Sunteți pe pagina 1din 7

Bioorganic & Medicinal Chemistry 24 (2016) 4492–4498

Contents lists available at ScienceDirect

Bioorganic & Medicinal Chemistry


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

Design, synthesis and anti-P. falciparum activity of pyrazolopyridine–


sulfonamide derivatives
Thais B. Silva a,b, Alice M. R. Bernardino b, Maria de Lourdes G. Ferreira a, Kamilla R. Rogerio a,c,d,
Leonardo J. M. Carvalho c, Nubia Boechat a, Luiz C. S. Pinheiro a,⇑
a
Departamento de Síntese de Fármacos, Instituto de Tecnologia em Fármacos, Farmanguinhos—FIOCRUZ, Fundação Oswaldo Cruz, Rua Sizenando Nabuco 100, Manguinhos,
Rio de Janeiro, RJ 21041-250, Brazil
b
Programa de Pós-Graduação em Química, Departamento de Química Orgânica, Universidade Federal Fluminense, Niterói, RJ, Brazil
c
Laboratório de Pesquisa em Malária, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
d
Programa de Pós Graduação em Química, PGQu Instituto de Química, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil

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

Article history: Ten 1-phenyl-1H-pyrazolo[3,4-b]pyridine derivatives connected by a linker group to benzenesulfon-


Received 24 June 2016 amide moieties with different substituents in the 4-position were synthesized and assayed against
Revised 20 July 2016 Plasmodium falciparum. These ten compounds exhibited activity in vitro against the chloroquine-resistant
Accepted 22 July 2016
clone W2 with IC50 values ranging from 3.46 to 9.30 lM. The most active derivatives with substituent
Available online 25 July 2016
R2 = Cl or CH3 at the benzenesulfonamide moiety exhibited the lowest IC50. Compounds with an
R1 = CO2Et substituent at the 5-position of the 1H-pyrazolo[3,4-b]pyridine ring presented lower activity
Keywords:
than those with a CN substituent. The 1H-pyrazolo[3,4-b]pyridine system appears to be promising for
Malaria
Pyrazolopyridine
further studies as an antimalarial for overcoming the burden of resistance in P. falciparum.
Sulfonamide Ó 2016 Elsevier Ltd. All rights reserved.
Plasmodium falciparum

1. Introduction against malaria, including a fixed dose of artemisinin-combined


therapies (ACTs) to treat simple and severe falciparum malaria,
Over the past fifteen years (2000 to 2015), there has been a sig- using artemether plus lumefantrine, artesunate plus amodiaquine
nificant reduction in the number of cases and deaths caused by or mefloquine, dihydroartemisinin plus piperaquine or artesunate
malaria according to data from the World Health Organization. In plus sulfadoxine-pyrimethamine, among other ACTs.3 Combination
2000, it was estimated that there were 262 million cases of malaria therapy is intended to prevent or delay the onset of resistance with
worldwide, leading to 839,000 deaths. In 2015, the number of the simultaneous use of drugs that have different modes of action.
malaria cases decreased to 214 million, and the number of deaths An artemisinin derivative with a quick and effective action is used,
decreased to 438,000. These figures represent an 18% decline in however, if a parasite-resistant mutant of the drugs arises during
cases of malaria and a 48% decrease in the number of deaths during the course of infection is rapidly cleared, allowing a longer lasting
this period.1 The progress made in reducing the number of deaths drug to kill the parasite that survived the initial application of the
in children under 5 years of age has been substantial; however, it is artemisinin derivative.3
still high, and malaria remains a major cause of death in children There is an urgent need for novel antimalarials with better
corresponding to one death every 2 min.1 safety profiles than current medicines in addition to new drugs
In addition to efforts to control the mosquito vector and the that prevent transmission and relapse due to the resistance against
search for an effective vaccine, developing malaria drugs is still quinoline derivatives and artemisinins.4,5 In the recent literature, a
the main disease control strategy due to the spread of parasites number of new antimalarial compounds in different states of
that are resistant to antimalarial drugs.2 preclinical and clinical development have been described.6–8
Due to the high parasitic resistance exhibited by Plasmodium Quinoline derivatives are still the dominate class of antimalarial
falciparum to most available drugs, monotherapy is no longer used compounds;9,10 however, due to resistance to this class of drugs,
to treat malaria. The WHO recommends the use of first-line drugs searching for other analogous compounds is extremely important.
The pyrazolopyridine ring is an isostere of quinoline, and the
literature has demonstrated that this system possesses anti-
⇑ Corresponding author. Tel.: +55 21 3977 2456; fax: +55 21 2560 2518.
inflammatory,11 anti-cancer,12 antibacterial13,14 and antiviral15–18
E-mail address: lpinheiro@far.fiocruz.br (L.C.S. Pinheiro).

http://dx.doi.org/10.1016/j.bmc.2016.07.049
0968-0896/Ó 2016 Elsevier Ltd. All rights reserved.
T. B. Silva et al. / Bioorg. Med. Chem. 24 (2016) 4492–4498 4493

