You are on page 1of 11

Medicinal Chemistry Research (2020) 29:538–548 MEDICINAL

https://doi.org/10.1007/s00044-020-02505-8
CHEMISTRY
RESEARCH
ORIGINAL RESEARCH

Design, synthesis, and characterization of novel substituted


1,2,4-oxadiazole and their biological broadcast
Paranjay H. Parikh1 Jignesh B. Timaniya1 Mrugesh J. Patel1 Kaushal P. Patel1
● ● ●

Received: 30 August 2019 / Accepted: 8 January 2020 / Published online: 18 January 2020
© Springer Science+Business Media, LLC, part of Springer Nature 2020

Abstract
In the current study, a novel combinatorial library of substituted 1,2,4-oxadiazoles bearing pyridine and thiazole have been
synthesized via the condensation reaction of amidoxime and carboxylic acids and its derivatives in two way. The titled
compounds were screened for their in vitro antimicrobial activity against three gram positive bacteria and three gram
negative bacteria, In vitro antituberculosis activity against Mycobacterium tuberculosis H37RV and in vitro antimalarial
activity against Plasmodium falciparum. Majority of the compounds were found to be active against gram positive bacteria.
Compounds 6e, 6f, 7e, and 7f elaborate moderate antituberculosis activity against the first-line drug while 6c, 6d, 7c, and 7d
1234567890();,:
1234567890();,:

intricate excellent antimalarial activity against Plasmodium falciparum strain.


Keywords Oxadiazole Pyridine Thiazole Plasmodium falciparum Mycobacterium tuberculosis
● ● ● ●

Introduction is also a major infectious disease affected by Mycobacter-


ium tuberculosis. Main cause of revival of tuberculosis is
Malaria remains one of the most vital infectious diseases the existence of multidrug-resistant M. tuberculosis strains
around the equator, particularly many parts of Americas, (Palomino and Martin 2014). WHO recent survey revealed
Asia, and much of the Africa also; however, 85–90% that still all do not have the MDR-TB facilely with them.
malaria fatalities occur in sub-Saharan Africa. Plasmodium They also declare TB to be the ‘global emergency’ and
falciparum, the most dangerous malarial parasite, is more than 30 million people will be effected by myco-
accountable for about 1 million deaths every year, accord- bacteria in the next 20 years (Sandhu 2011; WHO 2011).
ing to the World Health Organization latest report million The usually accessible drugs are not sufficient to treat the
documented cases of malaria were reported (The Malaria Multidrug-resistant TB and extensively drug-resistant TB
Eradication Research Agenda Group 2011; WHO 2018). (London et al. 2016; Patel et al. 2012). That’s need to
During the past few decade research to control the malaria developed the new drugs which has divergent and unique
infection has been carried by various prevalent countries, structure and with a mechanism of action possibly different
donors and global malaria partners gave us strengthened to from that of existing drugs. In this regard, it was thought
control malaria around the world (Karunamoorthi 2014). that it is worth design and synthesizing various novel
Various infectious diseases are influencing the world compounds which may have combined applications as
through their morbidity and mortality in which tuberculosis antimalarial, antituberculosis and antimicrobial agents
(Dramowski et al. 2012).
Oxadiazoles and their derivatives can be considered as
simple five membered heterocycles possessing one oxygen
Supplementary information The online version of this article (https:// and two nitrogen atoms. The oxadiazoles exist in different
doi.org/10.1007/s00044-020-02505-8) contains supplementary
isomeric forms such as 1,2,4-, 1,2,5-, 1,2,3-, and 1,3,4-
material, which is available to authorized users.
oxadiazoles (Hemming 2001). 1,2,4-, 1,2,5-, and 1,3,4-
* Kaushal P. Patel oxadiazoles are known but 1,2,3-isomer is quit unstable and
kaus_chem@yahoo.com reserve back to the diazoketone tautomer (Salahuddin et al.
1 2017; Singh and Chouhan 2014). Oxadiazole have occupied
Department of Advanced Organic Chemistry, P. D. Patel Institute
of Applied Sciences, Charotar University of Science and unique place in the field of bioorganic and pharmaceutical
Technology, Gujarat 388421, India chemistry due to their diverse activity like anti-microbial
Medicinal Chemistry Research (2020) 29:538–548 539

