ArticlePDF Available

Discovery of Novel Alkylated (bis)Urea and (bis)Thiourea Polyamine Analogues with Potent Antimalarial Activities

Authors:

Abstract and Figures

A series of alkylated (bis)urea and (bis)thiourea polyamine analogues were synthesized and screened for antimalarial activity against chloroquine-sensitive and -resistant strains of Plasmodium falciparum in vitro. All analogues showed growth inhibitory activity against P. falciparum at less than 3 μM, with the majority having effective IC(50) values in the 100-650 nM range. Analogues arrested parasitic growth within 24 h of exposure due to a block in nuclear division and therefore asexual development. Moreover, this effect appears to be cytotoxic and highly selective to malaria parasites (>7000-fold lower IC(50) against P. falciparum) and is not reversible by the exogenous addition of polyamines. With this first report of potent antimalarial activity of polyamine analogues containing 3-7-3 or 3-6-3 carbon backbones and substituted terminal urea- or thiourea moieties, we propose that these compounds represent a structurally novel class of antimalarial agents.
Content may be subject to copyright.
rXXXX American Chemical Society Adx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000000
ARTICLE
pubs.acs.org/jmc
Discovery of Novel Alkylated (bis)Urea and (bis)Thiourea Polyamine
Analogues with Potent Antimalarial Activities
Bianca K. Verlinden,
Jandeli Niemand,
Janette Snyman,
Shiv K. Sharma,
Ross J. Beattie,
Patrick M. Woster,
§
and Lyn-Marie Birkholtz*
,
Department of Biochemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria, PO Box x20, Pretoria, 0028,
South Africa
Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University,
Detroit, Michigan 48202, United States
§
Department of Pharmaceutical and Biomedical Sciences, Medical University of South Carolina, Charleston, South Carolina 29425,
United States
INTRODUCTION
Malaria remains one of the most deadly parasitic diseases, with
nearly 250 million new cases each year, resulting in approxi-
mately one million deaths (www.who.int). The spreading resis-
tance of Plasmodium falciparum to existing antimalarials including
chloroquine, antifolates, and artemisinin has resulted in a press-
ing need to discover new chemotherapeutic agents against this
disease.
1
One class of promising antiparasitic agents include
inhibitors of polyamine biosynthesis
2
as well as polyamine analogues,
3
with ample evidence indicating that these rapidly dividing cells
have an exquisite need for the presence of polyamines for a
myriad of cellular functions during cell growth and division.
4,5
The naturally occurring polyamines putrescine (1), spermi-
dine (2), and spermine (3) (Figure 1) interact with a variety of
cellular eector sites due to their highly cationic nature and
specic spatial orientation of positive charge and are therefore
able to stabilize DNA, RNA, and other acidic cellular con-
stituents.
4
Polyamine analogues are structurally similar to the
naturally occurring polyamines and act as either polyamine
antimetabolites that deplete intracellular polyamine pools or
polyamine mimetics that displace the natural polyamines from
their binding sites without substituting for their cellular functions.
6
Particularly, terminal alkylation of polyamines and polyamine
analogues results in a change of pK
a
of the amine groups of these
molecules, resulting in nonfunctional polyamine characteristics.
7,8
Moreover, these analogues may compete for polyamine uptake and,
in mammalian cells in particular, can induce polyamine catabolism.
3
The rst generation of antiparasitic alkylpolyamines, typied
by the N,N0-bis(benzyl)-substituted polyamine analogue MDL
27695 (4, Figure 1), exhibited antitrypanosomal and antiplas-
modial activity in the μM range.
911
A bis[(2-phenyl)benzyl)]-
spermine analogue of 4known as BW-1 (5, Figure 1) was sub-
sequently shown to have inhibitory activity against various strains
of Trypanosoma and the microsporidial, Encephalitozoon cuniculi,
particularly by blocking polyamine uptake and inhibiting poly-
amine oxidase activity
9,11,12
and additionally being curative in a
rodent model of infection with the microsporidial organism.
9
Derivatives of 5include analogues of the substituted (bis)biguanide
known as 2d (6, Figure 1) that, in addition to depleting the
polyamine pool, inhibits trypanothione reductase (a spermidine
glutathione conjugate) activity in trypanosomes.
8
Compound 6
and its derivatives are highly active antiparasitic agents, with in
vitro IC
50
s against Trypanosoma brucei as low as 90 nM. Several
urea- and thiourea-based isosteres of 6have subsequently been
shown to be eective epigenetic modulators in mammalian cells
by inuencing selective chromatin marks in tumor cell lines
through inhibition of lysine specic demethylase 1, thereby decreas-
ing cancerous cell growth.
13
On the basis of the success of terminally (bis)alkylated polyamine
analogues against other parasites, several analogues of 6, as well
as a new generation of (bis)urea and (bis)thiourea alkylated
Received: April 18, 2011
ABSTRACT: A series of alkylated (bis)urea and (bis)thiourea
polyamine analogues were synthesized and screened for anti-
malarial activity against chloroquine-sensitive and -resistant
strains of Plasmodium falciparum in vitro. All analogues showed
growth inhibitory activity against P. falciparum at less than
3μM, with the majority having eective IC
50
values in the
100650 nM range. Analogues arrested parasitic growth within
24 h of exposure due to a block in nuclear division and therefore asexual development. Moreover, this eect appears to be cytotoxic
and highly selective to malaria parasites (>7000-fold lower IC
50
against P. falciparum) and is not reversible by the exogenous
addition of polyamines. With this rst report of potent antimalarial activity of polyamine analogues containing 373or363
carbon backbones and substituted terminal urea- or thiourea moieties, we propose that these compounds represent a structurally
novel class of antimalarial agents.
Bdx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
isosteres of 6, were synthesized and evaluated for their ability to
inhibit the proliferation of malaria parasites. These compounds
contain a variety of carbon backbones, and terminal urea/
thiourea substituents that are symmetrically substituted aralkyl
substituents, and as such present a structurally novel class of
scaolds, unrelated to any known antimalarials. This study reports
the antimalarial activity against drug sensitive and resistant
P. falciparum strains in vitro, their eects on the parasites
DNA replication and polyamine-specic events.
RESULTS AND DISCUSSION
Chemical Syntheses of Urea and Thiourea Polyamine
Analogues. To access a library of urea and thiourea analogues
isosteric to 6(720, Table 1; and 2539, Table 2) and anal-
ogous amidine analogues (2124, Table 1), we employed our
previously published synthesis
8,14
of precursor molecules 43,as
shown in Scheme 1. The appropriate diamine 40 (n=1,2,4,5)
was (bis)cyanoethylated (acrylonitrile, EtOH, reflux) to afford
the corresponding (bis)cyano intermediates 41. The central
nitrogens in 41 were then N-Boc protected ((Boc)
2
O, CH
2
Cl
2
/
aq NaHCO
3
)
15
to form 42, and the cyano groups were reduced
(Raney Ni) to yield the desired diamines 43.
8,16
Compounds 43
(n= 1, 2, 4, 5) were then reacted with 2 equiv of appropriate
alkyl- or aryl-substituted isocyanates or isothiocyanates 44 in
anhydrous CH
2
Cl
2
, followed by acid removal of the N-Boc
protection groups (HCl in EtOAc)
15
to afford the desired urea
or thiourea products (720 and 2539). The amidine analo-
gues compounds 2124 were prepared (Scheme 2) by reacting
diamines 43 with 2 equiv of S-naphthylmethyl thioacetimidate
hydrobromide 46 (prepared by refluxing 2-bromomethyl-
naphthalene with thiacetamide in CHCl
3
according to literature
procedure
14
) using absolute ethanol, and the Boc protecting
groups were subsequently removed with HCl in EtOAc.
In Vitro Activity of Polyamine Analogues against P. falci-
parum.The first diverse library of isoteric (bis)urea and (bis)thiourea
alkylated polyamine analogues was tested for possible growth
inhibitory affect against intraerythrocytic P. falciparum in vitro
(Table 1). The majority of these compounds showed in vitro
inhibitory activity against both drug resistant (W2 chloroquine
resistant strain, HB3 antifolate resistant strain) and sensitive
P. falciparum (strain 3D7) at concentrations below 3 μM(Table1).
Compound 6, containing terminal (bis)diphenylpropylguanidine
moieties, is active against P. falciparum at 298 nM. Conversely, it
is clear that amidine substituted analogues that lacked terminal
alkyl groups were not active against the malaria parasite. Com-
pound 21, an amidine analogue containing a 333 carbon
backbone, which lacks any (bis)urea and (bis)thiourea substit-
uents, was the least effective compound (IC
50
= 147 μM,
Table 1). Of the 19 compounds tested, 15 have potent in vitro
antimalarial activity (<1 μM), with the six most active com-
pounds (6,9,13,15,16, and 20) displaying IC
50
values in the
range of 144405 nM against the 3D7 strain of P. falciparum
(Table 1). Moreover, all of these compounds had IC
50
values in
the nM range against drug resistant P. falciparum, with com-
pounds 13,15,16, and 20 being more active against chloroquine-
resistant P. falciparum (strain W2) displaying low resistance
factors (ranging from 0.19 to 0.61) compared to chloroquine
against this strain. This suggests that these analogues are mini-
mally affected by the resistance mechanisms of, e.g., chloroquine,
with a mechanistically distinct mode of action (Table 1).
Analysis of the antimalarial eects of this rst series of (bis)urea
and (bis)thiourea alkylated polyamine analogues suggests that the
most potent compounds contain either a 373or343
carbon backbones, with the 373 carbon backbone delivering
the best activity against the parasite. Compounds with (bis)urea
substituents exhibited the most potent antimalarial activity, followed
by the (bis)thiourea substituted compounds. The diphenylpropyl
substituents proved more eective than the diphenylethyl substit-
uentsasterminalgroupsofthesecompounds.Analysisofthe
amidine alkylated polyamine analogues suggests that the 333
and 343 carbon backbones are not eective against the parasite
and that analogues with a 373 carbon backbone are active in the
nM range, particularly when they have terminal diphenylpropyl
substituents. On the basis of results observed with this rst
evaluation of the antimalarial activity of polyamine analogues, the
selective design and synthesis of a second series of compounds,
predicted to have a higher antimalarial capacity, was attempted. This
second series of 15 compounds contained 363, 373, and
343 backbones but with a variety of terminal substituents.
Figure 1. The natural polyamines putrescine (1), spermidine (2), and spermine (3) and antimalarial polyamine analogues MDL 27695 (4), BW-1 (5),
and compound 6.
Cdx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
Table 1. In Vitro Antimalarial Activity of the Compounds 624, against P. falciparum Strains 3D7, HB3, and W2
a
Values are the means (SE of at least three independent experiments (ng3).
b
P. falciparum drug sensitive strain 3D7.
c
P. falciparum chloroquine
resistant strain HB3.
d
P. falciparum antifolate resistant strain W2.
e
Resistance index (RI) dened as the ratio of the IC
50
values of the resistance to
sensitive strains, W2/3D7.
f
Indicates IC
50
values in μM.
g
Control drug, chloroquine
Ddx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
Of the second series of analogues, 13 had potent antimalarial
activity with IC
50
values in the range of 88846 nM (Table 2).
