethoxyphosphoryl)acetate (15)
compared with untreated cells regarded as control, C. The results are representative of (4.0 mL, 20.0 mmol) in THF (50 mL) NaH (1.0 g, 42 mmol) was added
three independent experiments. Statistical significance was assessed by the Student t- at 0  C, and the resulting mixture was stirred at room temperature
test. *p < 0.05, **p < 0.01 was considered as significantly different from control. for 1 week. Cold (0  C) water (100 mL), 1 M HClaq (20 mL) and DCM
(150 mL) were added. The organic layer was separated and the
aqueous layer was extracted with DCM (2  50 mL). The combined
7.87 (dd, 3JHeH ¼ 7.6 Hz, 4JHeH ¼ 1.9 Hz, 1H, HeAr), 7.82e7.77 (m,
organic extracts were washed with brine, dried over MgSO4,
2H, HeAr), 7.39 (dd, 3JHeH ¼ 7.6 Hz, 3JHeH ¼ 4.9 Hz, 1H, HeAr),
filtered and concentrated. The crude product was purified by flash
4.30e4.09 (m, 4H, (CH3CH2O)2P(O)), 1.19 (t, 3JHeH ¼ 7.1 Hz, 6H,
chromatography (chloroform) to give pure product as yellow solid,
(CH3CH2O)2P(O)). 13C NMR (63 MHz, CDCl3) d 178.69 (s, C(O)),
mp 70e71  C; 1H NMR (250 MHz, CDCl3) d 8.24e8.08 (m, 2H,
173.51 (d, 3JCeP ¼ 1.8 Hz, C(O)), 160.25 (d, 2JCeP ¼ 22.1 Hz, C-2),
HeAr), 7.90e7.70 (m, 2H, HeAr), 5.02 (d, J ¼ 27.3 Hz, 1H, CHP),
153.07 (d, 2JCeP ¼ 12.1 Hz, C-9a), 151.44 (s, CHeAr), 150.97 (d,
3 4.37e4.09 (m, 6H, (CH3CH2O)2P(O), (CH3CH2O)C(O)), 1.29 (t, 3JH-
JCeP ¼ 1.1 Hz, CeAr), 141.33 (s, CCl), 134.45 (s, CHeAr), 134.08 (s, 3
H ¼ 7.1 Hz, 6H, (CH3CH2O)2P(O)), 1.25 (t, JH-H ¼ 7.1 Hz, 3H,
CHeAr), 133.18 (s, CeAr), 131.73 (s, CeAr), 130.46 (d, 2JCeP ¼ 9.9 Hz,
(CH3CH2O)C(O)). 13C NMR (63 MHz, CDCl3) d 180.15 (d, 2JC-
C-3a), 127.46 (s, CHeAr), 126.82 (s, CHeAr), 125.44 (s, CeAr), 121.74 3
P ¼ 2.4 Hz, C(O)), 176.81 (d, JC-P ¼ 1.8 Hz, C(O)), 165.15 (s, C(O)),
(s, CHeAr), 112.52 (d, 1JCeP ¼ 213.9 Hz, C-3), 63.10 (d, 2JCeP ¼ 6.0 Hz,
145.13 (d, 2JC-P ¼ 9.7 Hz, CCl), 140.06 (d, 2JC-P ¼ 8.4 Hz, C-2), 134.23
(CH3CH2O)2P(O)), 16.17 (d, 3JCeP ¼ 6.7 Hz, (CH3CH2O)2P(O)). 31P
(s, CH-Ar), 133.99 (s, CH-Ar), 130.87 (s, CeAr), 130.73 (s, CeAr),
NMR (101 MHz, CDCl3) d 6.72. IR n(cm1): 2983, 1676, 1383, 1261,
127.00 (s, CH-Ar), 126.94 (s, CH-Ar), 63.33 (d, 3JC-P ¼ 6.6 Hz,
1214, 1025, 938. HRMS m/z, calcd [MþH]þ 446.05548, observed
(CH3CH2O)C(O)), 63.00 (d, 2JC-P ¼ 6.7 Hz, (CH3CH2O)2P(O)), 46.23 (d,
446.05560. 1
JC-P ¼ 143.4 Hz, CHP), 15.99 (d, 3JC-P ¼ 4.3 Hz, (CH3CH2O)2P(O)),
13.64. 31P NMR (101 MHz, CDCl3) d 16.09. IR n(cm1): 2986, 2898,
4.1.2.6. Diethyl (2-(2,6-dichloro-5-fluoropyridin-3-yl)-4,9-dioxo-4,9-
1736, 1667, 1262, 1248, 962, 877, 720, 570; HRMS m/z, calcd
dihydronaphtho [2,3-b]furan-3-yl)phosphonate (13f). yellow solid,
[MþH]þ 415.07079, observed 415.07031.