activities. The literature suggests that pyrazolopyridine is a poten- The 7-chloroquinoline moiety was replaced by the system
tial system with antiparasitic activity against Trypanosoma cruzi,19 1-phenyl-1H-pyrazolo[3,4-b]pyridine by ring isosterism. An
as well as antileishmanial20 and antimalarial activities.21,22 N-(4-aminobutyl)benzenesulfonamide group was attached at the
In our previous search for new drugs against malaria, we 4 position of the heterocyclic ring. The 1H-pyrazolo[3,4-b]pyridine
demonstrated the importance of MEFAS (I), a hybrid salt active ring remains separated from the benzenesulfonamide moiety by
against the P. falciparum W2 clone that presents an IC50 value of the linker containing four methylene groups, similar to that found
0.001 lM.23 in the individual molecular framework of the precursor V.
The derivative 2-(trifluoromethyl)[1,2,4]triazolo[1,5-a]pyrim-
idine II was active against P. falciparum W2 with an 2. Results and discussion
IC50 = 0.023 lM and showed no toxicity. In this work, the impor-
tance of the CF3 group at position 2 of the nucleus triazolo[1,5-a] 2.1. Chemistry
pyrimidine for increased activity was demonstrated.24
A series of quinoline derivatives containing a 1,2,3-triazole ring The synthetic route for preparing the N-(4-((1-phenyl-1H-pyra-
at position 4 were synthesized and tested against strains of zolo[3,4-b]pyridin-4-yl)amino)butyl)benzenesulfonamides (1–10)
P. falciparum W2. Compound III was the most active, with an is shown in Scheme 1.
IC50 = 1.4 lM.25 Ethyl 5-amino-1-phenyl-1H-pyrazole-4-carboxylate (13) could
We have recently synthesized a new class of hybrids of atorvas- easily be prepared in 80% yield from the reaction of phenylhy-
tatin (AVA) and aminoquinolines (primaquine and chloroquine drazine (11) (0.01 mol) and ethyl 2-cyano-3-ethoxyacrylate (12)
derivatives). The quinolinic moiety was connected to the pentasub- (0.01 mol), in ethanol under reflux. The derivative 5-amino-1-phe-
stituted pyrrole from AVA by a linker group. The activity of the nyl-1H-pyrazol (14) was obtained in 85% yield via a hydrolysis
compounds increased with the size of the carbon chain. Compound reaction followed by decarboxylation of the derivative ethyl
IV with four methylene groups and a 7-chloroquinolinyl moiety 5-amino-1-phenyl-1H-pyrazole-4-carboxylate (13) with phospho-
exhibited better activity than CQ with an IC50 of 0.40 lM.26 ric acid (85%).
New derivatives of quinoline-sulfadoxine hybrids were planned The Michael addition of 5-aminopyrazole (14) with ethyl
by molecular hybridization between the quinoline ring and the 2-cyano-3-ethoxyacrylate (12) or diethyl 2-(ethoxymethylene)-
benzenesulfonamide moiety present in chloroquine and sulfadox- malonate (15) in ethanol under reflux afforded the derivatives 16
ine, which are drugs used in the treatment of malaria. All 15 com- and 17 in 86% and 88% yield, respectively.
pounds in this series were active in vitro against the P. falciparum Ethyl 4-chloro-1-phenyl-1H-pyrazolo[3,4-b]pyridine-5-car-
W2 clone, and none of these compounds were toxic to BGM cells. boxylate (18) was prepared from diethyl 2-(((1-phenyl-1H-pyra-
Ten compounds presented IC50 values lower than that of CQ, with zol-5-yl)amino)methylene)malonate (16) through a reaction with
values ranging from 0.05 to 0.40 lM. The most active hybrids have phosphorous oxychloride under reflux in 84% yield as described
a structural relationship between the four methylene groups used in the literature.16,20 However, the derivative 4-chloro-1-phenyl-
as linkers between the quinoline ring and the benzenesulfonamide 1H-pyrazolo[3,4-b]pyridine-5-carbonitrile (19) cannot be obtained
moiety. Four compounds exhibited SI values (3386.0–1031.3) using this methodology.
higher than that of chloroquine (834.74). When evaluated against The cyclization of pyrazole (17) was performed by refluxing in
Plasmodium berghei malaria, compound V was the most active dowtherm (250 °C) for 40 min and isolating 4-oxo-1-phenyl-4,7-
and inhibited parasitemia by 49% on day 5 after inoculation, con- dihydro-1H-pyrazolo[3,4-b]pyridine-5-carbonitrile (22) by precip-
tributing to the discovery of new prototypes with antimalarial itating from hexane with a yield of 84%.28 The derivative (22) was
activity (Fig. 1).27 refluxed with phosphorous oxychloride to produce (19) in 90%
In an effort to enhance the anti-P. falciparum activity of the yield.
precursor quinoline-sulfonamide hybrid V, new derivatives The compounds ethyl 4-((4-aminobutyl)amino)-1-phenyl-1H-
N-(4-((1-phenyl-1H-pyrazolo[3,4-b]pyridin-4-yl)amino)butyl)ben- pyrazolo[3,4-b]pyridine-5-carboxylate (20) and 4-((4-aminobutyl)
zenesulfonamides (1–10) were designed (Fig. 2). amino)-1-phenyl-1H-pyrazolo[3,4-b]pyridine-5-carbonitrile (21)

H H O
N H H
O
OH N
O
O H N
H H N N
O
O N
N
O N CF3 F3 C
O- N
CF3 N Cl N
O
(I) (II) (III)
IC50 = 0.001 µM IC 50 0.023 µM IC 50 1.4 µM

O Cl
HN
H
Cl N N
N H HN S
N O O

(IV) Cl N (V)
IC 50 0.4 µM IC 50 = 0.09 µM
F

Figure 1. Structures of compounds I–V.


4494 T. B. Silva et al. / Bioorg. Med. Chem. 24 (2016) 4492–4498

NH2
O
N S
HN O
O NH

O N
Cl N N
Cl
chloroquine sulfadoxine

H
N
HN S
O
O
isosteric ring
replacement
Cl N remained the group
linker benzenesulfonamide
(V)

R2

H
N
HN S
O
R1 O
N
N N R1 = CO2 Et, CN
R2 = H, F, Cl, CH 3
(1-10)

Figure 2. Rational approach to the design of compounds 1–10.

NHNH 2
CO2Et
R1
N NH 2 N
N N EtO CO2 Et N N
(i) NH 2 (ii) (iii) H CO2 Et
N
11 + 17 R 1 = CN
R1
+ (vii)
O
EtO CO 2Et 13 14 15 R 1 = CO2 Et;
12 R 1 = CN CN
16 R 1 = CO2 Et
CN 12 N
(iv) N
R2 N
H

NH2 Cl
H HN 22
N R1 (viii)
HN S (vi) R1 (v)
R1 O O N
N
N N N
N N
N N
R 1 = CO2 Et R1 = CN
20 R 1 = CO2 Et; 18 R 1 = CO2 Et;
1 R 2 = H; 6 R2 = H; 21 R 1 = CN 19 R 1 = CN
2 R 2 = CH 3; 7 R2 = CH3 ;
3 R 2 = F; 8 R2 = F;
4 R 2 = Cl; 9 R2 = Cl;
5 R 2 = OCH 3 10 R2 = OCH3

Scheme 1. Synthetic route used to prepare compounds 1–10. Reagents and conditions: (i) EtOH, reflux, 1 h, 70%; (ii) H3PO4, 170 °C, 6 h, 86%; (iii) EtOH, reflux, 2 h, 86–88%;
(iv) POCl3, reflux, 6 h, 84%; (v) butane-1,4-diamine, 1,4-dioxane, 25–80 °C, 1–4 h, 66–79%; (vi) appropriate sulfonyl chloride, MeOH, TEA, 25 °C, 24 h, 59–70%; (vii) dowtherm,
250 °C, 40 min, 71%; (viii) POCl3, reflux, 6 h, 90%.

were synthesized in 79% and 66% yield, respectively, by the nucle- The values are summarized in Table 1. All the compounds that
ophilic substitution reaction between 4-chloro-1-phenyl-1H-pyra- presented activity showed IC50 values ranging from 3.46 to
zolo[3,4-b]pyridines (18, 19) (1.0 mmol) and butane-1,4-diamine 9.30 lM in the anti-HPR2 assay, lower than that of sulfadoxine
(1.0 mmol).26,27 (IC50 = 15.0 lM); however, these compounds were not more active
The addition–elimination reaction between the appropriate than chloroquine (IC50 = 0.55 lM).
sulfonyl chloride (1.2 mmol) and amines (20, 21) (1.0 mmol) was Compounds 2, 7 with substituent (R2 = CH3) and 4 (R2 = Cl) at
performed in MeOH and TEA (1.0 mmol) at 25 °C to obtain the tar- the 4-position of the benzenesulfonamide moiety were the most
get compounds N-(4-((1-phenyl-1H-pyrazolo[3,4-b]pyridin-4-yl) active against P. falciparum, presenting IC50 values of 3.46 lM (2)
amino)butyl)benzenesulfonamides (1–10) in 70–59% yield.27 and 3.60 lM (4 and 7). Compound 4 also showed the highest SI
The 10 derivatives 1-phenyl-1H-pyrazolo[3,4-b]pyridine (1–10) values: >277.77.
that were synthesized with different substituents in the 4-position Comparing the series in which substituents R1 = CO2Et and CN
of the benzenesulfonamide group were tested against the W2 at the 5-position of the 1H-pyrazolo[3,4-b]pyridine ring, it can be
chloroquine and sulfadoxine-resistant P. falciparum clone and for observed that the 3 compounds with R1 = CO2Et showed higher
cytotoxicity (Table 1). activity than those with CN. Also is observed in the activity values
T. B. Silva et al. / Bioorg. Med. Chem. 24 (2016) 4492–4498 4495

Table 1
Evaluation of activity against P. falciparum parasites (W2 clone) resistant to chloroquine, cytotoxicity tests in normal monkey kidney cell line (BGM) and the selectivity index (SI)
of compounds 1–10, 20 and 21, chloroquine and sulfadoxine