The ve most potent compounds (25,29,30,38, and 39), with
IC
50
values ranging between 88211 nM, were also active
against chloroquine resistant P. falciparum (W2 strain). These
compounds all have a 373or363 carbon architecture,
the majority with (bis)urea and terminal aralkyl substituents. Com-
pounds 34 and 35 were the least eective against the parasite,
with 34 not active in the μMrange,and35 with an IC
50
= 14.08 μM
(Table 2). Both of these contain 363 carbon backbones with
(bis)urea substituents, similar to the structures of the most
eective compounds, with the exception that they feature alkyl
rather than aralkyl substituent rings. This second generation of
synthesized (bis)urea and (bis)thiourea alkylated polyamine
analogues suggests that the most potent compounds contain
either a 363or373 carbon backbones, with the 363
Table 2. In Vitro Antimalarial Activity of the Second-Generation Compounds (2539), against P. falciparum Strains 3D7 and W2
a
Values are the means (SE of at least 3 independent experiments (ng3).
b
P. falciparum drug sensitive strain 3D7.
c
P. falciparum antifolate resistan
strain W2.
d
Resistance index (RI) dened as the ratio of the IC
50
values of the resistance to sensitive strains, W2/3D7.
e
Indicates IC
50
values in μM.
f
Control drug, chloroquine
Edx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
carbon backbone delivering the best activity against the parasite.
Compounds with (bis)urea substituents exhibited the most potent
antimalarial activity, followed by the (bis)thiourea substituted
compounds. The selection of terminal substituents of these com-
pounds are of vital importance in enhancing their antimalarial
activity, compounds with terminal phenyl rings had the best
activity, followed by diphenylethyl substituents, benzyl rings, and
last diphenylmethyl substituents. Compounds with no terminal
aralkyl groups were the least eective against the parasite with
IC
50
values in the high μM range, thus demonstrating the impor-
tance of bulky terminal substituents for antimalarial activity.
Some terminally, symmetrically substituted polyamine analo-
gues target DNA and exert a cytotoxic eect through DNA
aggregation.
7
In most instances, the polyamine analogues
(compounds 9,13,15,16, and 25,29,30,38, and 39) elicited
a signicant cytotoxic response in the parasite (P< 0.05), as
measured by a decrease in viable cell numbers (% parasitemia,
Figure 2). However, even through compound 20 was active
against the parasite, it was not able to decrease viable cell numbers in
P. falciparum over time, indicating a cytostatic action on in vitro
growth. During its asexual intraerythrocytic development,
P. falciparum replicates its DNA as it develops from single-
nucleated ring (1N) and trophozoite stages (1N) to multinucleated
schizont (2N or >2N) stages that result in up to 32 daughter
merozoites (mononucleated) being formed from a single parent
parasite.
17,18
Polyamines have been shown to be important for
DNA replication and therefore implied life cycle development in
Plasmodia.
19
The eects of the polyamine analogues (compounds
9,13,15, and 16, and 25,29,30,38, and 39) on the ability of the
parasite to replicate its DNA were monitored. An untreated
parasite population contained 28% of parasites with single nuclei
(1N), whereas 38% and 34% were in multinucleated schizonts
forms of 2N or >2N, respectively (Figure 3), demonstrating
complete intraerytrocytic asexual development as expected after
72 h of development as measured by ow cytometry. Treatment
of P. falciparum with compound 20 produced similar cytometric
proles to untreated parasites and was not able to prevent DNA rep-
lication, conrming its cytostatic nature. However, for compound 16,
Scheme 1
Scheme 2
Fdx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
a dramatic halt in schizogony and associated nuclear division was
observed with parasites containing 78% 1N rings/trophozoites,
20% 2N and 2% >2N schizonts after treatment with this
compound. Compounds 9,13,15,25,29, and 30 revealed
similar proles to that of compound 16 (results not shown).
Compounds 16,38, and 39 had the greatest inhibitory aect on
DNA replication and nuclear division with the majority of the
parasites being conned to the ring stage (Figure 3).
Treatment of P. falciparum with polyamine biosynthesis
inhibitors like the substrate analogues α-diuoromethylornithine
and 3-aminooxy-1-aminopropane has been shown to be reversible,
and therefore the inhibitory eect is alleviated with the addition
of exogenous polyamines to the parasite.
20,21
To investigate the
inuence of exogenous polyamines on P. falciparum growth
inhibition observed with the current series of polyamine analo-
gues, polyamine reversal studies were performed to determine if
treated parasites could recover when supplemented with exo-
genous putrescine. The inhibitory eect observed with the poly-
amine compounds could not be reversed with exogenous poly-
amines for any of the most potent compounds (Figure 4). The
decreased cell viability observed for compound 16 was again
visible over a 72 h time period, and this was already evident after the
rst 24 h, during which the parasite needed to start nuclear division.
Therefore, the cytotoxic action of these polyamine analogues on
P. falciparum seems to be independent of changes in the polyamine
pool. This may be due to the analoguesability to block the
intracellular binding sites of the natural polyamines, or to displace
intracellular polyamines from their binding sites.
10
Alternatively,
the mode of action of these polyamine analogues against P. falciparum
may be independent of the polyamine pathway. In trypanosomes,
polyamine analogues act as competitive inhibitors of enzymes not
directly related to polyamine biosynthesis.
22
It remains to be seen
if the polyamine analogues manifest their eect on P. falciparum
through targeting epigenetic control mechanisms, as has been ob-
served in mammalian cells.
13
Theparasiteseemstobeexquisitely
sensitive toward epigenetic regulatory mechanisms, particularly for
the control of expression of variant gene families.
23
P. falciparum appear to be highly sensitive to the polyamine
analogues described above, with the majority showing parasite
IC
50
values below 500 nM. To ensure that this eect was not
merely due to general toxicity of the compounds, in vitro cyto-
toxicity testing was performed in a sensitive mammalian cell line.
A subset of the compounds described in this manuscript, notably
14,15,and16, have been evaluated as potential antitumor agents in
the Calu-6 nonsmall cell human lung carcinoma cell line.
13
It is
Figure 2. Viable cell count of P. falciparum (3D7) treated with
compounds 16 or 20 (2 IC
50
). Parasites were treated for 72 h, after
which parasitemia was determined microscopically with Giemsa stains
(counting 100 parasites per slide 10,n= 3) and DNA levels were
quantied using SYBR Green I incorporation. Black bars, % parasitemia
at 0 h; gray bars, after 72 h. Results are the mean of three independent
experiments, performed in triplicate, (SE. Signicance is indicated at
P< 0.05 (*) as determined with a Student-ttest.
Figure 3. Flow cytometric analysis of nuclear division of P. falciparum
treated with compounds 16,38, and 39 (2 IC
50
). Ring or trophozoite
stage parasites contain 1 nucleus (1N), followed by nuclear division in
late trophozoites (2N) and multinucleated schizonts (>2N), repre-
sented as the % parasites in each population. Flow cytometric measure-
ment of nuclear content was performed with SYBR Green I staining of
DNA tracked in the FITC channel. Data are represented as the mean of
three independent experiments, performed in duplicate, (SE.
Figure 4. Inuence of exogenous addition of polyamines to P. falcipar-
um (3D7) treated with polyamine analogues. Parasites were treated with
compound 16 alone (2 IC
50
, gray circles), or supplemented with
1 mM putrescine after either 24 h (upward triangles) or 48 h (downward
triangles). Parasitemia was monitored for 72 h using SYBR Green I.
Untreated parasites are indicated with squares. Data are represented as
the mean of three independent experiments, performed in duplicate,
(SE. Error bars fall within symbols where not shown. Signicance is
indicated at P< 0.05 (*) as determined with two-way ANOVA.
Gdx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
important to note that these compounds exert antitumor eects
through re-expression of aberrantly silenced tumor suppressor genes
and as such are not inherantly cytotoxic in mammalian cells when used
as single agents. Maximal cytotoxicity to tumor cells in vitro and in vivo
can only be achieved through synergistic eects with a traditional agents
such as 5-azacytidine. The GI
50
values in the Calu-6 cell line for
1416 alone range between 10 and 40 μM and thus they are not
generally cytotoxic agents. The most active compounds against
P. falciparum (compounds 9,13,15,16,and20,and25,29,30,
38,and39) showed remarkable selectivity toward the parasite
compared to a mammalian cell line (HepG2 human hepatocellular
liver carcinoma, Table 3), with the majority of the compounds
(particularly compounds 20,25,29,30,38,and39) showing >500-
fold selectivity toward the parasite. Remarkably, the most active
compound (30)is7000-fold more selective toward the parasite.
Comparatively, compounds 14,15,and16 were not the most
eective against P. falciparum and inhibited cell growth in Calu-6 cells
at μM concentrations (Table 3), but even these compounds showed
more than 10-fold selectivity against the malaria parasite.
13
CONCLUSIONS
The results presented here indicate that the title compounds
do not show general cytotoxicity in mammalian cells and that P.
falciparum seems to show selective sensitivity to these polyamine
analogues. These results are encouraging in implicating this
series of polyamine analogues as highly selective antimalarial
agents. Moreover, the ability of polyamine analogues to cure
in vivo infections of malaria in the murine model of Plasmodia
berghei should provide clues as to the ultimate antimalarial
potential of this structurally distinct class of compounds, and
these studies are currently underway.
EXPERIMENTAL SECTION
All reagents and dry solvents were purchased from Aldrich Chemical
Co. (Milwaukee, WI), Sigma Chemical Co. (St. Louis, MO), or Acros
Chemical (Chicago, IL) and were used without further purication
except as noted below. Triethylamine was distilled from potassium
hydroxide and stored in a nitrogen atmosphere. Methanol was distilled
from magnesium and iodine under a nitrogen atmosphere and stored
over molecular sieves. Methylene chloride was distilled from phosphorus
pentoxide, and chloroform was distilled from calcium sulfate. Tetrahy-
drofuran was puried by distillation from sodium and benzophenone.
Dimethyl formamide was dried by distillation from anhydrous calcium
sulfate and was stored under nitrogen. Preparative scale chromato-
graphic procedures were carried out using E. Merck silica gel 60,
230440 mesh. Thin layer chromatography was conducted on Merck
precoated silica gel 60 F-254. Ion exchange chromatography was
conducted on Dowex 1 8200 anion exchange resin. All
1
Hand
13
C
NMR spectra were recorded on a Varian Mercury 400 MHz spectro-
meter, and all chemical shifts are reported as δvalues referenced to TMS.
In all cases,
1
H NMR,
13
C NMR, and IR spectra were consistent with
assigned structures. Mass spectra were recorded on a Kratos MS 80 RFA
(EI and CI) or Kratos MS 50 TC (FAB) mass spectrometer. Prior to
biological testing, target molecules 739 were determined to be 95%
pure or greater by HPLC chromatography using an Agilent series 1100
high-performance liquid chromatograph tted with a C18 reversed-
phase column. Compounds 720 were previously synthesized and their
1
H and
13
C NMR spectra have been reported.