mp 103e105  C; 1H NMR (250 MHz, CDCl3) d 8.28e8.21 (m, 2H,
HeAr), 7.85e7.78 (m, 2H, HeAr), 7.70 (d, 3JHeF ¼ 7.0 Hz, 1H, HeAr),
4.35e4.13 (m, 4H, (CH3CH2O)2P(O)), 1.27 (t, 3JH-H ¼ 7.1 Hz, 6H, 4.1.5. General procedure for preparing compounds 18aee
(CH3CH2O)2P(O)). 13C NMR (63 MHz, CDCl3) d 178.32 (s, C(O)), To a stirred solution of phosphonate (16) (0.83 g, 2 mmol) in
173.24 (s, C(O)), 157.81 (d, 2JC-P ¼ 22.3 Hz, C-2), 153.22 (d, 2JC- MeOH (10 mL) amine (30 mmol, 21.4 mL, C ¼ 1.4 M methylamine
1 4 solution in MeOH or 8 mmol for n-propylamine, c-propylamine, n-
P ¼ 11.9 Hz, C-9a), 153.07 (d, JC-F ¼ 263.1 Hz, CF), 144.17 (d, JC-
3 hexylamine, 2-chlorobenzylamine) was added, and the resulting
P ¼ 3.2 Hz, CCl), 140.00 (d, JC-P ¼ 20.9 Hz, CeAr), 134.46 (s, CH-Ar),
134.10 (s, CH-Ar), 132.96 (s, CeAr), 131.51 (s, CeAr), 129.99 (d, 2JC- mixture was stirred at room temperature for 1 day. Then, solvent
2 was evaporated, and DCM (15 mL), 1 M HClaq (20 mL) were added.
P ¼ 9.3 Hz, C-3a), 129.42 (d, JC-P ¼ 21.5 Hz, CCl)), 127.38 (s, CH-Ar),
126.75 (s, CH-Ar), 125.24 (d, 2JC-P ¼ 3.6 Hz, CeAr), 113.21 (d, 1JC- The organic layer was separated and the aqueous layer was
2 extracted with DCM (2  10 mL). The combined organic extracts
P ¼ 212.5 Hz, C-3), 63.24 (d, JC-P ¼ 6.0 Hz, (CH3CH2O)2P(O)), 16.13
(d, JC-P ¼ 6.2 Hz, (CH3CH2O)2P(O)). 31P NMR (101 MHz, CDCl3)
3 were washed with brine, dried over MgSO4, filtered and concen-
d 5.71. IR n(cm1): 2989, 1679, 1394, 1283, 1207, 1018, 983. HRMS m/ trated. The crude product was purified by flash chromatography
z, calcd [MþH]þ 498.00708, observed 498.00714. (chloroform).

4.1.3. Ethyl hydrogen (2-(2-chlorophenyl)-4,9-dioxo-4,9- 4.1.5.1. Diethyl (2-hydroxy-1-methyl-4,9-dioxo-4,9-dihydro-1H-


dihydronaphtho [2,3-b]furan-3-yl)phosphonate (14) benzo[f]indol-3-yl)phosphonate (18a). red solid, mp 74e75  C; 1H
To a solution of diethyl phosphonate 13c (0.44 g, 1 mmol) in NMR (250 MHz, CDCl3) d 8.20e7.97 (m, 2H, HeAr), 7.76e7.57 (m,
butanone (10 mL) lithium bromide (0.11 g, 1.25 mmol) was added 2H, HeAr), 4.34e3.98 (m, 4H, (CH3CH2O)2P(O)), 3.91 (s, 3H, CH3),
and the mixture was stirred at 80  C for 5 h. The solvent was 1.33 (t, 3JH-H ¼ 7.0 Hz, 6H, (CH3CH2O)2P(O)). 13C NMR (63 MHz,
evaporated and 1 M HClaq (10 mL) was added to the solid residue. CDCl3) d 179.73 (s, C(O)), 175.22 (s, C(O)), 160.58 (d, 2JC-P ¼ 18.3 Hz,
Extraction with CHCl3 (3  30 mL), drying (MgSO4) and evaporation C-2), 133.29 (s, CeAr), 133.19 (s, CH-Ar), 132.84 (s, CH-Ar), 132.79 (s,
of the solvent gave the crude product. The solid was suspended in CeAr), 127.18 (d, 2JC-P ¼ 8.2 Hz, C-9a), 126.54 (d, 2JC-P ¼ 11.9 Hz, C-
Et2O (10 mL), stirred, filtered and dried to give 0.39 g (93%) of 14 as 3a), 126.30 (s, CH-Ar), 126.00 (s, CH-Ar), 81.76 (d, 1JC-P ¼ 215.7 Hz, C-
yellow solid, mp 208e209  C; 1H NMR (250 MHz, DMSO-d6) 3), 63.44 (d, 2JC-P ¼ 5.4 Hz, (CH3CH2O)2P(O)), 30.83 (s, CH3), 16.27 (d,
3
d 8.19e8.11 (m, 2H, HeAr), 7.99e7.92 (m, 2H, HeAr), 7.74e7.59 (m, JC-P ¼ 7.0 Hz, (CH3CH2O)2P(O)). 31P NMR (101 MHz, CDCl3) d 18.24.