Compounds Anti-P. falciparum activity IC50* (lM) Cytotoxicity to BGM cells MLD50** (lM) SI[MLD50/IC50]
1 R1 = CO2Et/R2 = H 6.52 ± 0.8 822.00 126.07
2 R1 = CO2Et/R2 = CH3 3.46 ± 0.25 596.00 172.25
3 R1 = CO2Et/R2 = F 4.12 ± 0.48 993.00 241.01
4 R1 = CO2Et/R2 = Cl 3.60 ± 2.04 >1000.00 >277.77
5 R1 = CO2Et/R2 = OCH3 9.30 ± 1.25 995.00 106.98
6 R1 = CN/R2 = H 4.20 ± 1.07 248.00 59.04
7 R1 = CN/R2 = CH3 3.60 ± 0.09 682.00 189.44
8 R1 = CN/R2 = F 9.30 ± 2.95 508.00 54.62
9 R1 = CN/R2 = Cl 7.50 ± 0.09 418.00 55.73
10 R1 = CN/R2 = OCH3 5.00 ± 1.15 498.00 99.60
20 R1 = CO2Et 2.10 ± 0.08 930.00 442.85
21 R1 = CN 5.00 ± 1.90 503.00 100.60
Chloroquine 0.55 385.00 700.00
Sulfadoxine 15.00 310.00 20.70
*
IC50 = inhibitory concentration for 50% of parasite growth, evaluated in three different experiments for each test (±standard deviation (SD)).
**
Minimal lethal dose for 50% of cells (MLD50), used to calculate the selectivity index.

of the intermediates 20 and 21 with IC50values of 2.10 and 5.00, chromatography was performed using silica gel 60
respectively. In fact, the amine 20 exhibited greater activity than (0.040–0.063 mm). The remaining reagents and solvents that were
the sulfonamides (1–10) and the highest SI value (442.85). used were of analytical grade.

3. Conclusions 4.2. Synthesis

Among the 1-phenyl-1H-pyrazolo[3,4-b]pyridine derivatives 4.2.1. Procedure for preparing ethyl 5-amino-1-phenyl-1H-pyra-
(1–10) synthesized with different substituents at the 4-position zole-4-carboxylate (13)
of the benzenesulfonamide moiety, all exhibited anti-P. falciparum A mixture of phenylhydrazine (9 mmol) and 10 mL of ethanol
activity in vitro against W2 chloroquine-resistant parasites and was stirred and allowed to reflux for 1 h. Then, ethyl
low toxicity to BGM cells. (ethoxymethylene)cyanoacetate (9 mmol) dissolved in 10 mL of
The ten compounds (1–10) presented IC50 values lower than ethanol was slowly added. The reaction mixture was refluxed for
that of the control drug sulfadoxine, with IC50 values ranging from 20 min. The reaction mixture was poured into 50 mL of ice-cold
3.46 to 9.30 lM. However, the observed activity was lower than water. The precipitate was collected by filtration and washed with
that of chloroquine and the quinolone-sulfonamide hybrid V. water to afford 13 in 70% yield.
Derivatives 2, 4 and 7 exhibited the lowest activity against Yield: 70%. Mp: 98 °C. IR (KBr. cm1): 3400–3140; 2995; 2912;
P. falciparum, with IC50 values of 3.46 lM (2) and 3.60 lM (4 and 1677; 1621; 1552; 1528; 1280; 781–697. 1H NMR (400 MHz,
7). Compounds with an R1 = CO2Et substituent at the 5-position CDCl3, TMS, d in ppm): 1.36 (t, 3H, J = 7.1 Hz; CH2CH3); 4.30 (q,
of the 1H-pyrazolo[3,4-b]pyridine ring showed higher activity than 2H, J = 7.1 Hz CH2CH3); 5.31 (s, 2H, NH2); 7.41–7.37 (m, 1H, H40 );
those with a CN substituent. The amine 20 showed higher activity 7.55–7.48 (m, 4H, H20 , H30 , H50 , H60 ); 7.78 (s, 1H, H3). 13C NMR
than the sulfonamides (1–10) with an IC50 = 2.10 lM and the high- (100 MHz, CDCl3, TMS, d in ppm): 14.5; 59.6; 96.2; 123.8; 128.1;
est SI value (442.85). 129.7; 137.6; 140.6; 149.0; 164.6. EI [M1] 230.2.
The 1H-pyrazolo[3,4-b]pyridine system appears to be promis-
ing for further studies as an antimalarial with the objective of over- 4.2.2. Procedure for preparing 1-phenyl-1H-pyrazol-5-amine
coming the burden of resistance in P. falciparum. (14)
A mixture of 1.50 g (6 mmol) of ethyl 5-amine-1-phenyl-1H-
4. Experimental pyrazole-4-carboxylate (13) and 20 mL of 85% phosphoric acid
was maintained under reflux and stirring for 6 h at 170 °C. After
4.1. Chemistry completion of the reaction, 50 mL of crushed ice was added, and
then the mixture was basified to pH 7 with NaOH (6 M aq). The
The 1H, 13C and 19F nuclear magnetic resonance (NMR) spectra mixture was extracted with dichloromethane (3  30 mL). The
were obtained at 400.00, 100.00 and 376.00 MHz, respectively, combined organic solution was washed with water (3  50 mL),
using a BRUKER Avance instrument equipped with a 5 mm probe. dried (anhydrous sodium sulfate), filtered, and concentrated under
Tetramethylsilane was used as an internal standard. The chemical vacuum to afford 14 as a brown oil.
shifts (d) are reported in ppm, and the coupling constants (J) are Yield: 86%. IR (KBr. cm1): 3418–3182; 1618; 1597; 1550;
reported in Hertz. Electron-ionization mass spectra (EI-MS, scan 760–694. 1H NMR (400 MHz, CDCl3, TMS, d in ppm): 3.84 (s, 2H,
ES+ capillary (3.0 kV)/cone (30 V)/extractor (1 V)/RF lens NH2); 5.61 (d, 1H, J = 2.0 Hz, H4); 7.42 (d, 1H, J = 1.8 Hz, H3);
(1.0 V)/source temperature (150 °C)/desolvation temperature 7.57–7.32 (m, 5H, H20 , H30 , H40 , H50 , H60 ). 13C NMR (100 MHz,
(300 °C)) were recorded using a Micromass/Waters Spectrometer CDCl3, TMS, d in ppm): 90.6; 123.9; 127.4; 129.4; 138.6; 140.3;
(model: ZQ-4000). HRMS data were obtained using LC-MS Bruker 144.7. EI [M1] 157.8.
Daltonics MicroTOF (time of flight analyzer). Fourier transform
infrared (FT-IR) absorption spectra were recorded on a Shimadzu 4.2.3. General procedure for preparing diethyl 2-(((1-phenyl-1H-
mode IR Prestige-21 spectrophotometer through KBr reflectance. pyrazol-5-yl)amino)methylene)malonate (16), and (E)-ethyl 2-cy-
The melting points (mp) were determined using a Büchi model ano-3-((1-phenyl-1H-pyrazol-5-yl)amino)acrylate (17)
B-545 apparatus. TLC (thin layer chromatography) was performed A mixture of 11.13 g (7 mmol) of 1-phenyl-1H-pyrazol-5-amine
using a silica gel F-254 glass plate (20  20 cm). Column 14 and 7 mmol of diethyl 2-(ethoxymethylene)malonate or ethyl
4496 T. B. Silva et al. / Bioorg. Med. Chem. 24 (2016) 4492–4498