(Bakht et al. 2010; Desai et al. 2013; Karakaya et al. 2019; 3,5-disubstituted 1,2,4-oxadiazoles. Scheme 1 shows the
Madhu Sekhar et al. 2018; Malladi et al. 2014; Ningaiah general reaction conditions and method for the synthesized
et al. 2014; Padmavathi et al. 2009; Patel et al. 2017; Rane the 1,2,4-oxadiazoles. First amide intermediate was pre-
et al. 2013; Thakkar et al. 2017; Tresse et al. 2019; Zhou pared from acid chloride and aniline to give intermediate
et al. 2017), anti-inflammatory, antitubercular, analgesic, 3a–b in excellent yield. Intermediate 3a–b were treated
anticonvulsant, anticancer, antifungal, herbicidal, and anti- with hydroxylamine to provide the required amidoxime
parkinson activities (Kumar et al. 2016; Palit et al. 2016). 4a–b in good yields. The reaction of amidoxime with var-
The five-member heterocyclic compounds; particu- ious aryl and alkyl acid in the presence of HATU gave 3,5-
larly1,2,4-oxadiazole have been successfully tested against disubstituted-1,2,4-oxadiazole 6a–f and 7a–f with good
several diseases and therefore received special attention in yield.
the field of chemistry due to their wide medicinal applica-
tions. Various 1,2,4-oxadiazoles have been found as Pharmacology
potential agonists for cortical muscarinic, benzodiazepine,
and 5-HT1D receptors, and as antagonists for histamine H3 Antimicrobial activity
receptors (O Bora et al. 2014). It was thought worthwhile to
incorporate above biological importance and heterocyclic The minimal inhibitory concentration (MIC) of all the
derivatives in one moiety to find the best biologically active synthesized compounds 3a–b, 4a–b, 6a–f, and 7a–f was
heterocycle. So, we planned to synthesized 1,2,4-oxadiazole resolute by broth microdilution method according to
derivatives and examine their biological potency. National Committee for Clinical Laboratory Standards
(NCCLS-2002). The antibacterial activity of all synthesized
compounds 3a–b, 4a–b, 6a–f, and 7a–f was screened for
Results and discussion activity on three Gram positive bacteria Bacillus subtilis
(MTCC 441), Clostridium tetani (MTCC 449) Strepto-
Chemistry coccus pneumoniae (MTCC 1936) and three Gram negative
bacteria Escherichia coli (MTCC 443), Salmonella typhi
Synthesis of 1,2,4-oxadiazole involves the condensation (MTCC 98), Vibrio cholerae (MTCC 3906) by using
between amidoxime and carboxylic acids and its derivatives ampicillin, norfloxacin and ciprofloxacin as the standard
in two way. Coupling between the amidoximes with acti- antibacterial drugs. Antifungal activity was screened against
vated carboxylic acid derivatives such as acid chlorides, two fungal species Candida albicans (MTCC 3008) and
fluorides, anhydrides, and esters to give 1,2,4-oxadiazole Aspergillus fumigatus (MTCC 227) where nystatin and
derivatives or less reactive carboxylic acids in the presence griseofulvin were used as the standard antifungal drugs. The
of some coupling reagents like DCC, EDC, HATU, PyBOP, outcome of anti-microbial screening is summarized in
TBTU, etc. Other transition metal-catalyzed method include Tables 1 and 2.
the reaction of amidoximes with aryl halides in presence of Upon exploration of antimicrobial screening data (Table 3),
Pd-Catalyst or with aldehydes and subsequently oxidation it has been observed that Compounds 3a–b and 4a–b are
and many other methods also available to synthesize less active against gram positive bacteria. While the

Scheme 1 Synthesis of 3,5-disubstituted-1,2,4-oxadiazole 6a–f and 7a–f. (i) Et3N (2.0 eq., DCM, rt,4 h, (ii) Hydroxylamine (50% w/w in H2O),
Ethanol, reflux, overnight, and (iii) HATU (1.2 eq.), CH3CN, 150 °C, MW, 20 min
540 Medicinal Chemistry Research (2020) 29:538–548

Table 1 Synthesized of 3,5-disubstituted-1,2,4-oxadiazole 6a–f and 7a–f

Sr Sr
Substrates Yield Substrates Yield
No No

1 7

6a 7a

2 8

6b 7b

3 9

6c 7c

4 10

6d 7d

5 11

6e 7e

6 12

6f 7f

majority of the compounds showed excellent activity 250 µg/mL). Whereas compounds 6c and 7a illustrated
against gram positive bacteria B. subtilis and C. tetani as the same activity against B. subtilis as compared with
compared with ampicillin (MIC = 250 µg/mL). Com- norfloxacin (MIC = 100 µg/mL). Whereas in inhibiting
pounds 6a, 6b, and 7d against S. pneumoniae showed gram negative bacteria, the compounds 6a and 7e showed
identical potency as compared with ampicillin (MIC = gleaming activity i.e., 62.5 µg/mL against V. cholera as
Medicinal Chemistry Research (2020) 29:538–548 541