3H, HeAr), 7.60e7.49 (m, 1H, HeAr), 3.86 (p, 3JH-H ¼ 2JH-P ¼ 7.2 Hz, IR n(cm1): 2983, 1652, 1552, 1289, 1021, 966, 854, 552. HRMS m/z,
2H, (CH3CH2O)P(O)), 1.01 (t, 3JH-H ¼ 7.0 Hz, 3H, (CH3CH2O)2P(O)). 13C calcd [MþH]þ 364.09445, observed 364.09448.
K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63 61

4.1.5.2. Diethyl (2-hydroxy-4,9-dioxo-1-propyl-4,9-dihydro-1H- (101 MHz, CDCl3) d 19.66. IR n(cm1): 2959, 2928, 2859, 1732, 1649,
benzo[f]indol-3-yl)phosphonate (18b). red solid, mp 81e82  C; 1H 1509, 1392, 1232, 1166, 1022, 963, 793, 622. HRMS m/z, calcd
NMR (250 MHz, CDCl3) d 8.11e8.06 (m, 2H, HeAr), 7.71e7.58 (m, [MþH]þ 474.08678, observed 474.08612.
2H, HeAr), 4.34 (t, 3JH-H ¼ 7.4 Hz, 2H, CH2CH2N), 4.29e4.04 (m, 4H,
(CH3CH2O)2P(O)), 1.83 (h, 3JH-H ¼ 7.4 Hz, 2H, CH2CH2N), 1.33 (t, 3JH- 4.2. Biological activity
3
H ¼ 7.1 Hz, 6H, (CH3CH2O)2P(O)), 0.97 (t, JH-H ¼ 7.4 Hz, 3H,
CH3CH2).13C NMR (63 MHz, CDCl3) d 179.78 (s, C(O)), 174.84 (s, 4.2.1. Cell culture and treatment
C(O)), 160.52 (d, 2JC-P ¼ 19.7 Hz, C-2), 133.42 (s, CeAr), 133.16 (s, Two human leukemia cell lines, promyelocytic leukemia (HL-
CeAr), 132.80 (s, CH-Ar), 132.75 (s, CH-Ar), 127.47 (d, 2JC-P ¼ 8.4 Hz, 60) and lymphoblastic leukemia (NALM-6), and a solid tumor-
C-9a), 126.21 (d, 2JC-P ¼ 12.1 Hz, C-3a), 126.24 (s, CH-Ar), 126.03 (s, derived human breast adenocarcinoma cell line (MCF-7) were
CH-Ar), 81.73 (d, 1JC-P ¼ 216.1 Hz, C-3), 63.42 (d, 2JC-P ¼ 5.5 Hz, purchased from the European Collection of Cell Cultures (ECACC).
(CH3CH2O)2P(O)), 45.48 (s, CH2N), 23.05 (s, CH3CH2), 16.24 (d, 3JC- Leukemia cells were cultured in RPMI 1640 plus GlutaMax I me-
31
P ¼ 7.0 Hz, (CH3CH2O)2P(O)), 11.02 (s, CH3CH2). P NMR (101 MHz, dium (Invitrogen, Grand Island, NY, USA), supplemented with 10%
1
CDCl3) d 18.45. IR n(cm ): 2974, 2932, 2874, 1660, 1589, 1387, 981, heat-inactivated FBS (Biological Industries, Beit-Haemek, Israel)
962, 751, 703, 575, 506. HRMS m/z, calcd [MþH]þ 392.12575, and antibiotics (100 mg/mL streptomycin and 100 U/mL penicillin)
observed 392.12552. (SigmaeAldrich, St. Louis, MO, USA). MCF-7 cells were cultured in
Minimum Essential Medium Eagle (MEME, SigmaeAldrich, St.
4.1.5.3. Diethyl (1-cyclopropyl-2-hydroxy-4,9-dioxo-4,9-dihydro-1H- Louis, MO, USA) supplemented with 10% heat-inactivated fetal
benzo[f]indol-3-yl)phosphonate (18c). red solid, mp 90e91  C; 1H bovine serum (FBS) (Biological Industries, Beit-Haemek, Israel),
NMR (250 MHz, CDCl3) d 11.95 (bs, 1H, OH), 8.31e8.10 (m, 2H, 2 mM glutamine, Men Non-essential amino acid solution and an-
HeAr), 7.88e7.66 (m, 2H, HeAr), 4.47e4.10 (m, 4H, tibiotics (100 mg/mL streptomycin and 100 U/mL penicillin), all from
(CH3CH2O)2P(O)), 3.47 (p, 3JH-H ¼ 3.5 Hz, 1H, CHN), 1.44 (t, 3JH- SigmaeAldrich (SigmaeAldrich, St. Louis, MO, USA). Normal hu-
H ¼ 7.1 Hz, 6H, (CH3CH2O)2P(O)), 1.38e1.31 (m, 2H, CH2), 1.25e1.19 man mammary gland/breast cell line (MCF10A) and normal human
(m, 2H, CH2). 13C NMR (63 MHz, CDCl3) d 179.59 (s, C(O)), 173.88 (s, umbilical vein endothelial cells (HUVECs) were purchased from the
C(O)), 161.63 (d, 2JC-P ¼ 20.3 Hz, C-2), 133.32 (s, CeAr), 133.07 (s, American Type Culture Collection (ATCC). MCF-10A cells were
CeAr), 132.47 (s, CH-Ar), 132.41 (s, CH-Ar), 127.59 (d, 2JC-P ¼ 11.6 Hz, cultured using MEGM Mammary Epithelial BulletKit, while HUVECs
C-9a), 127.29 (d, 2JC-P ¼ 8.2 Hz, C-3a), 125.99 (s, CH-Ar), 125.94 (s, using EGM-2 Endothelial Medium BulletKit, both purchased from
CH-Ar), 81.32 (d, 1JC-P ¼ 215.3 Hz, C-3), 63.26 (d, 2JC-P ¼ 5.6 Hz, Lonza (Lonza, Walkersville, MD, USA). Cells were maintained at
(CH3CH2O)2P(O)), 26.95 (s, CHN), 16.07 (d, 3JC-P ¼ 6.8 Hz, 37  C in a 5% CO2 atmosphere and were grown until 80% confluent.