2-cyano-3-ethoxyacrylate was maintained under reflux and stir- Yield: 90%. Mp: 146 °C. IR (KBr. cm1): 3110; 2229; 1593; 1463.
1
ring for 2 h. After completion of the reaction, the reaction mixture H NMR (400 MHz, DMSO-d6, TMS, d in ppm): 7.47–7.43 (m, 1H,
was poured into 50 mL of ice-cold water. The precipitate was col- H40 ); 7.63–7.59 (m, 2H, H30 , H50 ); 8.14–8.11 (m, 2H, H20 , H60 );
lected by filtration and washed with water to afford 16 and 17 in 8.81 (s, 1H, H3); 9.08 (s, 1H, H6). 13C NMR (100 MHz, DMSO-d6,
yields of 88% and 86%, respectively. TMS, d in ppm): 103.4; 114.9; 115.8; 121.5; 127.4; 129.3; 134.1;
137.7; 149.9; 141.3; 152.5. EI [M+23]+ 276.9.
4.2.3.1. Diethyl 2-(((1-phenyl-1H-pyrazol-5-yl)amino)methylene)-
malonate (16). Yield: 88%. Mp: 86 °C. IR (KBr. cm1): 3135; 4.2.7. General procedure for preparing 4-[(4-aminobutyl)amino]-
3105; 2976; 2904; 1683; 1638; 1594; 1556; 1513; 1272. 1H NMR 1-phenyl-1H-pyrazolo[3,4-b]pyridines (20, 21)
(400 MHz, CDCl3, TMS, d in ppm): 1.30 (t, J = 7.1 Hz, 3H, CH2CH3); A solution of 1 g of derivative 18 or 19 (3.3 mmol or 3.9 mmol,
4.23 (q, J = 7.1 Hz, 2H, CH2CH3); 6.19 (d, 1H, J = 1.9 Hz, H4); 7.58– respectively) in 20 mL of 1,4-dioxane was added dropwise to
7.45 (m, 5H, H20 , H30 , H40 , H50 , H60 ); 7.61 (d, 1H, J = 1.9 Hz, H3); 0.8 mL (8.2 mmol) of butane-1,4-diamine and stirred at 80 °C for
7.64 (d, 1H, J = 12.7 Hz, Hb); 10.89 (d, J = 12.4 Hz, NH). 13C NMR 4 h. The reaction mixture was poured into 50 mL of ice-cold water.
(100 MHz, CDCl3, TMS, d in ppm): 14.1; 61.6; 78.2; 94.0; 124.3; The precipitate was collected by filtration and washed with ethyl
128.9; 129.9; 137.1; 138.6; 140.6; 152.7; 166.7. EI [M1] 328.2. ether to afford 20 and 21 in yields of 79% and 66%.

4.2.3.2. Ethyl 2-cyano-3-((1-phenyl-1H-pyrazol-5-yl)amino)acry- 4.2.7.1. Ethyl 4-[(4-aminobutyl)amino]-1-phenyl-1H-pyrazolo


late (17). Yield: 86%. Mp: 169 °C. IR (KBr. cm1): 3175–3140; [3,4-b]pyridine-5-carboxylate (20). Yield: 79%. Mp: 149 °C.
3068; 2992; 2939; 2212; 1667; 1621; 1559; 1511; 1238. 1H NMR IR (KBr. cm1): 3271–3239; 2979; 2867; 1667; 1578–1473;
(400 MHz, CDCl3, TMS, d in ppm): 1.33–1.27 (m, 3H, CH2CH3); 1266. 1H NMR (400 MHz, DMSO-d6, TMS, d in ppm): 1.34 (t, 3H,
4.26–4.19 (m, 2H, CH2CH3); 6.21 (d, 1H, J = 1.9 Hz, H4); 7.57–7.43 J = 7.1 Hz, CH2CH3); 1.80–1.72 (m; 4H, H9, H10); 2.84 (m, 2H,
(m, 5H, H20 , H30 , H40 , H50 , H60 ); 7.61 (d, 1H, J = 1.9 Hz, H3); 8.22 H11); 3.73 (m, 2H, H8); 4.31 (q, 2H, J = 7.1 Hz, CH2CH3);
(d, 1H, J = 12.9 Hz, Hb); 11.03 (d, J = 12.8 Hz, NH). 13C NMR 7.40–7.34 (m, 1H, H40 ); 7.58–7.53 (m, 2H, H30 –H50 ); 8.19–8.16
(100 MHz, CDCl3, TMS, d in ppm): 14.2; 14.3; 60.4; 60.7; 94.0; (m, 2H, H20 –H60 ); 8.56 (s, 1H, H3); 8.77 (s, 1H, H6); 9.13 (t; 1H,
96.0; 124.3; 128.6; 130.0; 137.5; 140.0; 140.6; 152.5; 165.0; J = 5.2 Hz, NH). 13C NMR (100 MHz, DMSO-d6, TMS, d in ppm):
168.2. EI [M1] 280.8. 14.0; 25.2; 25.7; 38.5; 44.0; 60.2; 99.3; 104.0; 121.3; 126.2;
128.9; 136.1; 138.7; 151.4; 152.4; 152.5; 167.8. HRMS (ESI) calcd
4.2.4. Procedure for preparing ethyl 4-chloro-1-phenyl-1H-pyra for C19H23N5O2 354.1852, found [M+1]+ 354.1930.
zolo[3,4-b]pyridine-5-carboxylate (18)
To 1 g (3 mmol) of derivative 16 was added 10 mL of phospho- 4.2.7.2. 4-[(4-Aminobutyl)amino]-1-phenyl-1H-pyrazolo[3,4-b]
rus oxychloride. The mixture was stirred under reflux for 6 h. pyridine-5-carbonitrile (21). Yield: 66%. Mp: 102 °C. IR
Excess solvent was removed under reduced pressure, and the (KBr. cm1): 3302; 2939; 2861; 2214; 1581–1460. 1H NMR
resulting material was carefully added to 50 mL of crushed ice. (400 MHz, DMSO-d6, TMS, d in ppm): 1.55–1.50 (m; 1H, H10);