13
General Procedure for the Synthesis of Urea and Thiourea
Analogues. Step A. In a 100 mL round-bottom flask, the centrally Boc
substituted diamino compounds 43 (0.5 mmol) were dissolved in 10 mL
of HPLC grade CH
2
Cl
2
and a solution of alkyl-, benzyl-, or phenyl-
substituted isocyanate or isothiocyanate, 44 (1.0 mmol, 2 equiv) in 5 mL
of CH
2
Cl
2
under cold condition was added. The flask was protected with
N
2
atmosphere, and the reaction mixture was allowed to stir at room
temperature for 1824 h, the progress for formation of product was
monitored by TLC using CH
2
Cl
2
/MeOH/NH
4
OH (94.4:5.0:0.5 or
89:10:1). After completion of the reaction, CH
2
Cl
2
was removed under
reduced pressure on a rotary evaporator to produce a viscous colorless
material. The product was transferred into the next step without further
purification.
Step B. The above crude product was dissolved in anhydrous
MeCO
2
Et (6.0 mL) and after 1 M HCl in MeCO
2
Et (6.0 mL) was
added, the reaction mixture becomes cloudy. The flask was protected
with N
2
atmosphere and the reaction mixture was allowed to stir at room
temperature for 2448 h, and the progress for formation of product was
monitored by TLC using CH
2
Cl
2
:MeOH:NH
4
OH (89:10:1 and
78:20:2). After completion of the reaction as confirmed by TLC, and
the nature of the product (white crystalline materials separated from the
solution), MeCO
2
Et was removed under reduced pressure on a rotary
evaporator to produce a white powder. The solid product was well
stirred with 20 mL of fresh MeCO
2
Et and the soluble part was decanted,
and the solid so obtained was vacuum-dried to give pure product as a
white solid.
1,14-Bis-{3-[1-(10,10-diphenylmethyl)thioureado]}-4,11-diazatetrade-
cane Hydrochloride, 25 (RJB-92-09C).
1
H NMR (DMSO-d
6
): δ8.83
(bs, 6H, NH), 8.26 (bs, 2H, NH), 7.28 (bs, 16H, ArH), 7.21 (bs, 4H,
ArH), 6.71 (b, 2H, CHPh
2
), 3.39 (bs, 4H, NCH
2
), 2.87 (bs, 4H,
NCH
2
), 2.82 (bs, 4H, NCH
2
), 1.85 (bs, 4H, CH
2
CH
2
), 1.58 (bs, 4H,
CH
2
CH
2
), 1.28 (bs, 4H, CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ183.21
(CdS), 143.41, 129.07, 127.89, 127.57 (ArC), 61.28, 47.22, 45.23,
41.39, 26.38, 26.08, 25.85 (CH
2
).
1,14-Bis-{3-[1-(20,20-diphenylethyl)thioureado]}-4,11-diazatetrade-
cane Hydrochloride, 26 (RJB-92-09).
1
H NMR (DMSO-d
6
): δ9.00
(bs, 4H, NH), 7.80 (bs, 2H, NH), 7.54 (b, 2H, NH), 7.28
(bs, 16H, ArH), 7.17 (bs, 4H, ArH), 4.37 (b, 2H, CHPh
2
),
4.01 (b, 4H, NCH
2
), 3.43 (bs, 4H, NCH
2
), 2.79 (bs, 8H, NCH
2
),
1.80 (bs, 4H, CH
2
CH
2
), 1.60 (bs, 4H, CH
2
CH
2
), 1.30 (bs,
4H, CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ183.00 (CdS), 143.40,
Table 3. Selectivity of Polyamine Analogues for Growth
Inhibition of P. falciparum (3D7) Compared to Selected
Mammalian Cells
compd P. falciparum
(IC
50
,μM)
Calu-6
(GI
50
,μM)
a
HepG2
(GI
50
,μM)
b
SI
c
(Calu-6/Pf)
SI
c
(HepG2/Pf)
90.144 (0.031 23.92 (0.4 166
13 0.253 (0.003 24.52 (1.4 97
14 1.316 (0.01 9.4 nd 7
15 0.329 (0.009 38.3 18.92 (1.39 38 58
16 0.405 (0.008 10.3 24.67 (0.3 25 61
20 0.355 (0.09 >200 >500
25 0.211 (0.007 >200 >500
29 0.106 (0.011 >200 >1500
30 0.088 (0.007 619.3 (25.8
d
7038
38 0.130 (0.002 >200 >1500
39 0.1 (0.003 33.8 (3.24 338
CQ 0.009 22.16 (0.39 2462
a
Data obtained from Sharma et al.
13
Calu-6 are human nonsmall cell
lung carcinoma cells.
b
Values are the means (SE of at least 2
independent experiments performed in duplicate. HepG2 are human
hepatocellular liver carcinoma cells.
c
Selectivity indices were determined
as the compound GI
50
mammalian cell/IC
50
P. falciparum.
d
Values are
the means (SE of 3 independent experiments performed in duplicate.
Hdx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
129.17, 128.65, 127.08 (ArC), 50.46, 48.71, 47.22, 45.12, 41.22,
26.25, 26.14, 25.84 (CH
2
).
1,14-Bis-{3-[ 1-( 30,30-diphenylpropyl)thioureado]}-4,11-diazatetra-
decane Hydrochloride, 27 (RJB-92-11C).
1
H NMR (DMSO-d
6
): δ8.86
(bs, 4H, NH), 7.87 (bs, 4H, NH), 7.317.24 (m, 16H, ArH), 7.14
(t, 4H, J= 7.2 Hz, ArH), 4.01 (t, 2H, J= 7.2 Hz, CHPh
2
), 3.43 (b, 4H,
NCH
2
), 3.22 (b, 4H, NCH
2
), 2.83 (b, 8H, NCH
2
), 2.23 (b, 4H, NCH
2
),
1.82 (bs, 4H, CH
2
CH
2
), 1.52 (bs, 4H, CH
2
CH
2
), 1.29 (bs, 4H,
CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ145.37, 129.11, 128.30, 126.79
(ArC), 48.62, 47.22, 45.17, 41.00, 34.92, 26.36, 26.11, 25.86 (CH
2
).
1,14-Bis-{3-[ 1-( phenyl)thioureado]}-4,11-diazatetradecane Hydr-
ochloride 28 (RJB-92-06).
1
H NMR (DMSO-d
6
): δ10.09 (bs, 2H, NH),
8.90 (b, 4H, NH), 8.33 (bs, 2H, NH), 7.45 (d, 4H, J= 8.0 Hz, ArH),
7.28 (t, 4H, J= 8.0 Hz, ArH), 7.06 (t, 2H, J= 7.6 Hz, ArH), 3.54 (b,
4H, NHCH
2
), 2.84 (b, 8H, NCH
2
), 1.96 (bs, 4H, CH
2
CH
2
), 1.60 (bs,
4H, CH
2
CH
2
), 1.35 (bs, 4H, CH
2
CH
2
).
13
C NMR (DMSO-d
6
):
δ181.29 (CdS), 140.10, 129.16, 124.61, 123.38, 47.21, 45.25, 41.34,
28.74, 26.12, 25.88.
1,14-Bis-{3-[1-(benzyl)thioureado]}-4,11-diazatetradecane Hydro-
chloride 29 (RJB-92-11).
1
H NMR (DMSO-d
6
): δ8.96 (bs, 4H, NH),
8.25 (b, 2H, NH), 8.05 (bs, 2H, NH), 7.367.20 (m, 10H, ArH), 4.64
(s, 4H, ArCH
2
NH), 3.48 (bs, 4H, NHCH
2
), 2.82 (bs, 8H, NCH
2
), 1.85
(bs, 4H, CH
2
CH
2
), 1.60 (bs, 4H, CH
2
CH
2
), 1.30 (bs, 4H, CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ182.00 (CdS), 140.01, 128.90, 127.91,
127.44, 47.23, 45.15, 41.30, 31.98, 26.34, 26.14, 25.85.
1,14-Bis-{3-[1-( phenyl)ureado]}-4,11-diazatetradecane Hydrochloride
30 (RJB-92-04).
1
HNMR(DMSO-d
6
): δ8.99 (s, 2H, NH), 8.88 (bs, 4H,
NH), 7.37 (d, 4H, J=8.0Hz,ArH), 7.19 (t, 4H, J=7.2Hz,ArH), 6.84
(t, 2H, J=7.2Hz,ArH), 6.66 (bs, 2H, NH), 3.14 (b, 4H, NHCH
2
), 2.85
(b, 8H, NCH
2
), 1.77 (t, 4H, J=6.8Hz,CH
2
CH
2
), 1.59 (bs, 4H, CH
2
CH
2
),
1.29 (bs, 4H, CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ156.36 (CdO), 141.23,
129.28, 121.62, 118.20, 47.22, 45.28, 36.83, 27.29, 26.09, 25.86.
1,14-Bis-{3-[1-(benzyl)ureado]}-4,11-diazatetradecane Hydrochloride
31 (RJB-92-13).
1
H NMR (DMSO-d
6
): δ9.06 (bs, 4H, NH), 7.30
7.16 (m, 14H, ArH, and NH), 4.18 (s, 4H, ArCH
2
NH), 3.08 (b, 4H,
NHCH
2
), 2.79 (b, 8H, NCH
2
), 1.73 (b, 4H, CH
2
CH
2
), 1.58 (bs, 4H,
CH
2
CH
2
), 1.27 (bs, 4H, CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ159.33
(CdO), 141.50, 128.88, 127.60, 127.20, 47.16, 45.12, 43.56, 36.99,
27.44, 26.10, 25.82.
1,14-Bis-{3-[1-( ethyl)thioureado]}-4,11-diazatetradecane Hydro-
chloride 32 (RJB-92-08).
1
H NMR (DMSO-d
6
): δ9.92 (bs, 4H, NH),
7.79 (s, 2H, NH), 7.79 (s, 2H, NH), 3.42 (bs, 4H, NHCH
2
), 3.31 (bs,
4H, NCH
2
), 2.82 (bs, 8H, NCH
2
), 1.82 (b, 4H, CH
2
CH
2
), 1.59 (b, 4H,
CH
2
CH
2
), 1.30 (b, 4H, CH
2
CH
2
), 1.02 (t, 6H, J= 7.20 Hz, CH
3
).
13
C
NMR (DMSO-d
6
): δ45.13, 41.04, 39.00, 26.36, 26.11, 25.84, 15.11.
1,14-Bis-{3-[1-( n-propyl)thioureado]}-4,11-diazatetradecane Hy-
drochloride 33 (RJB-92-14C).