(CH3CH2O)2P(O)), 8.37 (s, CH2). 31P NMR (101 MHz, CDCl3) d 18.41. For all experiments tested compounds were dissolved in DMSO
IR n(cm1): 2982, 2929, 2906, 1719, 1507, 1248, 1017, 965, 789, 680, (SigmaeAldrich, Louis, MO, USA) and further diluted in culture
578, 560. HRMS m/z, calcd [MþH]þ 390.11010, observed 390.11017. medium to obtain less than 0.1% DMSO concentration. In each
experiment controls without and with 0.1% DMSO were performed.
4.1.5.4. Diethyl (1-hexyl-2-hydroxy-4,9-dioxo-4,9-dihydro-1H-benzo DMSO in this concentration had no effects on the observed
[f]indol-3-yl)phosphonate (18d). red solid, mp 67e68  C; 1H NMR parameters.
(250 MHz, CDCl3) d 11.95 (bs, 1H, OH), 8.14e8.07 (m, 2H, HeAr),
7.69e7.63 (m, 2H, HeAr), 4.38 (t, 3JH-H ¼ 7.4 Hz, 2H, CH2CH2N), 4.2.2. Metabolic activity assay (MTT)
4.30e4.08 (m, 4H, (CH3CH2O)2P(O)), 1.88e1.74 (m, 2H, CH2), The MTT (3-(4,5-dimethyldiazol-2-yl)-2,5 diphenyl tetrazolium
1.46e1.22 (m, 12H, (CH3CH2O)2P(O), 3  CH2), 0.88 (t, 3JH-H ¼ 6.7 Hz, bromide) assay, which measures activity of cellular de-
3H, CH3). 13C NMR (63 MHz, CDCl3) d 179.76 (s, C(O)), 174.94 (s, hydrogenases, was performed according to a known procedure
C(O)), 160.15 (d, 2JC-P ¼ 20.1 Hz, C-2), 133.37 (s, CeAr), 133.21 (s, CH- [41]. Exponentially growing cells were seeded on 96-well plates
Ar), 132.82 (s, CH-Ar), 132.73 (s, CeAr), 127.38 (d, 2JC-P ¼ 11.6 Hz, C- (Nunc, Roskilde, Denmark) in 100 mL and left to growth for 24 h.
9a), 126.28 (s, CH-Ar), 126.20 (d, 2JC-P ¼ 7.2 Hz, C-3a), 126.09 (s, CH- Subsequently, various concentrations of the tested compounds
Ar), 81.89 (d, 1JC-P ¼ 215.4 Hz, C-3), 63.47 (d, 2JC-P ¼ 5.5 Hz, (freshly prepared in DMSO and diluted with complete culture
(CH3CH2O)2P(O)), 44.27 (s, CH2N), 31.35 (s, CH2), 29.73 (s, CH2), medium) were added in 100 mL of culture medium to obtain the
26.29 (s, CH2), 22.55 (s, CH2), 16.27 (d, 3JC-P ¼ 6.7 Hz, concentration range from 107 to 103 M and the plates were
(CH3CH2O)2P(O)), 13.99 (s, CH3). 31P NMR (101 MHz, CDCl3) d 18.45. incubated for 24 or 48 h. Afterwards, MTT (5 mg/mL in PBS) was
IR n(cm1): 3060, 2976, 2925, 1643, 1591, 1541, 1235, 1019, 955, 787, added and incubation was continued for 2 h. The metabolically
724, 622, 576. HRMS m/z, calcd [MþH]þ 434.17270, observed active cells reduced MTT to blue formazan crystals. Then, the MTT-
434.17249. containing medium was carefully aspirated and 100 mL DMSO was
added to dissolve the crystals. The absorbance of the blue formazan
4.1.5.5. Diethyl (1-(2-chlorobenzyl)-2-hydroxy-4,9-dioxo-4,9- product was measured at 560 nm using an automated plate reader
dihydro-1H-benzo[f]indol-3-yl)phosphonate (18e). red solid, mp (iMark Bio-Rad, Hercules, CA, USA) and compared with control
86e88  C; 1H NMR (250 MHz, CDCl3) d 8.16e7.82 (m, 2H, HeAr), (untreated cells). The IC50 values were calculated from concen-
7.63e7.49 (m, 2H, HeAr), 7.32e7.15 (m, 1H, HeAr), 7.07e6.80 (m, trationeresponse curves.