The mixture was then basified to pH 7 with NaOH (6 M aq) and 1.77–1.72 (m, 1H, H9); 2.64–2.60 (m, 1H, H11); 3.70–3.67 (m,
stirred for 40 min. The mixture was diluted with water (30 mL) 1H, H8); 7.38–7.34 (m, 1H, H40 ); 7.54 (t, 2H, J = 7.6 Hz, H30 –H50 );
and extracted with chloroform (3  30 mL). The combined organic 8.14 (d, 2H, J = 7.6 Hz, H20 –H60 ); 8.36 (s, 1H, H6); 8.53 (s, 1H,
solution was washed with water (3  50 mL), dried (anhydrous H3). 13C NMR (100 MHz, DMSO-d6, TMS, d in ppm): 26.2; 29.7;
sodium sulfate), filtered, and concentrated under vacuum to afford 40.9; 118.5; 121.4; 126.4; 126.6; 128.9; 129.9; 138.5; 156.54;
14 in 86% yield. 157.7; 160.9. HRMS (ESI) calcd for C17H18N6 306.1593, found [M
Yield: 84%. Mp: 97 °C. IR (KBr. cm1): 3084; 2982; 2942; 1718; +1]+ 307.1668.
1591; 1463; 1178. 1H NMR (400 MHz, DMSO-d6, TMS, d in ppm):
1.38 (t, 3H, J = 7.1 Hz, CH2CH3); 4.40 (q, 2H, J = 7.1 Hz, CH2 CH3); 4.2.8. General procedure for preparing N-(4-((1-phenyl-1H-pyra-
7.45–7.41 (m, 1H, H40 ); 7.63–7.58 (m, 2H, H30 , H50 ); 8.18–8.14 zolo[3,4-b]pyridine-4-yl)amino)butyl)benzenesulfonamides (1–
(m, 2H, H20 , H60 ); 8.67 (s, 1H, H3); 9.09 (s, 1H, H6). 13C NMR 10)
(100 MHz, DMSO-d6, TMS, d in ppm): 13.9; 61.6; 116.9; 119.1; The amine 20 or 21 (1.4 mmol) was treated with the appropri-
121.1; 126.7; 129.2; 134.4; 138.1; 138.1; 150.2; 151.6; 163.2. EI ate sulfonyl chloride (2.8 mmol) in MeOH as the solvent and TEA
[M1] 302.1. (1 eq.). The reaction mixture was maintained under stirring at
70 °C for 24 h and was poured into ice-cold water (50 mL). The pre-
4.2.5. Procedure for preparing 4-oxo-1-phenyl-4,7-dihydro-1H- cipitate was filtered and dried. The residual crude product was
pyrazolo[3,4-b]pyridine-5-carbonitrile (22) purified via silica gel column chromatography using a gradient
The cyclization of 1.0 g (3.5 mmol) of 17 was performed by mixture of chloroform/methanol. Compounds 1–10 were obtained
refluxing in 15 mL of dowtherm at 250 °C for 40 min. After comple- as white solids in 59–70% yield.
tion of the reaction, the reaction mixture was cooled and then
poured into 50 mL of hexane. The precipitate was filtered and dried 4.2.8.1. Ethyl 1-phenyl-4-((4-(phenylsulfonamido)butyl)amino)-
to afford 22 in 71% yield. 1H-pyrazolo[3,4-b]pyridine-5-carboxylate (1). Yield: 60%.
Yield: 71%. Mp: 223 °C. IR (KBr. cm1): 3124; 2229; 1661; 1583; Mp: 52–54 °C. IR (KBr. cm1): 3290–3247; 2982–2872; 1682;
1497. 1H NMR (400 MHz, CDCl3, TMS, d in ppm): 7.45–7.41 (m, 1H, 1582; 1370; 1181; 1266; 1156. 1H NMR (400 MHz, DMSO-d6,
H40 ); 7.61–7.57 (m, 2H, H30 , H50 ); 8.06 (d, 1H, J = 7.9 Hz, H20 , H60 ); TMS, d in ppm): 1.34 (t, 3H, J = 7.1 Hz, CH2CH3); 1.60–1.53 (m, 2H,
8.53 (s, 1H, H3); 8.64 (s, 1H, H6). 13C NMR (100 MHz, CDCl3, TMS, d H10); 1.74–1.67 (m, 2H, H9); 2.86–2.81 (m, 2H, H11); 3.67–3.63
in ppm): 91.5; 108.4; 116.1; 121.9; 127.1; 129.2; 134.2; 138.0; (m, 2H, H8); 4.31 (q, 2H, J = 7.1 Hz, CH2CH3); 7.36 (t, 1H,
151.8; 164.4. EI [M1] 234.9. J = 7.4 Hz, H40 ); 7.62–7.54 (m, 5H, H30 –H50 , H400 , H300 –H500 or
H200 –H600 ); 7.68 (t, 1H, J = 5.8 Hz, SO2NH); 7.80–7.78 (m, 2H,
4.2.6. Procedure for preparing 4-chloro-1-phenyl-1H-pyrazolo H200 –H600 or H300 –H500 ); 8.19–8.17 (m, 2H, H20 –H60 ); 8.49 (s, 1H,
[3,4-b]pyridine-5-carbonitrile (19) H3); 8.76 (s, 1H, H6); 9.07 (t, 1H, J = 5.2, NH). 13C NMR (100 MHz,
To 1 g (4.2 mmol) of derivative 22 was used the same procedure DMSO-d6, TMS, d in ppm): 14.1; 25.8; 26.4; 42.1; 44.0; 60.2; 99.3;
described in Section 4.2.1. 104.0; 121.3; 126.2; 126.3; 129.1; 129.9; 132.2; 136.1; 138.7;
T. B. Silva et al. / Bioorg. Med. Chem. 24 (2016) 4492–4498 4497