Table 2 In vitro antimicrobial


Compounds Gram positive bacteria Gram negative bacteria Fungi
activity (MIC, µg/mL) of all the
synthesized compounds 3a–b, S.P. B.S. C.T. E.C. S.T. V.C. C.A. A.F.
4a–b, 6a–f, 7a–f MTCC MTCC MTCC MTCC MTCC MTCC MTCC MTCC
1936 441 449 443 98 3906 227 3008

3a 200 500 500 250 250 200 >1000 >1000


3b 125 200 500 200 250 200 >1000 1000
4a 200 500 250 100 250 250 1000 500
4b 250 500 500 200 100 100 1000 500
6a 100 100 500 250 62.5 62.5 100 250
6b 200 100 200 250 100 200 500 200
6c 250 250 100 200 250 200 >1000 200
6d 100 250 125 250 200 200 250 250
6e 250 200 250 100 125 100 500 1000
6f 250 250 200 100 250 200 1000 500
7a 500 500 100 200 200 250 >1000 500
7b 250 500 200 125 250 200 >1000 200
7c 200 250 250 200 100 200 500 500
7d 100 100 200 62.5 100 200 1000 500
7e 125 200 200 200 200 62.5 500 200
7f 100 250 500 100 62.5 100 100 100
A 100 250 250 100 100 100 n.t.a n.t.
B 10 100 50 10 10 10 n.t. n.t.
C 25 50 100 25 25 25 n.t. n.t.
D n.t. n.t. n.t. n.t. n.t. n.t. 100 100
E n.t. n.t. n.t. n.t. n.t. n.t. 500 100
S.P. Streptococcus pneumoniae, B.S. Bacillus subtilis, C.T. Clostridium tetani, E.C. Escherichia coli, S.T.
Salmonella typhi, V.C. Vibrio cholerae, C.A. Candida albicans, A.F. Aspergillus fumigatus, MTCC
microbial type culture collection, A Ampicillin, B Norfloxacin, C Chloramphenicol, D Ciprofloxacin,
E Nystatin, F Griseofulvin
a
n.t. not tested

Table 3 In vitro antituberculosis


Compound % Inhibition Compound % Inhibition
activity (% inhibition) of
compounds 3a–b, 4a–b, 6a–f, 250 µg/mL 100 µg/mL 250 µg/mL 100 µg/mL
and 7a–f against M. tuberculosis
H37Rv (at concentration 3a 16 14 6f 95 92
250 µg/mL and 100 µg/mL) 3b 24 17 7a 42 31
4a 11 5 7b 71 61
4b 36 47 7c 36 25
6a 88 77 7d 78 68
6b 20 17 7e 84 77
6c 74 24 7f 96 91
6d 66 54 Rifampicin 98 98
6e 92 92 Isoniazid 99 99

compared with ampicillin (MIC = 100 µg/mL). Com- and 7f against E. coli showed equal activity as compared
pounds 6a and 7f illustrate excellent activity i.e., 62.5 µg/ with ampicillin (MIC = 100 µg/mL), while against S.
mL against Salmonella typhi as well as Compound 7d typhi Compounds 4a, 6b, 7c, and 7d well as compounds
showed brilliant activity against E. Coli as compared with 4b and 7f against V. cholerae showed the same potency
ampicillin (MIC = 100 µg/mL). Compounds 4a, 6e, 6f as compared with ampicillin (MIC = 250 µg/mL).
542 Medicinal Chemistry Research (2020) 29:538–548