1
H NMR (DMSO-d
6
): δ8.93 (bs, 4H,
NH), 7.83 (bs, 2H, NH), 7.76 (bs, 2H, NH), 3.43 (bs, 4H, NHCH
2
),
3.25 (bs, 4H, NCH
2
), 2.83 (bs, 8H, NCH
2
), 1.82 (bs, 4H, CH
2
CH
2
),
1.59 (bs, 4H, CH
2
CH
2
), 1.44 (q, 4H, J= 7.2 Hz, CH
2
CH
3
), 1.29 (b, 4H,
CH
2
CH
2
), 0.82 (t, 6H, J= 7.20 Hz, CH
3
).
13
C NMR (DMSO-d
6
): δ
47.20, 46.00, 45.13, 41.03, 26.36, 26.12, 25.84, 22.70,12.08.
1,14-Bis-{3-[1-(ethyl)ureado]}-4,11-diazatetradecane Hydrochlor-
ide 34 (RJB-92-15C).
1
H NMR (DMSO-d
6
): δ9.09 (bs, 4H, NH), 7.27
(b, NH), 7.79 (s, 2H, NH), 3.042.95 (m, 8H, NHCH
2
), 2.80 (bs, 8H,
NCH
2
), 1.71 (b, 4H, CH
2
CH
2
), 1.60 (b, 4H, CH
2
CH
2
), 1.29 (b, 4H,
CH
2
CH
2
), 0.95 (t, 6H, J= 6.8 Hz, CH
3
).
13
C NMR (DMSO-d
6
): δ
159.22 (CdO), 47.15, 45.10, 37.00, 34.92, 27.34, 26.11, 25.81, 16.23.
1,14-Bis-{3-[1-( n-propyl)ureado]}-4,11-diazatetradecane Hydro-
chloride 35 (RJB-92-13C).
1
H NMR (DMSO-d
6
): δ9.07 (bs, 4H,
NH), 7.77 (b, 4H, NH), 3.04 (t, 4H, J= 6.0 Hz, NHCH
2
), 2.90
(t, 4H, J=7.2Hz,NCH
2
), 2.79 (b, 8H, NCH
2
), 1.71 (q, 4H, J=6.8
Hz, CH
2
CH
2
), 1.60 (b, 4H, CH
2
CH
2
), 1.361.28 (m, 8H,
CH
2
CH
2
), 0.79 (t, 6H, J=7.6Hz,CH
3
).
13
C NMR (DMSO-d
6
):
δ159.36 (CdO), 47.14, 45.10, 41.87, 36.96, 27.38, 26.10, 25.80,
23.77, 12.02.
1,12-Bis-{3-[1-(10,10-diphenylmethyl)ureado]}-4,9-diazadodecane
Hydrochloride, 36 (SKS-96-02).
1
H NMR (DMSO-d
6
): δ8.95 (b, 4H,
NH), 8.19 (b, 2H, NH), 7.397.18 (m, 20H, ArH), 6.48 (b, 2H, NH),
5.86 (d, 2H, J= 8.4 Hz, CHPh
2
), 3.07 (t, 4H, J= 5.6 Hz, NCH
2
), 2.75 (b,
8H, NCH
2
), 1.79 (b, 4H, CH
2
CH
2
), 1.58 (b, 4H, CH
2
CH
2
).
13
C NMR
(DMSO-d
6
): δ158.64 (CdO), 144.45, 129.04, 127.59, 127.37 (ArC),
57.78, 46.55, 45.09, 36.86, 27.47, 23.20 (CH
2
).
1,12-Bis-{3-[ 1-( 20,20-diphenylethyl)ureado]}-4,9-diazadodecane Hydro-
chloride, 37 (SKS-96-01).
1
HNMR(DMSO-d
6
): δ9.11 (bs, 4H, NH),
7.347.15 (m, 20H, ArH), 4.10 (t, 2H, J= 6.0 Hz, CHPh
2
), 3.64 (m, 4H,
NCH
2
), 3.01 (bs, 4H, NCH
2
), 2.81 (bs, 4H, NCH
2
), 2.75 (bs, 4H, NCH
2
),
1.68 (bs, 8H, CH
2
CH
2
).
13
CNMR(DMSO-d
6
): δ159.05 (CdO), 143.77,
129.10, 128.58, 126.92 (ArC), 51.72, 46.51, 45.06, 44.53, 36.83, 27.39, 23.25
(CH
2
).
1,15-Bis-{3-[1-(10,10-diphenylmethyl)ureado]}-4,12-diazapentade-
cane Hydrochloride, 38 (SKS-96-02C).
1
H NMR (DMSO-d
6
): δ8.93
(bs, 4H, NH), 7.287.14 (m, 20H, ArH), 5.88 (bs, 2H, CHPh
2
), 3.08
(bs, 4H, NCH
2
), 2.77 (bs, 4H, NCH
2
), 2.69 (bs, 4H, NCH
2
), 1.71 (bs,
4H, CH
2
CH
2
), 1.53 (bs, 4H, CH
2
CH
2
), 1.20 (b, 6H, CH
2
CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ159.30 (CdO), 144.02, 129.60, 127.38,
127.60 (ArC), 57.70, 42.26, 45.07, 36.81, 28.56, 27.47, 26.37, 25.88
(CH
2
).
1,15-Bis-{3-[1-( 20,20-diphenylethyl)ureado]}-4,12-diazapentadecane
Hydrochloride, 39 (SKS-96-01C).
1
H NMR (DMSO-d
6
): δ9.06 (bs, 4H,
NH), 7.317.14 (m, 20H, ArH), 4.10 (t, 2H, J= 7.6 Hz, CHPh
2
), 3.64
(m, 4H, NCH
2
), 3.01 (t, 4H, J= 5.6 Hz, NCH
2
), 2.74 (bs, 8H, NCH
2
),
1.68 (m, 4H, CH
2
CH
2
), 1.64 (m, 4H, CH
2
CH
2
), 1.16 (bs, 6H,
CH
2
CH
2
CH
2
).
13
C NMR (DMSO-d
6
): δ159.11 (CdO), 143.75,
129.11, 128.58, 126.94 (ArC), 51.71, 47.26, 45.06, 44.55, 36.82, 28.62,
27.39, 26.42, 25.91 (CH
2
).
S-2-Naphthylmethyl Thioacetimidate Hydrobromide 46 (SKS-84-
31). To a stirred solution of thioacetamide (1.127 g, 15 mmol) in
anhydrous CHCl
3
(40 mL) was added 2-bromomethylnaphthalene 45
(3.40 g, 15 mmol) by cooling the reaction flask. The reaction mixture
was allowed to stir at room temperature for 5 min and then heated to
reflux for 2 h, cooled to back to room temperature, and placed in an ice
bath. The resulting solid was filtered off, washed with 50 mL CHCl
3
, and
dried in vacuum for 3 h to afford 46 (3.83 g, 86%) as a white solid,
1
H
NMR (DMSO-d
6
): δ8.00 (s, 1H ArH), 7.96 (m, 3H, ArH), 7.56
(m, 3H, ArH), 3.80 (s, 2H, SCH
2
), 2.62 (s, 3H, CH
3
).
13
C
NMR (DMSO-d
6
): δ193.48 (CdS), 133.48, 133.15, 131.42, 129.48,
128.83, 128.40, 128.37, 127.51, 127.42, 127.34 (ArC), 36.74 (CH
2
),
24.86 (CH
3
).
General Procedure for the Synthesis of Amidine Analo-
gues. The diamino compound 43 (0.50 mmol) was dissolved in 12 mL
of absolute ethanol, and a solution of S-2-naphthylmethyl thioacetimi-
date hydrobromide 46 (310 mg, 1.0 mmol, 2 equiv) was added under
cold condition. The flask was protected with N
2
atmosphere, the
resulting suspension that eventually becomes homogeneous was allowed
to stir at room temperature for 4872 h, and the progress for formation
of product was monitored by TLC using CH
2
Cl
2
/MeOH/NH
4
OH
(89:10:1). After completion of the reaction, the ethanol was removed
under reduced pressure on a rotary evaporator to produce a viscous
colorless material, which was purified by stirring the mixture with dry
ether, discarded ether soluble part, and insoluble material was again
stirred with fresh ether (25 mL). The white solid so obtained was dried
in vacuum.
1,11-Bis-(acetamidinyl)-4,8-di( tert-butyloxycarbonyl)-4,8-diazoun-
decane Hydrobromide 47a (SKS-84-40).
1
H NMR (DMSO-d
6
): δ9.38
(bs, 1H, NH), 9.06 (bs, 1H, NH), 8.55 (bs, 1H, NH), 8.27 (bs, 1H, NH),
7.19 (t, 2H, NH), 3.203.05 (m, 12H, NCH
2
), 2.11 (s, 6H, CH
3
),
1.721.60 (m, 6H, CH
2
), 1.33 (s, 18H, C[CH
3
]
3
).
Idx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
1,12-Bis-(acetamidinyl)-4,9-di( tert-butyloxycarbonyl)-4,9-diazodo-
decane Hydrobromide 47b (SKS-84-31C).
1
H NMR (DMSO-d
6
):
δ9.18 (b, 1H, NH), 9.04 (b, 1H, NH), 8.50 (b, 1H, NH), 7.91 (b,
1H, NH), 7.08 (t, 2H, NH), 3.1o (b, 12H, NCH
2
), 2.11 (s, 6H, CH
3
),
1.71 (m, 4H, CH
2
), 1.421.30 (bs, 22H, CH
2
and C[CH
3
]
3
).
1
H NMR
(CD
3
OD): δ3.30 (t, 4H, J= 7.2 Hz, NCH
2
), 3.24 (bs, 8H, NCH
2
), 2.24
(s, 6H, CH
3
), 1.90 (m, 4H, CH
2
), 1.53 (m, 4H, CH
2
), 1.45 (s, 18H,
C[CH
3
]
3
).
1,14-Bis-(acetamidinyl)-4,11-di( tert-butyloxycarbonyl)-4,11-diazo-
tetradecane Hydrobromide 47c (SKS-99-05C).
1
H NMR (CD
3
OD):
δ3.303.19 (m, 12H, NCH
2
), 2.24 (s, 6H, CH
3
), 1.87 (b, 4H, CH
2
),
1.55 (b, 4H, CH
2
), 1.45 (s, 18H, C[CH
3
]
3
), 1.32 (b, 4H, CH
2
).
1,15-Bis-(acetamidinyl)-4,12-di( tert-butyloxycarbonyl)-4,12-diazo-
pentadecane Hydrobromide 47d (SKS-84-33C).
1
H NMR (CD
3
OD):
δ3.283.19 (m, 12H, NCH
2
), 2.22 (s, 6H, CH
3
), 1.86 (m, 4H, CH
2
),
1.54 (m, 4H, CH
2
), 1.45 (s, 18H, C[CH
3
]
3
), 1.29 (m, 6H, CH
2
).