140.4; 151.3; 152.3; 152.5; 167.7. HRMS (ESI) calcd for C25H28N5O4S HRMS (ESI) calcd for C26H29N5O5S 523.1889, found [M+1]+
493.1784, found [M+1]+ 494.1871. HPLC: 98%. 524.1972. HPLC: 100%.

4.2.8.2. Ethyl 4-((4-(4-methylphenylsulfonamido)butyl)amino)-1- 4.2.8.6. N-(4-((5-Cyano-1-phenyl-1H-pyrazolo[3,4-b]pyridin-4-yl)


phenyl-1H-pyrazolo[3,4-b]pyridine-5-carboxylate (2). Yield: amino)butyl)benzenesulfonamide (6). Yield: 67%. Mp: 158–
66%. Mp: 130–132 °C. IR (KBr. cm1): 3293–3068; 2963–2851; 159 °C. IR (KBr. cm1): 3305; 2941–2876; 2213; 1592–1422; 1320;
1682; 1578; 1328; 1268; 1161. 1H NMR (400 MHz, DMSO-d6, TMS, 1152. 1H NMR (400 MHz, DMSO-d6, TMS, d in ppm): 1.55–1.48 (m,
d in ppm): 1.34 (t, 3H, J = 7.1 Hz, CH2CH3); 1.59–1.53 (m, 2H, H10); 2H, H10); 1.71–1.63 (m, 2H, H9); 2.83–2.78 (m, 2H, H11);
1.73–1.66 (m, 2H, H9); 2.34 (s, 3H, CH3); 2.83–2.78 (m, 2H, H11); 3.65–3.60 (m, 2H, H8); 7.39–7.35 (m, 1H, H40 ); 7.64–7.54 (m, 6H,
3.66–3.61 (m, 2H, H8); 4.31 (q, 2H, J = 7.0 Hz, CH2CH3); 7.38–7.35 H30 –H50 , H400 , H200 –H600 or H300 –H500 , SO2NH); 7.79–7.77 (t, 2H,
(m, 3H, H40 , H200 –H600 or H300 –H500 ), 7.59–7.54 (m, 3H, SO2NH, H200 –H600 or H300 –H500 ); 8.14–8.12 (m, 2H, H20 –H60 ); 8.39 (s, 1H,
H30 –H50 ); 7.67 (d, 2H, J = 8.2 Hz, H200 –H600 or H300 –H500 ); 8.19–8.17 H6); 8.49 (s, 1H, H3). 13C NMR (100 MHz, DMSO-d6, TMS, d in
(m, 2H, H20 –H60 ); 8.48 (s, 1H, H3); 8.77 (s, 1H, H6); 9.06 (t, 1H, ppm): 25.9; 26.1; 41.9; 42.2; 43.4; 118.5; 121.4; 123.3; 126.3;
J = 5.2, NH). 13C NMR (100 MHz, DMSO-d6, TMS, d in ppm): 14.1; 128.9; 129.0; 129.9; 132.2; 134.9; 138.5; 140.4; 150.8; 151.4;
20.8; 25.8; 26.3; 42.0; 44.0; 60.2; 99.3; 104.0; 121.3; 126.2; 126.4; 153.8. HRMS (ESI) calcd for C23H22N6O2S 446.1525, found [M+1]+
128.9; 129.5; 136.1; 137.5; 138.7; 142.4; 151.3; 152.4; 152.5; 447.1609. HPLC: 98.3%.
167.7. HRMS (ESI) calcd for C26H29N5O4S 507.1940, found [M+1]+
508.2024. HPLC: 97%. 4.2.8.7. N-(4-((5-Cyano-1-phenyl-1H-pyrazolo[3,4-b]pyridin-4-yl)
amino)butyl)-4-methylbenzenesulfonamide (7). Yield: 65%.
4.2.8.3. Ethyl 4-((4-(4-fluorophenylsulfonamido)butyl)amino)-1- Mp: 160–161 °C. IR (KBr. cm1): 3295; 2943–2874; 2207; 1592–
phenyl-1H-pyrazolo[3,4-b]pyridine-5-carboxylate (3). Yield: 1455; 1322; 1153. 1H NMR (400 MHz, DMSO-d6, TMS, d in ppm):
59%. Mp: 160–162 °C. IR (KBr. cm1): 2987–2864; 1667; 1588; 1.55–1.48 (m, 2H, H10); 1.69–1.63 (m, 2H, H9); 2.35 (s, 3H, CH3);
1368; 1261; 1153. 1H NMR (400 MHz, DMSO-d6, TMS, d in ppm): 2.80–2.76 (m, 2H, H11); 3.65–3.59 (m, 2H, H8); 7.39–7.35 (m, 3H,
1.34 (t, 3H, J = 7.1 Hz, CH2CH3); 1.60–1.53 (m, 2H, H10); 1.74–1.67 H40 , H200 –H600 or H300 –H500 ); 7.58–7.52 (m, 3H, H30 –H50 , SO2NH);
(m; 2H, H9); 2.87–2.82 (m, 2H, H11); 3.68–3.64 (m, 2H, H8); 4.31 7.66 (d, 2H, J = 8.2 Hz, H200 –H600 or H300 –H500 ); 8.13 (d, 2H,
(q, 2H, J = 7.0 Hz, CH2CH3); 7.43–7.34 (m, 3H, H40 , H200 –H600 ); 7.55 J = 7.7 Hz, H20 –H60 ); 8.39 (s, 1H, H6); 8.49 (s, 1H, H3). 13C NMR
(t, 2H, J = 7.7 Hz, H30 –H50 ); 7.71 (t, 1H, J = 5.9, SO2NH); 7.87–7.83 (100 MHz, DMSO-d6, TMS, d in ppm): 20.8; 26.1; 42.2; 43.4; 121.4;
(m, 2H, H300 –H500 ); 8.19–8.17 (m, 2H, H20 –H60 ); 8.50 (s, 1H, H3); 126.4; 128.9; 129.5; 134.9; 137.5; 138.5; 142.4; 150.8; 151.4;
8.77 (s, 1H, H6); 9.07 (t, 1H, J = 5.1 Hz, NH). 13C NMR (100 MHz, 153.8. HRMS (ESI) calcd for C24H24N6O2S 460.1681, found [M+1]+
DMSO-d6, TMS, d in ppm): 14.1; 25.8; 26.3; 42.1; 44.0; 60.2; 99.3; 461.1763. HPLC: 95.3%.
104.0; 116.2 (J = 22.5; C300 –C500 ); 121.3; 126.2; 128.9; 129.3
(J = 9.4; C200 –C600 ); 136.1; 136.8; 138.7; 151.4; 152.4; 152.5; 164.0 4.2.8.8. N-(4-((5-Cyano-1-phenyl-1H-pyrazolo[3,4-b]pyridin-4-
(J = 248.8; C400 ); 167.8. 19F NMR (376 MHz, DMSO-d6, TMS, d in yl)amino)butyl)-4-fluorobenzenesulfonamide (8). Yield:
ppm): 107.14. HRMS (ESI) calcd for C25H26FN5O4S 511.1690, found 63%. Mp: 128–129 °C. IR (KBr. cm1): 3285; 3150; 2869; 2221;