2H, HeAr), 6.73e6.51 (m, 1H, HeAr), 5.50 (s, 2H, CH2), 4.21e3.91
(m, 4H, (CH3CH2O)2P(O)), 1.16 (t, 3JH-H ¼ 6.6 Hz, 6H, 4.2.3. Analysis of cell proliferation, apoptosis and DNA damage by
(CH3CH2O)2P(O)). 13C NMR (63 MHz, CDCl3) d 179.83 (s, C(O)), flow cytometry
174.97 (s, C(O)), 160.72 (d, 2JC-P ¼ 20.0 Hz, C-2),133.78 (s, CCl), 133.39 Cell proliferation, DNA damage and apoptotic cell death were
(s, CeAr), 133.24 (s, CH-Ar), 133.05 (s, CH-Ar), 132.80 (s, CeAr), determined using the “Apoptosis, DNA Damage, and Cell Prolifer-
132.49 (s, CeAr), 129.76 (s, CH-Ar), 128.76 (s, CH-Ar), 127.72 (d, 2JC- ation Kit” (BD Bioscience, San Jose, CA, USA), according to the
P ¼ 8.0 Hz, C-3a), 127.14 (s, CH-Ar), 126.46 (s, CH-Ar), 126.29 (s, CH- manufacturer guidelines. The test kit includes all the necessary
Ar), 126.26 (s, CH-Ar), 126.25 (d, 2JC-P ¼ 11.8 Hz, C-9a), 82.34 (d, 1JC- components, such as: BD Cytofix/Cytoperm™ Fixation/Per-
2
P ¼ 215.7 Hz, C-3), 63.70 (d, JC-P ¼ 5.6 Hz, (CH3CH2O)2P(O)), 45.25 meabilization Solution; BD Perm/Wash™ Buffer; BD Cytofix/Cyto-
(s, CH2), 16.35 (d, 3JC-P ¼ 6.6 Hz, (CH3CH2O)2P(O)). 31P NMR perm™ Plus Permeabilization Buffer, DAPI, (Bromodeoxyuridine)
62 K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63

BrdU, DNase and antibodies (PerCP-Cy™5.5 Mouse Anti-BrdU; determined using the Cell Meter™ JC-10 Mitochondrial Membrane
Alexa Fluor® 647 Mouse Anti-H2AX (pS139); PE Mouse Anti- Potential Assay Kit (AAT, Bioquest Inc., Sunnyvale, CA, USA), ac-
Cleaved PARP (Asp214)). Briefly, cells were seeded on 25 cm2 cell cording to the manufacturer guidelines. JC-10 dye enters selectively
culture flask at a density 8.0  104/mL in 10 mL of culture medium into mitochondria and reversibly changes color from green to or-
(MCF-7 cells) or on 6-well plates at a density 2.0  105/mL in 2 mL ange as membrane potential increases. Briefly, cells were seeded on
of growth medium (HL-60 cells) and left to grow for 18e24 h. Af- 6-well plates (4.5  105 cells/well for MCF-7 cells and
terwards, compound 13c diluted in medium was added to the cells 4.0  105 cells/well for HL-60 cells) and left to growth for 18e24 h.
at IC50 concentration. The cells incubated without tested com- Then, cells were treated with tested compound 13c, as previously
pounds were used as control. After 24 h of incubation, cells were described. After 24 h of treatment MCF-7 cells were harvested by
treated with BrdU (final concentration of 10 mM) for additional 8 h. trypsinization and centrifuged (200  g, 5 min). HL-60 cells were
Then, MCF-7 cells were harvested by trypsinization and collected by centrifugation (200  g, 5 min). Afterwards, cells were
centrifuged (200  g, 5 min). HL-60 cells were collected by washed once with PBS, stained with JC-10 at 37  C for 45 min in
centrifugation (200  g, 5 min). The total number of cells was dark and analyzed by flow cytometry (Becton Dickinson Canto II). A
determined. Cells were fixed, permeabilized according to the mitochondrial uncoupler - carbonyl cyanide-4-(trifluoromethoxy)
manufacturer's protocol and treated with DNase (300 mg/mL in phenylhydrazone (FCCP, SigmaeAldrich, Louis, MO, USA) was
DPBS) for 1 h at 37  C in order to expose BrdU epitopes. Following used as a positive control. Changes in DJm were visualized and
this treatment, cells were simultaneously stained with quantified by constructing a dot-plot using the BD FACSDiva
fluorochrome-labeled anti-BrdU, H2AX (pS139), and cleaved PARP software.