Cleavage of Boc Group. The compound 47ad(0.430.45
mmol) was stirred with 6 mL of anhydrous MeCO
2
Et for 5 min, and 1 M
HCl in MeCO
2
Et (5 mL) was added. The flask was protected with N
2
atmosphere, and the reaction mixture was allowed to stir at room
temperature for 1824 h, the progress for formation of product was
monitored by TLC (CH
2
Cl
2
:MeOH:NH
4
OH 78:20:2). After comple-
tion of the reaction, MeCO
2
Et was removed under reduced pressure on
a rotary evaporator to produce a white powder. The solid product was
well stirred with 20 mL of fresh MeCO
2
Et, the decanted the soluble part
was decanted, and the solid so obtained was vacuum-dried to give pure
product 2124 as a white solid.
1,11-Bis-(acetamidinyl)-4,8-diazoundecane Hydrochloride 21 (SKS-
84-40C).
1
H NMR (DMSO-d
6
): δ9.75 (s, 2H, NH), 9.29 (s, 2H,
NH), 9.23 (s, 4H, NH), 8.79 (s, 2H, NH), 2.98 (b, 12H, NCH
2
), 2.13 (s,
6H, CH
3
), 2.07 (b, 2H, CH
2
), 1.89 (b, 4H, CH
2
).
13
C NMR (DMSO-
d
6
): δ164.79 (dCH), 44.83, 44.61, 39.54 (NCH
2
), 24.73, 22.85(CH
2
),
19.27 (CH
3
).
1
H NMR (D
2
O): δ3.35 (t, 4H, J= 6.4 Hz, NCH
2
),
3.183.11 (m, 8H, NCH
2
), 2.19 (s, 6H, CH
3
), 2.122.09 (m, 2H,
CH
2
), 2.082.01 (m, 2H, CH
2
).
13
C NMR (D
2
O): δ164.80 (dCH),
45.35, 44.83, 39.37 (NCH
2
), 29.86, 24.07(CH
2
), 18.70 (CH
3
).
1,12-Bis-(acetamidinyl)-4,9-diazododecane Hydrochloride 22 (SKS-
84-35).
1
H NMR (DMSO-d
6
): δ9.88 (s, 2H, NH), 9.10 (s, 6H, NH),
8.81 (s, 2H, NH), 3.34 (m, 4H, NCH
2
), 2.94 (b, 4H, NCH
2
), 2.87 (b, 4H,
NCH
2
), 2.14 (s, 6H, CH
3
), 1.90 (m, 4H, CH
2
), 1.71 (b, 4H, CH
2
).
13
C
NMR (DMSO-d
6
): δ164.79 (dCH), 46.62, 44.72, 39.84 (NCH
2
), 24.71,
23.21 (CH
2
), 19.26 (CH
3
);
1
HNMR(D
2
O): δ3.34 (t, 4H, J=7.2Hz,
NCH
2
), 3.133.06 (m, 8H, NCH
2
), 2.20 (s, 6H, CH
3
), 2.02 (m, 4H,
CH
2
), 1.76 (m, 4H, CH
2
).
13
CNMR(D
2
O): δ47.26, 45.19, 39.40
(NCH
2
), 24.09, 23.07 (CH
2
), 18.69 (CH
3
). HR-MS m/z285.4
1,14-Bis-(acetamidinyl)-4,11-diazotetradecane Hydrochloride, 23
(SKS-99-06).
1
H NMR (DMSO-d
6
): δ9.79 (s, 2H, NH), 9.27 (s, 2H,
NH), 9.13 (s, 4H, NH), 8.82 (s, 2H, NH), 3.33 (b, 4H, NCH
2
), 2.93
(b, 4H, NCH
2
), 2.83 (b, 4H, NCH
2
), 2.14 (s, 6H, CH
3
), 1.90 (b, 4H,
CH
2
), 1.63 (b, 4H, CH
2
) 1.30 (b, 4H, CH
2
).
13
C NMR (DMSO-d
6
):
δ164.77 (dCH), 47.28, 44.77, 39.98 (NCH
2
), 26.11, 25.74, 24.73-
(CH
2
), 19.22 (CH
3
).
1
H NMR (D
2
O): δ3.21 (t, 4H, J= 6.4 Hz,
NCH
2
), 2.96 (t, 4H, J= 7.2 Hz, NCH
2
), 2.90 (t, 4H, J= 7.6 Hz, NCH
2
),
2.06 (s, 6H, CH
3
), 1.89 (m, 4H, CH
2
), 1.54 (b, 4H, CH
2
) 1.25 (b, 4H,
CH
2
).
13
C NMR (D
2
O): δ165.30 (dCH), 47.81, 44.97, 39.36
(NCH
2
), 25.45, 25.34, 23.99(CH
2
), 19.10 (CH
3
).
1,15-Bis-(acetamidinyl)-4,12-diazopentadecane Hydrochloride 24
(SKS-84-34).
1
H NMR (DMSO-d
6
): δ9.78 (s, 2H, NH), 9.25 (s, 2H,
NH), 9.12 (s, 4H, NH), 8.81 (s, 2H, NH), 3.33 (m, 4H, NCH
2
), 2.93 (b,
4H, NCH
2
), 2.82 (b, 4H, NCH
2
), 2.14 (s, 6H, CH
3
), 1.89 (m, 4H,
CH
2
), 1.62 (b, 4H, CH
2
) 1.27 (b, 6H, CH
2
).
13
C NMR (DMSO-d
6
):
δ164.77 (dCH), 47.35, 44.75, 39.84 (NCH
2
), 28.58, 26.39, 25.86,
24.75 (CH
2
), 19.26 (CH
3
).
1
H NMR (D
2
O): δ3.34 (t, 4H, J= 6.4 Hz,
NCH
2
), 3.09 (t, 4H, J= 8.0 Hz, NCH
2
), 3.03 (t, 4H, J= 8.0 Hz, NCH
2
),
2.20 (s, 6H, CH
3
), 2.04 (m, 4H, CH
2
), 1.66 (m, 4H, CH
2
), 1.35 (b, 6H,
CH
2
).
13
C NMR (D
2
O): δ48.05, 45.05, 39.46 (NCH
2
), 27.97, 25.73,
24.09 (CH
2
), 18.71 (CH
3
).
In Vitro Cultivation of P. falciparum.P. falciparum 3D7 (chloroquine
sensitive), W2 (chloroquine resistant), and HB3 (antifolate resistant) strains
were used to assess the in vitro antimalarial efficacy of the polyamine analogues.
The parasites were maintained in O
+
human red blood cells suspended at 5%
hematocrit in RPMI-1640 culture medium containing 23.81 mM sodium
bicarbonate, 0.024 mg/mL gentamycin, 25 mM HEPES, 0.2% glucose, 0.2 mM
hypoxanthine, and 5 g/L Albumax II. The parasites were incubated with
moderate shaking at 60 rpm at 37 °Cinanatmosphereof5%CO
2
,5%O
2
,and
90% N
2
.
24
Cultures were synchronized to >95% in the ring stage with 10%
(w/v) D-sorbitol treatment.
In Vitro Assessment of Antimalarial Activity. In vitro activity
against erythrocytic stages of P. falciparum (strains 3D7 and W2) was
determined using the Malaria SYBR Green I-based fluorescence assay
(MSF)
25
based on the DNA binding properties of this dye. Compounds
were dissolved in a nonlethal DMSO concentration (<0.013%),
26
serially diluted and added to the ring stage P. falciparum (1% parasitemia,
2% hematocrit) and incubated at 37 °C, static for 96 h. Subsequently,
equal volumes (100 μL) of the parasite suspension were added to SYBR
Green I lysis buffer (0.2 μL/mL of 10000SYBR Green I (Invitrogen),
20 mM Tris, pH 7.5, 5 mM EDTA, 0.008% (w/v) saponin, 0.08% (v/v)
Triton X-100) and incubated in the dark for 1 h at room temperature.
Fluorescence was read with a Fluoroskan Ascent FL microplate reader at
an excitation of 485 nm and emission of 538 nm. The data was
represented as percentage of untreated control to determine cell
proliferation. Nonlinear regression curves were generated using Sigma
Plot 11.0, from which the 50% inhibitory concentrations (IC
50
) could be
determined. Each compound was tested in duplicate for at least three
independent biological replicates.
Determination of Parasite DNA Replication. The effect of the
compounds on P. falciparum DNA replication and nuclear division was
determined using flow cytometry. Parasites (2% parasitemia, 2% hemat-
ocrit) were treated with test compounds (2 IC
50
), and 50 μL samples
were isolated at set time intervals following drug exposure. Parasites
were fixed with of 1 mL of 0.025% glutaraldehyde for 45 min and kept at
4°C until use. Fixed cells were washed twice with 1PBS, resuspended
in 20 μL PBS, and stained with 20 μL 1:1000 SYBR Green I for 30 min in
the dark at room temperature. DNA fluorescence was measured with a
BD FACS Aria I flow cytometer (Becton Dickinson) analyzing 10
6
cells
for each sample with fluorescence emission collected at an excitation
wavelength of 488 nm with 502 nm long band-pass and 530 nm band-
pass emission filters. BD FACS Diva Software (6.1.1) and FlowJo v9.1
(Tree Star) were used to analyze the data.
Determination of Polyamine Reversal of Inhibition. The
ability of exogenous putrescine to rescue polyamine analogue treated
parasites was determined using polyamine reversal studies that were set
up in the format similar to that of the MSF assay. P. falciparum 3D7
cultures were treated in early ring stages with test compounds at 2
IC
50
. After 24 and 48 h, the cultures were supplemented with 1 mM
putrescine and incubated for a further 24 or 48 h, after which parasitemia
was determined as described for the MSF assay. Statistical analysis was
performed with GraphPad InStat 3.10, all data given are the mean of at
least three independent biological repeats.
Cytotoxicity Determinations in Mammalian Cells. Human
hepatocellular liver carcinoma cells (HepG2, kind gift by Duncan
Cromarty, University of Pretoria) were maintained in Dulbeccos
Modified Eagles Medium (DMEM) supplemented with 10% heat
inactivated fetal bovine serum and 1% penicillin/streptomycin at
37 °C (5% CO
2
, 90% humidity). Cytotoxicity was measured using the
lactate dehydrogenase assay (LDH). Cells (100000) were seeded in 96-
well plates and grown for 24 h at 37 °C, after which cells were treated
with various concentrations of the compounds. After 48 h exposure, cells
Jdx.doi.org/10.1021/jm200463z |J. Med. Chem. XXXX, XXX, 000–000
Journal of Medicinal Chemistry ARTICLE
were pelleted at 250gfor 10 min and LDH activity was measured in the
supernatant (10 μL) by adding 100 μL LDH reaction mix (BioVision)
and incubating for 30 min at room temperature. Colorimetric detection
of NADH levels (as a measurement of LDH-mediated oxidation of
lactate indicating LDH activity) occurred at 450 nm. Experiments were
performed in duplicate for at least two independent biological repeats.
AUTHOR INFORMATION
Corresponding Author
*Phone: +27 12 420 2479. Fax: +27 12 362 5302. E-mail:
lbirkholtz@up.ac.za.