[M+1]+ 512.1764. HPLC: 100%. 1590–1427; 1327; 1152. 1H NMR (400 MHz, DMSO-d6, TMS, d in
ppm): 1.55–1.48 (m, 2H, H10); 1.71–1.64 (m, 2H, H9); 2.83–2.78
4.2.8.4. Ethyl 4-((4-(4-chlorophenylsulfonamido)butyl)amino)-1- (m, 2H, H11); 3.65–3.60 (m, 2H, H8); 7.42–7.36 (m, 3H, H40 ,
phenyl-1H-pyrazolo[3,4-b]pyridine-5-carboxylate (4). Yield: H200 –H600 or H300 –H500 ); 7.58–7.54 (m, 2H, H30 –H5); 7.67 (t; 1H,
70%. Mp: 143–145 °C. IR (KBr. cm1): 3268; 2944–2869; 1666; J = 5.8 Hz, SO2NH); 7.86–7.82 (m, 2H, H200 –H600 or H300 –H500 );
1581; 1324; 1267; 1157. 1H NMR (400 MHz, DMSO-d6, TMS, d in 8.14–8.12 (m, 2H, H20 –H60 ); 8.38 (s, 1H, H6); 8.49 (s, 1H, H3). 13C
ppm): 1.34 (t, 3H, J = 7.1 Hz, CH2CH3); 1.61–1.54 (m, 2H, H10); NMR (100 MHz, DMSO-d6, TMS, d in ppm): 25.9; 26.1; 42.2; 43.4;
1.72–1.67 (m, 2H, H9); 2.87–2.83 (m, 2H, H11); 3.67–3.63 (m, 2H, 116.2 (J = 22.5 Hz, C300 –C500 ); 118.5; 121.4; 123.3; 126.4; 128.9;
H8); 4.31 (q, 2H, J = 7.0 Hz, CH2CH3); 7.36 (t, 1H, J = 7.4 Hz, H40 ); 129.3 (J = 9.4 Hz, C200 –C600 ); 129.9; 134.8; 136.8 (J = 3.0 Hz, C100 );
7.56 (t, 2H, J = 7.9 Hz, H30 –H50 ); 7.64 (t, 2H, J = 8.6 Hz, H200 –H600 or 138.5; 150.8; 151.4; 153.8; 156.5; 163.9 (J = 249.1 Hz, C400 ). 19F
H300 –H500 ); 7.81–7.77 (m, 3H, H200 –H600 or H300 –H500 , SO2NH); 8.18 NMR (376 MHz, DMSO-d6, TMS, d in ppm): 107.14. HRMS (ESI)
(d, 2H, J = 7.7 Hz, H20 –H60 ); 8.50 (s, 1H, H3); 8.77 (s, 1H, H6); 9.07 calcd for C23H21FN6O2S 464.1431, found [M+1]+ 465.1516. HPLC:
(t, 1H, J = 5.1 Hz, NH). 13C NMR (100 MHz, DMSO-d6, TMS, d in 97.3%.
ppm): 14.1; 25.8; 26.3; 42.1; 44.0; 60.2; 99.3; 104.0; 121.3; 126.2;
128.3; 128.9; 129.2; 136.1; 137.1; 138.7; 139.3; 151.3; 152.4; 4.2.8.9. 4-Chloro-N-(4-((5-cyano-1-phenyl-1H-pyrazolo[3,4-b]
152.5; 167.8. HRMS (ESI) calcd for C25H26ClN5O4S 528.1394, found pyridin-4-yl)amino)butyl)benzenesulfonamide (9). Yield:
[M+1]+ 528.1488. HPLC: 99.8%. 68%. Mp: 99–100 °C. IR (KBr. cm1): 3319; 3111; 2947–2875;
2214; 1578–1461; 1329; 1151. 1H NMR (400 MHz, DMSO-d6,
4.2.8.5. Ethyl 4-((4-(4-methoxyphenylsulfonamido)butyl) TMS, d in ppm):1.56–1.51 (m, 2H, H10); 1.70–1.64 (m, 2H, H9);
amino)-1-phenyl-1H-pyrazolo[3,4-b]pyridine-5-carboxylate (5). 2.84–2.80 (m, 2H, H11); 3.65–3.60 (m, 2H, H8); 7.37 (t, 1H,
Yield: 60%. Mp: 165–167 °C. IR (KBr. cm1): 2960–2872; 1667; J = 7.4 Hz, H40 ); 7.56 (t, 2H, J = 7.7 Hz, H30 –H50 ); 7.64 (d, 2H,
1579; 1341; 1179; 1263; 1149. 1H NMR (400 MHz. DMSO-d6. J = 8.6 Hz, H200 –H600 or H300 –H500 ); 7.78 (d, 2H, J = 8.6 Hz, H200 –H600
TMS. d in ppm): 1.34 (t, 3H, J = 7.0 Hz, CH2CH3); 1.60–1.54 (m, or H300 –H500 ); 8.13 (d, 2H, J = 7.8 Hz, H20 –H60 ); 8.39 (s, 1H, H6);
2H, H10); 1.72–1.68 (m, 2H, H9); 2.82–2.77 (m, 2H, H11); 8.49 (s, 1H, H3). 13C NMR (100 MHz, DMSO-d6, TMS, d in ppm):
3.64–3.63 (m, 2H, H8); 3.80 (s, 3H, OCH3); 4.31 (q, 2H, J = 7.0 Hz, 25.9; 26.1; 42.2; 43.4; 118.5; 121.4; 126.4; 128.3; 128.9; 129.2;
CH2CH3); 7.07 (d, 1H, J = 8.7 Hz, H200 –H600 or H300 –H500 ); 7.36 (t, 134.9; 137.1; 138.5; 139.3; 150.8; 151.4; 153.9. HRMS (ESI) calcd
1H, J = 7.3 Hz, H40 ); 7.57–7.49 (m, 3H, H30 –H50 , SO2NH); 7.72 (d, for C23H21ClN6O2S 480.1135, found [M+1]+ 481.1216. HPLC: 91%.
2H, J = 8.7 Hz, H200 –H600 or H300 –H500 ); 8.18 (d, 2H, J = 8.0 Hz,
H20 –H60 ); 8.48 (s, 1H, H3); 8.76 (s, 1H, H6); 9.06 (s, 1H, NH). 13C 4.2.8.10. N-(4-((5-Cyano-1-phenyl-1H-pyrazolo[3,4-b]pyridin-4-
NMR (100 MHz, DMSO-d6, TMS, d in ppm): 14.0; 25.8; 26.2; 42.0; yl)amino)butyl)-4-methoxybenzenesulfonamide (10). Yield:
44.0; 55.4; 60.2; 99.3; 104.0; 114.2; 121.3; 126.9; 126.2; 128.5; 63%. Mp: 156–157 °C. IR (KBr. cm1): 3340; 3277; 2973–2873;
128.9; 132.1; 136.1; 138.7; 151.3; 152.4; 152.5; 161.9; 167.7. 2211; 1596–1413; 1318; 1147. 1H NMR (400 MHz, DMSO-d6, TMS,
4498 T. B. Silva et al. / Bioorg. Med. Chem. 24 (2016) 4492–4498