Antifungal activity Table 4 In vitro antimalarial activity of compounds 6a–f and 7a–f
Compound IC50 (µg/mL) Compound IC50 (µg/mL)
The fallout of antifungal study (Table 2) of all the synthesized
3a 1.55 6f 0.198
compounds 3a–b, 4a–b, 6a–f, and 7a–f revealed that all the
compounds have poor activity against A. fumigates, except 3b 1.36 7a 1.54
only one compound 6f which shows the same potency as 4a 1.88 7b 0.97
compared with Griseofulvin and Nystatin i.e., 100 µg/mL. 4b 1.47 7c 0.082
Whereas in dissimilarity with standard fungicidal griseofulvin 6a 0.42 7d 0.096
(MIC = 500 µg/mL), compounds 4b, 6d, and 7f have a say 6b 0.78 7e 0.38
luminous antifungal activity i.e., 100 µg/mL, 250 µg/mL, 6c 0.054 7f 0.47
100 µg/mL against C. albicans. Whereas compounds 6b, 7c, 6d 0.322 Quinine 0.268
and 7e showed the same effect as that of griseofulvin i.e., 6e 0.57 Chloroquine 0.020
500 µg/mL against C. albicans. Other all compounds have
frail antifungal activity than nystatin and griseofulvin.
These compounds elaborate major activity against P. fal-
Antituberculosis activity ciparum strain as compared with quinine IC50 0.268 µg/mL.
Whereas compound 6d was found to possesses sensible
All the synthesized targets 3a–b, 4a–b, 6a–f, and 7a–f were activity i.e., IC50 0.322 µg/mL aligned with chloroquine. All
evaluated for their in vitro antituberculosis activity against other compounds were found to be not as much active as
Mycobacterium tuberculosis H37Rv strain. Primary chloroquine and quinine against P. falciparum strain.
screening of all the newly synthesized compounds 3a–b,
4a–b, 6a–f, and 7a–f was carried out at 100 and 250 µg/mL Structural activity relationship (SAR)
by using Lowenstein–Jensen medium as described by Rat-
tan (Rattan 2000). Isoniazid and Rifampicin were used as The outcome of biological assessment revealed that the
standard drugs. The results of antituberculosis screening activity was meaningfully affected by replacement of various
data are present in Table 3. functional groups at R position in oxadiazole ring. cyclo-
In the present study of screening, compound 6e exhibit propyl group was increased antibacterial activity against S.
better activity and showed 95% inhibition at 250 µg/mL typhi and V. cholera. While moderate active against S.
concentration and 92% inhibition at 100 µg/mL concentra- pneumonia and B.subtilis. and very less active against E. Coli.
tion. Compound 6f found to acquire outstanding activity at While methyl group was increased antibacterial activity
both the concentrations i.e., 92% at 250 and 100 µg/mL. against almost all gram positive bacteria but in case of gram
While compounds 7e and 7f were found to possesses great negative bacteria importance of methyl group is poor. Tert-
potency i.e., 96% inhibition at 250 µg/mL concentration and butyl group was inhibited potent antibacterial activity against
91% inhibition at 100 µg/mL concentration against M. gram positive bacteria M. tuberculosis H37Rv strain B.subtilis
tuberculosis H37Rv. While compounds 6c and 7e were and C. tetani as well as repressed strong antituberculosis
found reasonably active against M. tuberculosis H37Rv. activity against M. tuberculosis H37Rv strain and moderate
Remaining all compounds showed deprived inhibition of M. antimalarial activity Phenyl group having different sub-
tuberculosis growth. From the above results, it can be stituents i.e., nitro group, cyano group and methoxy group
accomplished that, compounds 6e and 7f may turn into an present at fourth position on phenyl ring responsible for
innovative class of antitubercular agents in the future. antimalarial activity compete against P. falciparum strain as
compared with quinine and chloroquine.
Antimalarial activity Phenyl group particularly chloro and iodo enhance the
antibacterial activity against all three gram positive bacteria but
All new oxadiazole derivatives 3a–b, 4a–b, 6a–f, and 7a–f very less active against gram negative bacteria. While phenyl
were tested for their antimalarial activity against P. falci- group having methoxy group at fourth position responsible for
parum strain. The obtained results are presented in Table 4. good antibacterial activity against gram negative bacteria S.
Quinine and Chloroquine were used as the reference stan- typhi. While thiazole ring attached to amide bond responsible
dard drugs. The results of the pharmacological screening are for increased antibacterial activity against S. pneumonia, C.
articulated as the drug concentration resulting in 50% tetani and B. subtilis but not as much active against gram
inhibition (IC50) of parasite growth. negative bacteria and pyridine ring attached to amide bond
The compounds 6c, 7c, and 7d were originated to have responsible for increased antibacterial activity against S.
IC50 in the array of 0.032 to 0.096 µg/mL upon P. falci- pneumoniae and B. subtilis and increased antibacterial activity
parum strain as compared with quinine and chloroquine. against C. tetani and B. subtilis, respectively (Fig. 1).
Medicinal Chemistry Research (2020) 29:538–548 543

Fig. 1 Structural activity relationship (SAR)