ACKNOWLEDGMENT
We thank Wayne Barnes at the Flow Cytometry and Cell
Sorting Unit in the Department of Biochemistry, University of
Pretoria, for technical assistance and Bridgette Cummings for
proofreading the manuscript. This work was supported by the
South African Malaria Initiative (www.sami.org.za), the South
African National Research Foundation (NRF Grant FA2007050-
300003) and the University of Pretoria (L.M.B.) and National
Institutes of Health grant 7RO1-CA149095 (P.M.W.). B.V. was
supported by grants from TATA Africa and the South African
Malaria Initiative. Any opinion, ndings and conclusions or
recommendations expressed in this paper are those of the
author(s) and therefore the NRF does not accept any liability
in regard hereto.
ABBREVIATIONS USED
MSF, malaria SYBR Green I-based uorescence assay
REFERENCES
(1) Dondorp, A.; Yeung, S.; White, L.; Nguon, C.; Day, N.; Socheat,
D.; von Seidlein, l. Artemisinin resistance: current status and scenarios
for containment. Nature Rev. Microbiol. 2010,8, 272280.
(2) Heby, O.; Persson, L.; Rentala, M. Targeting the polyamine
biosynthetic enzymes: a promising approach to therapy of african
sleeping sickness, Chagasdisease, and leishmaniasis. Amino Acids 2007
33 (2), 359366.
(3) Casero, R. A.; Marton, L. J. Targeting polyamine metabolism and
function in cancer and other hyperproliferative diseases. Nature Rev.
Drug Discovery 2007,6(5), 373390.
(4) Wallace, H. M.; Fraser, A. V.; Hughes, A. A perspective of
polyamine metabolism. Biochem. J. 2003,15 (376), 114.
(5) Clark, K.; Niemand, J.; Reeksting, S.; Smit, S.; Van Brummelen,
A.; Williams, M.; Louw, A. I.; Birkholtz, L. Functional consequences of
perturbing polyamine metabolism in the malaria parasite. Amino Acids
2010,38 (2), 633644.
(6) Wallace, H. M.; Niiranen, K. Polyamine analoguesan update.
Amino Acids 2007,33 (2), 261265.
(7) Casero, R. A.; Woster, P. M. Recent advances in the development
of polyamine analogues as antitumor agents. J. Med. Chem. 2009,52
(15), 45514573.
(8) Bi, X.; Lopez, C.; Bacchi, C.; Rattendib, D.; Woster, P. Novel
alkylpolyaminoguanidines and alkylpolyaminobiguanides with potent
antitrypanosomal activity. Bioorg. Med. Chem. Lett. 2006,16 (12),
32293232.
(9) Zou, Y.; Wu, Z.; Sirisoma, N.; Woster, P. M.; Casero, R. A.;
Weiss, L. M.; Rattendi, D.; Lane, S.; Bacchi, C. J. Novel alkylpolyamine
analogues that possess both antitrypanosomal and antimicrosporidial
activity. Bioorg. Med. Chem. Lett. 2001,11 (12), 16131617.
(10) Bitonti, A.; Dumont, J.; Bush, T.; Edwards, M.; Stemerick, D.;
McCann, P.; Sjoerdsma, A. Bis(benzyl)polyamine analogs inhibit the
growth of chloroquineresistant human malaria parasites (Plasmodium
falciparum) in vitro and in combination with α-diuoromethylornithine
cure murine malaria. Proc. Natl. Acad. Sci. U.S.A. 1989,86 (2), 651655.
(11) Bacchi, C. J.; Yarlett, N.; Faciane, E.; Bi, X.; Rattendi, D.; Weiss,
L. M.; Woster, P. M. Metabolism of an alkyl polyamine analog by a
polyamine oxidase from the microsporidian Encephalitozoon cuniculi.
Antimicrob. Agents Chemother. 2009,53 (6), 25992604.
(12) Bacchi, C. J.; Rattendi, D.; Faciane, E.; Yarlett, N.; Weiss, L. M.;
Frydman, B.; Woster, P. M.; Wei, B.; Marton, L. J.; Wittner, M. Polyamine
metabolism in a member of the phylum Microspora (Encephalitozoon
cuniculi): eects of polyamine analogues. Microbiology 2004,150 (5),
12151224.
(13) Sharma, S.; Wu, Y.; Steinbergs, N.; Crowley, M.; Hanson, A.;
Casero, R. A. J.; Woster, P. Bis)urea and (bis)thiourea inhibitors of
lysine-specic demethylase 1 as epigenetic modulators. J. Med. Chem.
2010,53 (14), 51975212.
(14) Shearer, B. G.; Oplinger, J. A.; Lee, S. S-2-Naphthylmethyl
thioacetimidate hydrobromide: a new odorless reagent for the mild synthesis
of substituted acetamidines. Tetrahedron Lett. 1997,38 (2), 179182.
(15) Keller, O.; Keller, W. E.; van Look, G.; Wersin, G. tert-
Butoxycarbonylation of amino acids and their derivatives: N-tert-Butox-
ycarbonyl-l-phenylalanine. Org. Synth. 1985,63, 160171.
(16) Bellevue, F. H.; Baohbedason, M.; Wu, R.; Woster, P. M.;
Casero, R. A. J.; Rattendi, D.; Lane, S.; Bacchi, C. J. Structural com-
parison of alkylpolyamine analogues with potent in vitro antitumor or
antiparasitic activity. Bioorg. Med. Chem. Lett. 1996,6(22), 27652770.
(17) Reilly, H. B.; Want, H.; Steuter, J. A.; Marx, A. M.; Ferdig, M. T.
Quantitative dissection of clone-specic growth rates in cultures malaria
parasites. Int. J. Parasitol. 2007,37 (14), 15991607.
(18) Izumiyama, S.; Omura, M.; Takasaki, T.; Ohmae, H.; Asahi, H.
Plasmodium falciparum: development and validation of a measure of
intraerythrocytic growth using SYBR Green I in a ow cytometer. Exp.
Parasitol. 2009,121 (2), 144150.
(19) Assaraf, Y. G.; Abu-Elheiga, L.; Spira, D. T.; Desser, H.;
Bachrach, U. Eect of polyamine depletion on macromolecular synthesis
of the malarial parasite, Plasmodium falciparum, cultured in human
erythrocytes. Biochem. J. 1987,242 (1), 221226.
(20) Das Gupta, R.; Krause-Ihle, T.; Bergmann, B.; Muller, I.;
Khomutov, A.; Muller, S.; Walter, R.; Luersen, K. 3-Aminooxy-1-
aminopropane and derivatives have an antiproliferative eect on cul-
tured Plasmodium falciparum by decreasing intracellular polyamine
concentrations. Antimicrob. Agents Chemother. 2005,49 (7), 28572864.
(21) Assaraf, Y. G.; Golenser, J.; Spira, D. T.; Messer, G.; Bachrach,
U. Cytostatic eect of DL-alpha-diuoromethylornithine against Plasmo-
dium falciparum and its reversal by diamines and spermidine. Parasitol.
Res. 1987,73 (4), 313318.
(22) Li, Z.; Fennie, M. W.; Ganem, B.; Hancock, M. T.; Kosbaslija,
M.; Rattendi, D.; Bacchi, C. J.; OSullivan, M. C. Polyamines with N-(3-
phenylpropyl) substituents are eective competitive inhibitors of trypa-
nothione reductase and trypanocidal agents. Bioorg. Med. Chem. Lett.
2001,11 (2), 251254.
(23) Duraisingh, M. T.; Voss, T. S.; Marty, A. J.; Duy, M. F.; Good,
R. T.; Thompson, J. K.; Freitas-Junior, L. H.; Scherf, A.; Crabb, B. S.;
Cowman, A. F. Heterochromatin silencing and locus repositioning
linked to regulation of virulence genes in Plasmodium falciparum.Cell
2005,121 (1), 1324.
(24) Trager, W.; Jensen, J. D. Human malaria parasites in continuous
culture. Science 1976,193 (4254), 637.
(25) Smilkstein, M.; Sriwilaijaroen, N.; Kelly, J.; Wilairat, P.; Riscoe,
M. Simple and inexpensive uorescence-based technique for high-
throughput antimalarial drug screening. Antimicrob. Agents Chemother.
2004,48 (5), 18031806.
(26) Grobusch, M. P.; Hanscheid, T.; Gobels, K.; Slevogt, H.; Zoller,
T.; Rogler, G.; Teichmann, D. Comparison of three antigen detection
tests for diagnosis and follow-up of falciparum malaria in travellers
returning to Berlin, Germany. Parasitol. Res. 1998,89 (5), 354357.
... In this context, thiourea is a relevant class of sulfur-bearing substances with notable synthetic and biological versatility. Thiourea derivatives have shown promising pharmaceutical potential due to their diverse pharmacological activities, including antitumoral [11], antioxidant [12], antidiabetic [13], and antimicrobial [14][15][16][17][18][19][20] properties. Notably, the thiourea moiety has been identified in several compounds with potential for the development of new therapeutic compounds to treat diseases caused by trypanosomatid species [21][22][23]. ...
... accessed on 28 April 2023) [29]. Thiourea derivatives with a promising inhibitory effect against different species of protozoans have been described in the literature, including Cryptosporidium hominis [16], Plasmodium falciparum [14,15], Leishmania amazonensis [22], and Trypanosoma spp. [21,23]. ...
... Thiourea derivatives with a promising inhibitory effect against different species of protozoans have been described in the literature, including Cryptosporidium hominis [16], Plasmodium falciparum [14,15], Leishmania amazonensis [22], and Trypanosoma spp. [21,23]. ...
Article
Full-text available
For decades, only two nitroheterocyclic drugs have been used as therapeutic agents for Chagas disease. However, these drugs present limited effectiveness during the chronic phase, possess unfavorable pharmacokinetic properties, and induce severe adverse effects, resulting in low treatment adherence. A previous study reported that N-(cyclohexylcarbamothioyl) benzamide (BTU-1), N-(tert-butylcarbamothioyl) benzamide (BTU-2), and (4-bromo-N-(3-nitrophenyl) carbamothioyl benzamide (BTU-3) present selective antiprotozoal activity against all developmental forms of Trypanosoma cruzi Y strain. In this study, we investigated the mechanism of action of these compounds through microscopy and biochemical analyses. Transmission electron microscopy analysis showed nuclear disorganization, changes in the plasma membrane with the appearance of blebs and extracellular arrangements, intense vacuolization, mitochondrial swelling, and formation of myelin-like structures. Biochemical results showed changes in the mitochondrial membrane potential, reactive oxygen species content, lipid peroxidation, and plasma membrane fluidity. In addition, the formation of autophagic vacuoles was observed. These findings indicate that BTU-1, BTU-2, and BTU-3 induced profound morphological, ultrastructural, and biochemical alterations in epimastigote forms, triggering an autophagic-dependent cell death pathway.
... The extract and SPE fractions of A. marlothii roots were subjected to antiplasmodial in vitro activity assessment against P. falciparum NF54 (drug-sensitive) ABS parasites using the malaria SYBR Green I-based fluorescence assay (Johnson et al., 2007;Smilkstein et al., 2004;Verlinden et al., 2011). Parasitology work and volunteer human blood donation at the University of Pretoria is covered under ethical approval from the Health Sciences Ethics Committee (506/2018) and Natural and Agricultural Sciences Ethics Committee (180000094) to LB. ...