d in ppm):1.55–1.48 (m, 2H, H10); 1.70–1.63 (m, 2H, H9); 2.79–2.74 References and notes
(m, 2H, H11); 3.65–3.60 (m, 2H, H8); 3.81 (s, 3H, OCH3); 7.09–7.05
(m, 2H, H200 –H600 or H300 –H500 ); 7.39–7.35 (m, 1H, H40 ); 7.45 (t, 1H, 1. World Health Organization (WHO), World Malaria Report 2014. Available at:
http://www.who.int/malaria/publications/world_malaria_report_2014/en/
J = 5.9 Hz, SO2NH); 7.58–7.54 (m, 2H, H30 –H50 ); 7.72–7.68 (m, 2H, Accessed: April/2016.
H200 –H600 or H300 –H500 ); 8.14–8.12 (m, 2H, H20 –H60 ); 8.39 (s, 1H, 2. Severini, C.; Menegon, M. J. Glob. Antimicrob. Resist. 2015, 3, 58.
H6); 8.49 (s, 1H, H3). 13C NMR (100 MHz, DMSO-d6, TMS, d in 3. World Health Organization(WHO), Guidelines for the Treatment of Malaria 3rd
ed, 2015. Available at: http://www.who.int/malaria/publications/atoz/
ppm): 26.0; 26.1; 42.2; 43.4; 55.5; 114.2; 118.5; 121.4; 126.4; 9789241549127/en/ Accessed: April/2016.
128.5; 128.9; 132.0; 134.9; 138.5; 150.7; 151.4; 161.9. HRMS (ESI) 4. White, N. J. J. Clin. Invest. 2004, 113, 1084.
calcd for C24H24N6O3S 476.1631, found [M+1]+ 477.1715. HPLC: 5. Dondorp, A. M.; Yeung, S.; White, L.; Nguon, C.; Day, N. P.; Socheat, D.; von
Seidlein, L. Nat. Rev. Microbiol. 2010, 8, 272.
95.3%.
6. Schrader, F. C.; Barho, M.; Steiner, I.; Ortmann, R.; Schlitzer, M. Int. J. Med.
Microbiol. 2012, 302, 165.
7. Barnett, D. S.; Guy, R. K. Chem. Rev. 2014, 114, 11221.
4.3. Biological evaluation
8. Anthony, M. P.; Burrows, J. N.; Duparc, S.; JMoehrle, J.; Wells, T. N. C. Malaria J.
2012, 11, 316.
4.3.1. Continuous cultures of P. falciparum-infected 9. Teixeira, C.; Vale, N.; Pérez, B.; Gomes, A.; Gomes, J. R. B.; Gomes, P. Chem. Rev.
erythrocytes and in vitro assays 2014, 114, 11164.
10. Vandekerckhove, S.; D’hooghe, M. Bioorg. Med. Chem. 2015, 23, 5098.
The cultivation of the P. falciparum W2 strain (chloroquine 11. Hamdy, N. A.; Gamal-Eldeen, A. M. Eur. J. Med. Chem. 2009, 44, 4547.
resistant) was performed according to the protocol of Trager & 12. Orlikova, B.; Chaouni, W.; Schumacher, M.; Aadil, M.; Diederich, M.; Kirsch, G.
Jensen28 using human erythrocytes (A+) in RPMI 1640 medium Eur. J. Med. Chem. 2014, 85, 450.
13. Leal, B.; Afonso, I. F.; Rodrigues, C. R.; Abreu, P. A.; Garrett, R.; Pinheiro, L. C. S.;
supplemented with human serum. Parasites were synchronized Azevedo, A. R.; Borges, J. C.; Vegi, P. F.; Santos, C. C. C.; Silveira, F. C. A.; Cabral, L.
with sorbitol in order to obtain the young (ring) parasite forms.29,30 M.; Frugulhetti, I. C. P. P.; Bernardino, A. M. R.; Santos, D. O.; Castro, H. C. Bioorg.
Parasitemia was adjusted to 0.05% and hematocrit to 1.5%; subse- Med. Chem. 2008, 16, 8196.
14. Parveen, N.; Aslam, S.; Maqbool, T.; Bernardino, A. M. R.; Munawar, M. A.; Khan,
quently, cultures were placed in microplates together with the M. A. Afinidad LXXI 2014, 566, 152.
drugs to be tested in serial dilutions (50–0.78 lM), as well as pos- 15. Gudmundsson, K. S.; Johns, B. A.; Wang, Z.; Turner, E. M.; Allen, S. H.; Freeman,
itive (chloroquine and sulfadoxine) and negative (no drug) con- G. A.; Jr, Boyd. F. L.; Sexton, C. J.; Selleseth, D. W.; Monirib, K. R.; Creech, K. L.
Bioorg. Med. Chem. 2005, 13, 5346.
trols, and incubated for 72 h. The microplates were then
16. Bernardino, A. M. R.; Azevedo, A. R.; Pinheiro, L. C. S.; Borges, J. C.; Carvalho, V.
subjected to three cycles of freezing and thawing, and parasite L.; Miranda, M. D.; Meneses, M. D. F.; Nascimento, M.; Ferreira, D.; Rebello, M.
growth was measured through quantification of histidine-rich pro- A.; Silva, V. A. G. G.; Frugulhetti, I. C. P. P. Med. Chem. Res. 2007, 16, 352.
17. Azevedo, A. R.; Ferreira, V. F.; Mello, H. M.; Ferreira, L. R. L.; Jabor, A. V.;
tein II (HRPII) using specific monoclonal antibodies in sandwich
Frugulhetti, I. C. P. P.; Pereira, H. S.; Moussatche, N.; Bernardino, A. M. R.
ELISA.31 Absorbances were read at 450 nm using a SpectraMax Heterocycl. Commun. 2002, 8, 427.
190 spectrophotometer, and the determination of IC50 and SD 18. Gomez, R.; Jolly, S. J.; Williams, T.; Vacca, J. P.; Torrent, M.; McGaughey, G.; Lai,
was performed using dose–response curves with a non-linear M.-T.; Felock, P.; Munshi, V.; DiStefano, D.; Flynn, J.; Miller, M.; Yan, Y.; Reid, J.;
Sanchez, R.; Liang, Y.; Paton, B.; Wan, B.-L.; Anthony, N. J. Med. Chem. 2011, 54,
regression function. (Origin software, OriginLab Corporation, 7920.
Northampton, MA, USA). 19. Dias, L. R. S.; Santos, M. B.; Albuquerque, S.; Castro, H. C.; Souza, A. M. T.;
Freitas, A. C. C.; DiVaio, M. A. V.; Cabral, L. M.; Rodrigues, C. R. Bioorg. Med.
Chem. 2007, 15, 211.
4.3.2. Cell cultures and cytotoxicity tests 20. Mello, H.; Echevarria, A.; Bernardino, A. M.; Canto-Cavalheiro, M.; Leon, L. L. J.
BGM cells were cultured in RPMI 1640 medium supplemented Med. Chem. 2004, 47, 5427.
with 10% fetal bovine serum (FBS) at a concentration of 5  103 21. Manach, C. L.; Paquet, T.; Brunschwig, C.; Njoroge, M.; Han, Z.; Cabrera, D. G.;
Bashyam, S.; Dhinakaran, R.; Taylor, D.; Reader, J.; Botha, M.; Churchyard, A.;
cells per milliliter. Cells were added together with the test com- Lauterbach, S.; Coetzer, T. L.; Birkholtz, L.-M.; Meister, S.; Winzeler, E. A.;
pounds in serial dilution. The microplates were incubated for Waterson, D.; Witty, M. J.; Wittlin, S.; Jiménez-Díaz, M.-B.; Martínez, M. S.;
24 h at 37 °C. Subsequently, MTT salt at a concentration of 3 mg/ Ferrer, S.; Angulo-Barturen, I.; Street, L. J.; Chibale, K. J. Med. Chem. 2015, 58,
8713.
mL was added after 3 h, the supernatant was removed, and the
22. Dias, R. S.; Freitas, A. C. C.; Barreiro, E. J.; Goins, D. K.; Nanayakkara, D.; Mc-
dye present on the bottom of the plate wells was dissolved in Chesney, J. D. Boll. Chim. Farm. 2000, 139, 14.
DMSO in a volume of 100 lL/well. The microplates were then read 23. Varotti, F. P.; Botelho, A. C. C.; Andrade, A. A.; Paula, R. C.; Fagundes, E. M. S.;
Valverde, A.; Mayer, L. M. U.; Mendonca, J. S.; Souza, M. V. N.; Boechat, N.;
using a Spectramax 190 spectrophotometer with a 570 nm filter.32
Krettli, A. U. Antimicrob. Agents Chemother. 2008, 52, 3868.
The determination of MLD50 was performed using dose response 24. Boechat, N.; Pinheiro, L. C. S.; Silva, T. S.; Aguiar, A. C. C.; Carvalho, A. S.; Bastos,
curves with a nonlinear regression function.32 (Origin software, M. M.; Costa, C. C. P.; Pinheiro, S.; Pinto, A. C.; Mendonça, J. S.; Dutra, K. D. B.;
OriginLab Corporation, Northampton, MA, USA). Selectivity index Valverde, A. L.; Santos-Filho, O. A.; Ceravolo, I. P.; Krettli, A. U. Molecules 2012,
17, 8285.
(SI) was defined as the ratio of the MLD50 to the IC50 values of 25. Boechat, N.; Ferreira, M. L. G.; Pinheiro, L. C. S.; Jesus, A. M. L.; Leite, M. M. M.;
the active compounds. Aguiar, A. C. C.; Andrade, I. M.; Krettli, A. U. Chem. Biol. Drug. Des. 2014, 84, 325.
26. Carvalho, R. C. C.; Martins, W. A.; Silva, T. P.; Kaiser, C. R.; Bastos, M. M.;
Pinheiro, L. C. S.; Krettli, A. U.; Boechat, N. Bioorg. Med. Chem. Lett. 2016, 26,
Acknowledgments 1881.
27. Pinheiro, L. C. S.; Boechat, N.; Ferreira, M. L. G.; Junior, C. C. S.; Jesus, A. M. L.;
Leite, M. M. M.; Souza, N.; Krettli, A. U. Bioorg. Med. Chem. 2015, 23, 5979.
The authors thank the Coordination of Improvement of Higher 28. Bernardino, A. M. R.; Pinheiro, L. C. S.; Rodrigues, C. R.; Loureiro, N. I.; Castro, H.
Education (CAPES) and the National Council of R&D of Brazil C.; Lanfredi-Rangel, A.; Sabatini-Lopes, J.; Borges, J. C.; Carvalho, J. M.; Romeiro,
G. A.; Ferreira, V. F.; Frugulhettic, I. C. P. P.; Vannier-Santos, M. A. Bioorg. Med.
(CNPq) for the fellowships granted to the authors. We also thank Chem. 2006, 14, 5765.
the Foundation for Research of the State of Rio de Janeiro (FAPERJ), 29. Trager, W.; Jensen, J. Science 1976, 193, 673.
Technological Development Program on Products for Health 30. Lambros, C.; Vanderberg, J. J. Parasitol. 1979, 65, 418.
31. Noedl, H. C.; Wongsrichanalai, R.; Miller, K.; Myint, S.; Looareesuwan, Y.;
(PDTIS), for financial support. LJMC and NB is recipient of research Sukthana, V.; Wongchotigul, H.; Kollaritsch, G.; Wiedermann, W. Wernsdorfer.
productivity fellowships from the CNPq and from FAPERJ (‘Cientista Exp. Parasitol. 2002, 102, 157.
do Nosso Estado’). 32. Denizot, F.; Lang, R. J. Immunol. Methods 1986, 89, 271.

S-ar putea să vă placă și