(Asp214) antibodies for 20 min at room temperature. After
washing, DNA staining for cell cycle analysis using DAPI solution 4.2.7. Statistical analysis
(1mg/mL of staining buffer) was performed. Cells were re- Statistical analysis was performed using Prism 4.0 (GraphPad
suspended in a staining buffer and analyzed by flow cytomtery Software Inc., San Diego, CA, USA). Results from at least three in-
(Becton Dickinson Canto II). The data was visualized and quantified dependent experiments in triplicate are expressed as mean ± SD or
by constructing a dot-plot using the BD FACSDiva software. SEM. Statistical significance was analyzed using either Student's t
test (for two-group comparison) or one-way ANOVA followed by a
4.2.4. Analysis of apoptosis by flow cytometry using APC-Annexin post-hoc multiple comparison StudenteNewmaneKeuls test (for
Vand propidium iodide (PI) double staining multiple-group comparison). p < 0.05 was considered statistically
Apoptotic cell death was determined using APC-Annexin V significant.
Apoptosis Detection Kit (BD Bioscience, San Jose, CA, USA). Briefly,
the cells were seeded on 6-well plates (4.5  105 cells/well for MCF- Notes
7 cells and 4.0  105 cells/well for HL-60 cells) and left to growth for
18e24 h. Subsequently, tested compound 13c diluted in medium The authors declare no competing financial interest.
was added to the cells at IC50 concentration. The cells incubated
without tested compounds were used as a control. After 24 h of
Acknowledgments
incubation MCF-7 cells were harvested by trypsinization and
centrifuged (200g, 5 min). HL-60 cells were collected by centri-
Financial support from Lodz University of Technology Statutory
fugation (200  g, 5 min). Cells were stained with annexin V and
founds I-18/13/DzS and from Medical University of Lodz (No 502-
propidium iodide (PI) according to the manufacturer guidelines
03/1-156-02/502-14-191 to KG and No 503/1-156-02/503-11-002).
(15 min, at room temperature). Apoptosis of cells was analyzed by
flow cytometry (Becton Dickinson Canto II). Early and late apoptosis
Appendix A. Supplementary data
was visualized and quantified by constructing a dot-plot using the
BD FACSDiva software.
Supplementary data related to this article can be found at http://
dx.doi.org/10.1016/j.ejmech.2016.05.002.
4.2.5. Analysis of ROS induction by flow cytometry
The change of intracellular ROS levels was determined by flow
cytometry, based on the staining with cell-permeable CellROX® References
Green Oxidative Stress Reagent (Molecular Probes, Life Technolo-
[1] S.R. de Paiva, M.R. Figueiredo, T.V. Araga~o, M.A. Kaplan, Antimicrobial activity
gies) which is a fluorogenic probe designed to reliably measure ROS in vitro of plumbagin isolated from Plumbago species, Mem. Inst. Oswaldo
in the live cells. CellROX® Green Reagent is non-fluorescent or very Cruz 98 (2003) 959e961.
weakly fluorescent while in a reduced state and upon oxidation it [2] C.C. Wang, Y.M. Chiang, S.C. Sung, Y.L. Hsu, J.K. Chang, P.L. Kuo, Plumbagin
induces cell cycle arrest and apoptosis through reactive oxygen species/c-Jun
exhibits a strong fluorogenic signal localized in the mainly in nu- N-terminal kinase pathways in human melanoma A375.S2 cells, Cancer Lett.
cleus and mitochondria. Briefly, cells were seeded on 6-well plates 259 (2008) 82e98.
(4.5  105 cells/well for MCF-7 cells and 4.0  105 cells/well for HL- n, G. Burton, J.M. Padro
[3] E.L. Bonifazi, C. Ríos-Luci, L.G. Leo  n, R.I. Misico, Anti-
proliferative activity of synthetic naphthoquinones related to lapachol. First
60 cells) and left to growth for 18e24 h. Cells were treated with synthesis of 5-hydroxylapachol, Bioorg. Med. Chem. 18 (2010) 2621e2630.
tested compound 13c as described above. After 24 h of treatment, [4] S. Sagar, L. Esau, B. Moosa, N.M. Khashab, V.B. Bajic, M. Kaur, Cytotoxicity and
cells were incubated with 5 mM CellROX® Green Reagent at 37  C for apoptosis induced by a plumbagin derivative in estrogen positive MCF-7
breast cancer cells, Anticancer Agents Med. Chem. 14 (2014) 170e180.
30 min in the dark. Then, MCF-7 cells were harvested by trypsini- [5] P.R. Duchowicz, D.O. Bennardi, D.E. Bacelo, E.L. Bonifazi, C. Rios-Luci,
zation and centrifuged (200  g, 5 min). HL-60 cells were collected J.M. Padron, G. Burton, R.I. Misico, QSAR on antiproliferative naphthoquinones
by centrifugation (200  g, 5 min). Cells were washed twice with based on a conformation-independent approach, Eur. J. Med. Chem. 77 (2014)
176e184.
PBS, re-suspended in PBS and analyzed by flow cytometry (Becton
[6] S.L. de Castro, F.S. Emery, E.N. da Silva Júnior, Synthesis of quinoidal mole-
Dickinson Canto II). A redox-cycling agent - menadione (Sigma- cules: strategies towards bioactive compounds with an emphasis on lapa-
eAldrich, Louis, MO, USA) was used as a positive control. chones, Eur. J. Med. Chem. 69 (2013) 678e700.