... were treated with an equal volume of either the crude extract or SPE fractions in sterile 96 well plates, for 96 h at 37 • C under hypoxic conditions. The SYBR Green I fluorescence was determined as before (Johnson et al., 2007;Smilkstein et al., 2004;Verlinden et al., 2011). ...
... The activity of the isolated compounds was determined against drug sensitive NF54 P. falciparum ABS (Table 2). Chloroquine served as a positive control giving an IC 50 of 0.004 μg/mL (0.008 μM) consistent with prior studies (Verlinden et al., 2011). Compounds 1-5 displayed varied potency against ABS P. falciparum ranging from good (IC 50 < 10 μg/mL) to moderate (IC 50 of 10-50 μg/mL) activity, as per the Fig. 3. ...
Article
Ethnopharmacological relevance Aloe marlothii A.Berger (Xanthorrhoeaceae) is indigenous to southern African countries where its aqueous preparations are used in traditional medicine to treat several ailments including hypertension, respiratory infections, venereal diseases, chest pain, sore throat and malaria. Aim of the study The aims of this study were as follows: (i) isolate and identify the antiplasmodial active compounds in A. marlothii roots. As the water extract was previously inactive, the dichloromethane:methanol (DCM:MeOH) (1:1) was used, (ii) examine the activity of the isolated compounds against Plasmodium falciparum asexual blood stage (ABS) parasites as well as for transmission-blocking activity against gametocytes and gametes, and (iii) to use in silico tools to predict the target(s) of the active molecules. Materials and methods The crude DCM:MeOH (1:1) extract of A. marlothii roots was fractionated on a reverse phase C8 column, using a positive pressure solid-phase extraction (ppSPE) workstation to produce seven fractions. The resulting fractions and the crude DCM:MeOH extract were tested in vitro against P. falciparum (NF54) ABS parasites using the malaria SYBR Green I based-fluorescence assay. Flash silica chromatography and mass-directed preparative high-performance liquid chromatography were utilised to isolate the active compounds. The isolated compounds were evaluated in vitro against P. falciparum asexual (NF54 and K1 strains) and sexual (gametocytes and gametes) stage parasites. Molecular docking was then used for the in silico prediction of targets for the isolated active compounds in P. falciparum. Results The crude extract and two SPE fractions displayed good antiplasmodial activity with >97% and 100% inhibition of ABS parasites proliferation at 10 and 20 μg/mL, respectively. Following UPLC-MS analysis of these active fractions, a targeted purification resulted in the isolation of six compounds identified as aloesaponol I (1), aloesaponarin I (2), aloesaponol IV (3), β-sorigenin-1-O-methylether (4), emodin (5), and chrysophanol (6). Aloesaponarin I (2) was the most bioactive, compared to other isolated constituents, against P. falciparum ABS parasites exhibiting equipotency against the drug-sensitive (NF54) (IC50 = 1.54 μg/mL (5 μM)) and multidrug-resistant (K1) (IC50 = 1.58 μg/mL (5 μM)) strains. Aloesaponol IV (3) showed pronounced activity against late-stage (>90% stage IV/V) gametocytes (IC50 = 6.53 μg/mL (22.6 μM)) demonstrating a 3-fold selective potency towards these sexual stages compared to asexual forms of the parasite (IC50 = 19.77 ± 6.835 μg/mL (68 μM)). Transmission-blocking potential of aloesaponol IV (3) was validated by in vitro inhibition of exflagellation of male gametes (94% inhibition at 20 μg/mL). In silico studies identified β-hematin and DNA topoisomerase II as potential biological targets of compounds 2 and 3, respectively. Conclusion The findings from our study substantiate the traditional use of A. marlothii to treat malaria. To our knowledge, this study has provided the first report on the isolation and identification of antiplasmodial compounds from A. marlothii roots. Furthermore, our study has provided the first report on the transmission-blocking potential of one of the compounds from the genus Aloe, motivating for the investigation of other species within this genus for their potential P. falciparum transmission-blocking activity.
... Antiplasmodial activity of extracts, fractions and isolated compounds was carried out using the malaria SYBR Green I-based fluorescence assay (Smilkstein et al. 2004;Johnson et al. 2007;Verlinden et al. 2011). Parasitology work and volunteer human blood donation at the University of Pretoria was covered under ethical approval from the Health Sciences Ethics Committee (506/2018) and Natural and Agricultural Sciences Ethics Committee (180000094) to LB. ...
Article
Full-text available
Three new steroids, turranin M, N and O (1-3), together with four known limonoids, nymania 1 (4), rubralin B (5), aphapolynin C (6) and Trichillia substance Tr B (7), were isolated from the leaves of Turraea obtusifolia. Their chemical structures were elucidated using NMR and MS. Rubralin B (5) displayed good activity against the asexual parasites from the drug sensitive Plasmodium falciparum NF54 strain with an IC50 value of 3.47 µg/mL (4.57 µM), nymania 1 (4) showed a weak activity (IC50 13.36 µg/mL (19.40 µM)) and the rest of compounds had IC50 > 20 µg/mL.
... P. falciparum NF54 cultures were maintained at 37°C in human erythrocytes (O + /A + ) suspended in a complete culture medium (RPMI 1640 medium (Sigma-Aldrich) supplemented with 25 mM HEPES (Sigma-Aldrich), 20 mM D-glucose (Sigma-Aldrich), 200 μM hypoxanthine (Sigma-Aldrich), 0.2% sodium bicarbonate, 24 μg ml −1 Gentamicin (Sigma-Aldrich) and 0.5% AlbuMAX II) in a gaseous environment of 90% N 2 , 5% O 2 , and 5% CO 2 as previously described. [26] In vitro ring-stage, intraerythrocytic P. falciparum cultures (genotyped drug-sensitive strain, 200 μl at 1% hematocrit, 1% parasitemia), were treated with PNAM-Lumefantrine, unconjugated lumefantrine, and PNAM. The controls for this assay included chloroquine diphosphate (1 μM, as positive control) and complete RPMI media (as negative control), and grown for 96 h at 37°C under the 90% N 2 , 5% O 2 , and 5% CO 2 gas mixture in 96-well plates. ...
Article
Full-text available
Uncomplicated malaria is effectively treated with oral artemisinin-based combination therapy (ACT). Yet, there is an unmet clinical need for the intravenous treatment of the more fatal severe malaria. There is no combination intravenous therapy for this form of the disease mainly due to the nonavailability of a water-soluble partner drug for the artemisinin, artesunate. The currently available treatment is a two-part regimen split into an intravenous monotherapy artesunate administered while the patient is admitted to a health centre and followed by the conventional oral ACT that is taken after the patient has been discharged. In a novel application of polymer therapeutics, we conjugated the aqueous insoluble but very important antimalarial lumefantrine to a carrier polymer to create a new water-soluble chemical entity (p-N-acryloylmorpholine-stat-p-acrylic acid-Lumefantrine) suitable for intravenous administration in a clinically relevant formulation. The conjugate is characterized by spectroscopic and analytical techniques, and the aqueous solubility of lumefantrine is determined to have increased by about three orders of magnitude. Pharmacokinetic studies in mice indicates that there is significant release of lumefantrine in the plasma at relatively high concentrations and production of the liver metabolite desbutyl-lumefantrine (area under the curve of metabolite is about 10% that of the parent). In a Plasmodium falciparum severe malaria mouse model, parasitaemia clearance by PNAM-Lumefantrine is 50% higher than that of reference unconjugated lumefantrine. The polymer-lumefantrine shows potential for entering the clinic as a partner drug to artesunate to meet the need for a one-course treatment for severe malaria. This article is protected by copyright. All rights reserved.
... On the other hand, thiourea and isothiourea derivatives were reported to exhibit diverse chemotherapeutic activities including anticancer, 14−16 anti-HIV, 17,18 anti-tuberculosis, 19 and antimalarial activities. 20 In view of the previously reported observations and as a continuation of an interest in chemotherapeutic properties, 21−24 and structural informatics 25−29 of adamantanebased derivatives, we report herein the synthesis, characterization, solid-state structural features, electronic properties, vibrational spectroscopy, and preliminary antimicrobial and antiproliferative activities of two adamantane-linked isothiourea derivatives. ...
Article
Full-text available
In this study, two adamantane-linked isothiourea derivatives containing a common 4-chlorophenyl substituent coupled with 4-nitrobenzyl or 4-bromobenzyl moieties were synthesized. Both derivatives were characterized, in the solid state and in solution, through a synergistic combination of experimental and in silico techniques, and the results are of great value for the chemical and structural characterization of related compounds. The crystal structures of both derivatives were analyzed in depth, including Hirshfeld surface analysis and lattice energy calculations, revealing a predominant dispersive component of the total energy that stabilizes crystal packing. Both compounds showed potent broad-spectrum antibacterial activity and moderate activity against the pathogenic fungus Candida albicans. In addition, in vitro anti-proliferative activity assays showed that the 4-bromobenzyl analogue displays higher activity than the 4-nitrobenzyl one, with IC50 values under 30 μM against five human cancer cell lines. Our results give evidence of the potential of the adamantane/isothiourea combination to render auspicious scaffolds for new potential chemotherapeutic agents.
... Cell Growth Inhibition Assays. Cell growth inhibition assays were performed as previously reported 23,46,47 and are described in the Supporting Information including representative dose−response curves of SQ109 analogs against the protozoan parasites ( Figure S1). ...
Article
SQ109 is a tuberculosis drug candidate that has high potency against Mycobacterium tuberculosis and is thought to function at least in part by blocking cell wall biosynthesis by inhibiting the MmpL3 transporter. It also has activity against bacteria and protozoan parasites that lack MmpL3, where it can act as an uncoupler, targeting lipid membranes and Ca2+ homeostasis. Here, we synthesized 18 analogs of SQ109 and tested them against M. smegmatis, M. tuberculosis, M. abscessus, Bacillus subtilis, and Escherichia coli, as well as against the protozoan parasites Trypanosoma brucei, T. cruzi, Leishmania donovani, L. mexicana, and Plasmodium falciparum. Activity against the mycobacteria was generally less than with SQ109 and was reduced by increasing the size of the alkyl adduct, but two analogs were ∼4-8-fold more active than SQ109 against M. abscessus, including a highly drug-resistant strain harboring an A309P mutation in MmpL3. There was also better activity than found with SQ109 with other bacteria and protozoa. Of particular interest, we found that the adamantyl C-2 ethyl, butyl, phenyl, and benzyl analogs had 4-10× increased activity against P. falciparum asexual blood stages, together with low toxicity to a human HepG2 cell line, making them of interest as new antimalarial drug leads. We also used surface plasmon resonance to investigate the binding of inhibitors to MmpL3 and differential scanning calorimetry to investigate binding to lipid membranes. There was no correlation between MmpL3 binding and M. tuberculosis or M. smegmatis cell activity, suggesting that MmpL3 is not a major target in mycobacteria. However, some of the more active species decreased lipid phase transition temperatures, indicating increased accumulation in membranes, which is expected to lead to enhanced uncoupler activity.
... The in vitro cytotoxicity was evaluated against human Caucasian hepatocellular carcinoma cells (HepG2) as previously described. [50] Cells were cultivated in vitro in complete DMEM media (Hyclone, U.S.A.) supplemented with 10 % (v/v) heat inactivated foetal bovine serum and 1 % (v/v) penicillin/streptomycin at 37°C in 5 % CO 2 . Cells were detached with 0.25 % (v/v) Trypsin-EDTA once~80 % confluency was observed, [51] and cell viability was monitored microscopically with 0.2 % (w/v) Trypan-Blue using a Neubauer chamber. ...
Article
Full-text available
Malaria elimination requires multipronged approaches, including the application of antimalarial drugs able to block human‐to‐mosquito transmission of malaria parasites. The transmissible gametocytes of Plasmodium falciparum seem to be highly sensitive towards epidrugs, particularly those targeting demethylation of histone post‐translational marks. Here, we report exploration of compounds from a chemical library generated during hit‐to‐lead optimization of inhibitors of the human histone lysine demethylase, KDM4B. Derivatives of 2‐([1,1′‐biphenyl]‐4‐carboxamido) benzoic acid, around either the amide or a sulfonamide linker backbone (2‐(arylcarboxamido)benzoic acid, 2‐carboxamide (arylsulfonamido)benzoic acid and N‐(2‐(1H‐tetrazol‐5‐yl)phenyl)‐arylcarboxamide), showed potent activity towards late‐stage gametocytes (stage IV/V) of P. falciparum, with the most potent compound reaching single digit nanomolar activity. Structure‐activity relationship trends were evident and frontrunner compounds also displayed microsomal stability and favourable solubility profiles. Simplified synthetic routes support further derivatization of these compounds for further development of these series as malaria transmission‐blocking agents.
Article
Full-text available
Artemisinin, isolated from the traditional Chinese medicinal plant qīng hāo 青蒿 (Artemisia annua) and its derivatives are used for treatment of malaria. With treatment failures now being recorded for the derivatives and companion drugs used in artemisinin combination therapies new drug combinations are urgently required. The amino-artemisinins artemiside and artemisone display optimal efficacies in vitro against asexual and sexual blood stages of the malaria parasite Plasmodium falciparum and are active against tumour cell lines. In continuing the evolution of combinations of the amino-artemisinins with new drugs, we examine the triterpenoid quinone methide celastrol isolated from the traditional Chinese medicinal plant léi gōng téng 雷公藤 (Tripterygium wilfordii). This compound is redox active, and has attracted considerable attention because of potent biological activities against manifold targets. We report that celastrol displays good IC50 activities ranging from 0.50–0.82 µM against drug-sensitive and resistant asexual blood stage Pf, and 1.16 and 0.28 µM respectively against immature and late stage Pf NF54 gametocytes. The combinations of celastrol with each of artemisone and methylene blue against asexual blood stage Pf are additive. Given that celastrol displays promising antitumour properties, we examined its activities alone and in combinations with amino-artemisinins against human liver HepG2 and other cell lines. IC50 values of the amino-artemisinins and celastrol against HepG2 cancer cells ranged from 0.55–0.94 µM. Whereas the amino-artemisinins displayed notable selectivities (SI > 171) with respect to normal human hepatocytes, in contrast, celastrol displayed no selectivity (SI < 1). The combinations of celastrol with artemiside or artemisone against HepG2 cells are synergistic. Given the promise of celastrol, judiciously designed formulations or structural modifications are recommended for mitigating its toxicity.
Article
Full-text available
A series of symmetrically or unsymmetrically alkyl-substituted polyamine analogues were synthesized containing either a 3-3-3 or 3-7-3 polyamine backbone. These analogues were evaluated in vitro as antitumor agents in the NCI H157 and NCI H82 lung carcinoma cell lines, and as antitrypanosomal agents against four strains of Trypanosoma brucei brucei or Trypanosoma brucei rhodesiense. The resulting biological data suggest that it is possible to specifically target tumor cells or parasitic organisms with alkylpolyamine analogues based on structure.
Article
Full-text available
The recently discovered enzyme lysine-specific demethylase 1 (LSD1) plays an important role in the epigenetic control of gene expression, and aberrant gene silencing secondary to LSD1 overexpression is thought to contribute to the development of cancer. We recently reported a series of (bis)guanidines and (bis)biguanides that are potent inhibitors of LSD1 and induce the re-expression of aberrantly silenced tumor suppressor genes in tumor cells in vitro. We now report a series of isosteric ureas and thioureas that are also potent inhibitors of LSD1. These compounds induce increases in methylation at the histone 3 lysine 4 (H3K4) chromatin mark, a specific target of LSD1, in Calu-6 lung carcinoma cells. In addition, these analogues increase cellular levels of secreted frizzle-related protein (SFRP) 2 and transcription factor GATA4. These compounds represent an important new series of epigenetic modulators with the potential for use as antitumor agents.
Article
Full-text available
Artemisinin combination therapies are the first-line treatments for uncomplicated Plasmodium falciparum malaria in most malaria-endemic countries. Recently, partial artemisinin-resistant P. falciparum malaria has emerged on the Cambodia–Thailand border. Exposure of the parasite population to artemisinin monotherapies in subtherapeutic doses for over 30 years, and the availability of substandard artemisinins, have probably been the main driving force in the selection of the resistant phenotype in the region. A multifaceted containment programme has recently been launched, including early diagnosis and appropriate treatment, decreasing drug pressure, optimising vector control, targeting the mobile population, strengthening management and surveillance systems, and operational research. Mathematical modelling can be a useful tool to evaluate possible strategies for containment.
Article
Full-text available
Inhibition of polyamine biosynthesis and/or the perturbation of polyamine functionality have been exploited with success against parasitic diseases such as Trypanosoma infections. However, when the classical polyamine biosynthesis inhibitor, alpha-difluoromethylornithine, is used against the human malaria parasite, Plasmodium falciparum, it results in only a cytostatic growth arrest. Polyamine metabolism in this parasite has unique properties not shared by any other organism. These include the bifunctional arrangement of the catalytic decarboxylases and an apparent absence of the typical polyamine interconversion pathways implying different mechanisms for the regulation of polyamine homeostasis that includes the uptake of exogenous polyamines at least in vitro. These properties make polyamine metabolism an enticing drug target in P. falciparum provided that the physiological and functional consequences of polyamine metabolism perturbation are understood. This review highlights our current understanding of the biological consequences of inhibition of the biosynthetic enzymes in the polyamine pathway in P. falciparum as revealed by several global analytical approaches. Ultimately, the evidence suggests that polyamine metabolism in P. falciparum is a validated drug target worth exploiting.
Article
Full-text available
Encephalitozoon cuniculi is a microsporidium responsible for systemic illness in mammals. In the course of developing leads to new therapy for microsporidiosis, we found that a bis(phenylbenzyl)3-7-3 analog of spermine, 1,15-bis{N-[o-(phenyl)benzylamino}-4,12-diazapentadecane (BW-1), was a substrate for an E. cuniculi amine oxidase activity. The primary natural substrate for this oxidase activity was N′-acetylspermine, but BW-1 had activity comparable to that of the substrate. As the sole substrate, BW-1 gave linear reaction rates over 15 min and Km of 2 μM. In the presence of N′-acetylspermine, BW-1 acted as a competitive inhibitor of oxidase activity and may be a subversive substrate, resulting in increased peroxide production. By use of 13C-labeled BW-1 as a substrate and nuclear magnetic resonance analysis, two products were determined to be oxidative metabolites, a hydrated aldehyde or dicarboxylate and 2(phenyl)benzylamine. These products were detected after exposure of 13C-labeled BW-1 to E. cuniculi preemergent spore preparations and to uninfected host cells. In previous studies, BW-1 was curative in a rodent model of infection with E. cuniculi. The results in this study demonstrate competitive inhibition of oxidase activity by BW-1 and support further studies of this oxidase activity by the parasite and host.
Article
Plasmodium falciparum can now be maintained in continuous culture in human erythrocytes incubated at 38°C in RPMI 1640 medium with human serum under an atmosphere with 7 percent carbon dioxide and low oxygen (1 or 5 percent). The original parasite material, derived from an infected Aotus trivirgatus monkey, was diluted more than 100 million times by the addition of human erythrocytes at 3- or 4-day intervals. The parasites continued to reproduce in their normal asexual cycle of approximately 48 hours but were no longer highly synchronous. They have remained infective to Aotus.
Article
Artemisinin combination therapies are the first-line treatments for uncomplicated Plasmodium falciparum malaria in most malaria-endemic countries. Recently, partial artemisinin-resistant P. falciparum malaria has emerged on the Cambodia-Thailand border. Exposure of the parasite population to artemisinin monotherapies in subtherapeutic doses for over 30 years, and the availability of substandard artemisinins, have probably been the main driving force in the selection of the resistant phenotype in the region. A multifaceted containment programme has recently been launched, including early diagnosis and appropriate treatment, decreasing drug pressure, optimising vector control, targeting the mobile population, strengthening management and surveillance systems, and operational research. Mathematical modelling can be a useful tool to evaluate possible strategies for containment.
Chapter
tert ‐Butoxycarbonylation of amino acids and their derivatives: N‐tert ‐butoxycarbonyl‐l‐phenylalanine reactant: 223 g (1 mol) of di‐tert‐butyl dicarbonate product: N‐tert‐butoxycarbonyl‐L‐phenylalanine product: Boc‐Ala‐OH product: Boc‐β‐Ala‐OH product: Boc‐Arg‐OH product: Boc‐Arg(NO2)‐OH product: Boc‐Asn‐OH product: Boc‐Asp(OBzl)‐OH product: Boc‐Cys(Bzl)‐OH product: (Boc‐Cys‐OH)2 product: Boc‐Gln‐OH product: Boc‐Glu(OBzl)‐OH product: Boc‐Gly‐OH product: Boc‐His(Boc)‐OH product: Boc‐Ile‐OH product: Boc‐Leu‐OH product: Boc‐Lys(Boc)‐OH product: Boc‐Lys(CBZ)‐OH product: Boc‐Met‐OH product: Boc‐Met‐OH product: Boc‐Pro‐OH product: Boc‐Ser‐OH product: Boc‐Ser(Bzl)‐OH product: Boc‐Thr‐OH product: Boc‐Trp‐OH product: Boc‐Trp‐(FOR)‐OH product: Boc‐Tyr‐OH product: Boc‐Tyr‐OH product: Boc‐Tyr(Bzl)‐OH product: Boc‐Tyr(2,6‐Cl2‐Bzl)‐OH product: Boc‐Val‐OH
Article
The development, synthesis and use of S-2-naphthylmethyl thioacetimidate hydrobromide 7 as a highly reactive reagent for the mild conversion of amines to substituted acetamidines in a nonodorous process is described.