[7] E.H. da Cruz, C.M. Hussene, G.G. Dias, E.B. Diogo, I.M. de Melo, B.L. Rodrigues,
M.G. da Silva, W.O. Valença, C.A. Camara, R.N. de Oliveira, Y.G. de Paiva,
4.2.6. Assessment of mitochondrial membrane potential M.O. Goulart, B.C. Cavalcanti, C. Pessoa, E.N. da Silva Júnior, 1,2,3-triazole-,
Changes in mitochondrial membrane potential (DJm) were arylamino- and thio-substituted 1,4-naphthoquinones: potent antitumor
K. Gach et al. / European Journal of Medicinal Chemistry 120 (2016) 51e63 63

activity, electrochemical aspects, and bioisosteric replacement of C-ring- Suppression of human keratinocyte hyperproliferation by 2-substituted
modified lapachones, Bioorg Med. Chem. 22 (2014) 1608e1619. naphtho[2,3-b]furan-4,9-diones, activation by enzymatic one- and two-
[8] N. Kongkathip, B. Kongkathip, P. Siripong, C. Sangma, S. Luangkamin, electron reduction, and intracellular generation of superoxide, J. Med. Chem.
M. Niyomdecha, S. Pattanapa, S. Piyaviriyagul, P. Kongsaeree, Potent anti- 55 (2012) 7273e7284.
tumor activity of synthetic 1,2-Naphthoquinones and 1,4-Naphthoquinones, [26] M. Efdi, S. Fujita, T. Inuzuka, M. Koketsu, Chemical studies on Goniothalamus
Bioorg Med. Chem. 14 (2003) 3179e3191. tapis Miq, Nat. Prod. Res. 24 (2010) 657e662.
[9] R.B. Weiss, The anthracyclines: will we ever find a better doxorubicin? Semin. [27] J.D. Bauer, R.W. King, S.F. Brady, Utahmycins a and B, azaquinones produced
Oncol. 19 (1992) 670e686. by an environmental DNA clone, J. Nat. Prod. 73 (2010) 976e979.
[10] O. Tacar, P. Sriamornsak, C.R. Dass, Doxorubicin: an update on anticancer [28] M.E. Suh, S.Y. Park, ChO. Lee, Synthesis of pyridino[2,3-f]indole-4,9-dione and
molecular action, toxicity and novel drug delivery systems, J. Pharm. Phar- 6,7-disubstituted quinoline-5,8-dione derivatives and evaluation on their
macol. 65 (2012) 157e170. cytotoxic activity, Bioorg. Med. Chem. 9 (2001) 2979e2986.
[11] D.A. Gewirtz, A critical evaluation of the mechanisms of action proposed for [29] H.J. Lee, M.E. Suh, Ch.O. Lee, Synthesis and cytotoxicity evaluation of 2-amino-
the antitumor effects of the anthracycline antibiotics adriamycin and and 2-hidroxy-3-ethoxycarbonyl-N-substituted-benzo[f]indole-4,9-dione de-
daunorubicin, Biochem. Pharmacol. 57 (1999) 727e741. rivatives, Bioorg. Med. Chem. 11 (2003) 1511e1519.
[12] E.A. Hillard, F.C. Abreu, D.C. Ferreira, G. Jaouen, M.O.F. Goulart, C. Amatore, [30] H.J. Park, H.-J. Lee, E.-J. Lee, H.J. Hwang, S.-H. Shin, M.-E. Shu, Ch Kim, H.J. Kim,
Electrochemical parameters and techniques in drug development, with an E.-K. Seo, S.K. Lee, Cytotoxicity and DNA topoisomerase inhibitory activity of
emphasis on quinones and related compounds, Chem. Commun. 23 (2008) benz[f]indole-4,9-dione analogs, Biosci. Biotechnol. Biochem. 67 (2003)
2612e2628. 1944e1949.
[13] T.J. Monks, R.P. Hanslik, G.M. Cohen, D. Ross, D.G. Graham, Quinone chemistry [31] H.-J. Lee, S.-Y. Park, J.S. Kim, H.M. Song, M.-E. Suh, Ch-O. Lee, Synthesis and
and toxicity, Toxicol. Appl. Pharmacol. 112 (1992) 2e16. cytotoxicity evaluation of pyridin[2,3-f]indole-2,4,9-trione and benz[f]indole-
[14] J.L. Bolton, M.A. Trush, T.M. Penning, G. Dryhurst, T.J. Monks, Role of quinines 2,4,9-trione derivatives, Bioorg. Med. Chem. 11 (2003) 4791e4796.
in toxicology, Chem. Res. Toxicol. 13 (2000) 135e160. [32] I. Choi, S. Woo, H.S. Park, H.-Y.P. Choo, Ch Kim, Correlation between the
[15] P.T. Schumacker, Reactive oxygen species in cancer cells: live by the sword, binding mode and inhibitory activity of benz[f]indole-4,9-dione analogs with
die by the sword, Cancer Cell 10 (2006) 175e176. human topoisomerase I-DNA, J. Korean Chem. Soc. 49 (2005) 449e460.
[16] J. Correa, J. Romo, The constituents of cacalia decomposita a. gray. structures [33] P. Kafarski, B. Lejczak, Biological activity of aminophosphonic acids, Phos-
of maturin, maturinin, maturone and maturinone, Tetrahedron 22 (1966) phorus Sulfur 63 (1991) 193e215.
685e691. [34] R. Engel, The use of carbon-phosphorus analogue compounds in the regula-
[17] P.M. Brown, R.H. Thompson, Naturally occurring quinones. Part XVI. Structure tion of biological processes, in: R. Engel, M. Dekker (Eds.), Handbook of
of maturinone and related compounds, J. Chem. Soc. C (1969) 1184e1186. Organophosphorus Chemistry, 1992, pp. 559e600. New York.
[18] H. Kakisawa, Y. Inouye, J. Romo, Diels-alder reaction of 3-methylbenzofuran- [35] S. Van der Jeught, C.V. Stevens, Direct phosphonylation of aromatic azahe-
4,7-quinone II. A revised structure of maturinone, Tetrahedron Lett. 10 (1969) terocycles, Chem. Rev. 109 (2009) 2672e2702.
1929e1932. [36] L.D. Quin, Organophosphorus compounds in medicine, in: A Guide to
[19] M. Aso, A. Ojida, G. Yang, O.-J. Cha, E. Osawa, K. Kanematsu, Furannulation Organophosphorus Chemistry, John Wiley & Sons, 2000, pp. 362e367. New
strategy for synthesis of the naturally occurring fused 3-methylfurans: effi- York.
cient synthesis of evodone and menthofuran and regioselective synthesis of [37] S.B. Tsogoeva, Recent progress in the development of synthetic hybrids of
maturone via a Lewis acid catalyzed Diels-Alder reactions. Some comments natural or unnatural bioactive compounds for medical chemistry, Mini-Rev.
for its mechanistic aspects, J. Org. Chem. 58 (1993) 3960e3968. Med. Chem. 10 (2010) 773e793.
[20] C. Ito, S. Katsuno, Y. Kondo, H.T.-W. Hugh, H. Furukawa, Chemical constituents [38] M. Decker, Hybrid molecules incorporating natural products: application in
of Avicennia alba. Isolation and structural elucidation of new naph- cancer therapy, neurodegenerative disorders and beyond, Curr. Med. Chem.
thoquinones and their analogues, Chem. Pharm. Bull. 48 (2000) 339e343. 18 (2011) 1464e1475.
[21] R.B. Williams, A. Norris, J.S. Miller, L.J. Razafitsalama, R. Andriantsiferana, [39] H.-Y. Hu, Y. Zhu, L. Wang, J.-H. Xu, A direct one-pot synthesis of naphtho[2,3-
V.E. Rasamison, D.G.I. Kingston, Two new cytotoxic naphthoquinones from b]furan-4,9-dione derivatives via C,O-dialkylation of b-dicarbonyl compounds
Mendoncia cowanii from the rainforest of Madagasca, Planta Med. 72 (2006) by 2,3-dichloro-1,4-naphthoquinones, Synthesis 10 (2005) 1605e1610.
564e566. [40] J. Ke˛ dzia, J. Modranka, T. Janecki, Efficient syntheses of 3-phosphorylquinolin-
[22] H. Kuo, K. Hotta, M. Yogo, S. Yoshina, Facile synthesis of unesterified enol- 4-ones and 3-phosphoryl-1,8-naphthyridin-4-ones, Tetrahedron Lett. 58
phosphates, Synthesis 3 (1979) 189e191. (2011) 6623e6626.
[23] H.-Y. Hu, Y. Zhu, L. Wang, J.-H. Xu, A direct one-pot synthesis of naphtho[2,3- [41] T. Mosmann, Rapid colorimetric assay for cellular growth and survival:
b]furan-4,9-dione derivatives via C,O-Dialkylation of b-dicarbonyl compounds application to proliferation and cytotoxicity assays, J. Immunol. Methods 65
by 2,3-Dichloro-1,4-naphthoquinones, Synthesis 10 (2005) 1605e1610. (1983) 55e63.
[24] K.-I. Hiral, J. Koyama, J. Pan, E. Shimamura, H. Shimada, T. Yamori, S. Sato, [42] D. Trachootham, J. Alexandre, P. Huang, Targeting cancer cells by ROS-
K. Tagahara, T. Tsuruo, Furanonaphthoquinone analogs possessing preferen- mediated mechanisms: a radical therapeutic approach? Nat. Rev. Drug Dis-
tial antitumor activity compared to normal cells, Cancer Detect. Prev. 23 cov. 8 (2009) 579e591.
(1999) 539e550. [43] V. Nogueira, N. Hay, Molecular pathways: reactive oxygen species homeo-
[25] A. Reichstein, S. Vortherms, S. Bannwitz, J. Tentrop, H. Prinz, K. Müller, Syn- stasis in cancer cells and implications for cancer therapy, Clin. Cancer Res. 19
thesis and structure-activity relationships of lapacho analogues. 1. (2013) 4309e4314.

You might also like