Material and methods stirred at room temperature for 2 h followed by addition of


2-Thiophenecarbonyl chloride or nicotinoyl chloride
All the reagents were used without further purification and hydrochloride (5.7 mmol) and triethylamine (1.4 mL,
were obtained commercially. Thin-layer chromatography 10.4 mmol). On completion, Mixture was added to aqueous
(TLC, on aluminium plates coated with silica gel 60 F254, sodium bicarbonate solution, and the mixture was extracted
0.25 mm thickness, Merck) was used for monitoring the with (30 mL) chloroform. The organic layer was washed
progress of all reactions. Solvents used were of analytical with saturated brine and dried over anhydrous magnesium
grade. Mass and LCMS spectra were recorded on Shimadzu sulfate. The residue obtained upon concentration under
LCMS 2010 spectrometer (Shimadzu, Tokyo, Japan). The reduced pressure was purified by silica gel column chro-
IR spectra were recorded on Shimadzu FTIR 8401 spectro- matography using the solvent system of Ethyl acetate:
photometer using potassium bromide pellets in the range n-Hexane (2:3) to give the desired compound.
4000–400 cm−1 and frequencies of only characteristic peaks
are expressed in cm−1. The elemental analysis was done by N-(3-Chloro-4-cyanophenyl)thiophene-2-carboxamide (3a)
using PerkinElmer 2400 series-II elemental analyzer (Per-
kinElmer, USA) and all compounds are originated within ± Pale yellow solid (MeOH) (This compound was prepared by
0.4% of the theoretical compositions. 1H-NMR and 13C- the general procedure mentioned for the synthesis of com-
NMR spectra were recorded in DMSO-d6 on a Bruker pound 3a & 3b); Yield = 78%; mp 212–214 °C; IR (KBr)
Advance 400 (MHz), 500 (MHz) spectrometer using sol- νmax 1620 (amide, C=O), 3150–3290 (NH), 2240 (CN)
vent peak as an internal standard at 400, 500 and 101, (cm−1); 1H NMR (DMSO-d6, 500 MHz): δ = 10.69 (s, 1H,
126 MHz, respectively. Chemical shifts are reported in parts N-H), 8.16 (s, 1H, H-15), 8.05 (d, J = 3.63 Hz, 1H, H-13),
per million (ppm). Melting points were recorded in melting 7.90–7.96 (m, 2H, H-10, 6), 7.83–7.89 (m, 1H, H-7), 7.26 (t,
point apparatus µThermoCal10 (Analab Scientific Pvt. Ltd, J = 4.41 Hz, 1H, H-14); 13C NMR (DMSO-d6, 126 MHz):
India) and are uncorrected. δ = 160.5 (C-2), 144.2 (C-5), 138.7 (C-1), 135.8 (C-9), 135.1
(C-7), 133.2 (C-15), 130.4 (C-13), 128.3 (C-14), 119.9 (C-6),
118.5 (C-10), 116.2 (C-16), 105.7 (C-8); Anal. Calcd. for
Experimental procedure C12H7ClN2OS (262.71): C, 54.86; H, 2.69; Cl, 13.49; N,
10.66; O, 6.09; S, 12.20; found C, 54.88; H, 2.64; N, 10.71.
General procedure for Synthesis of N-(3-chloro-4-
cyanophenyl)thiophene-2-carboxamide (3a) and N-(3-Chloro-4-cyanophenyl)nicotinamide (3b)
N-(3-chloro-4-cyanophenyl)nicotinamide (3b)
Pale yellow solid (MeOH) (This compound was prepared
4-amino-2-chlorobenzonitrile (0.8 g, 5.2 mmol) was dis- by the general procedure mentioned for the synthesis of
solved in dichloromethane (20 mL), and the mixture was Compound 3a & 3b); Yield = 67%; mp 222–224 °C; IR
544 Medicinal Chemistry Research (2020) 29:538–548

(KBr) νmax: 1620 (amide, C=O), 3130–3280 (NH), 2230 Compound 4a & 4b); Yield = 71%; mp: 218–220 °C; IR
(CN) (cm−1); 1H NMR (DMSO-d6, 400 MHz): δ = 10.94 (s, (KBr) νmax: 1625 (amide, C=O), 3160–3280 (NH),
1H, N-H), 9.08–9.16 (m, 1H, H-16), 8.80 (d, J = 5.09 Hz, 1650 (C = N) (cm−1); 1H NMR (DMSO-d6, 500 MHz): δ
1H, H-14), 8.30 (d, J = 7.83 Hz, 1H, H-12), 8.15–8.22 (m, = 10.41 (s, 1H, O-H), 9.44 (s, 1H, N-H), 8.04 (d,
1H, H-10), 7.97 (d, J = 8.61 Hz, 1H, H-13), 7.88 (d, J = J = 3.63 Hz, 1H, H-19), 7.93 (s, 1H, H-12), 7.89 (d,
8.61 Hz, 1H, H-6), 7.60 (dd, J = 4.89, 8.02 Hz, 1H, H-7); 13C J = 4.67 Hz, 1H, H-17), 7.68 (d, J = 8.82 Hz, 1H, H-9),
NMR (DMSO-d6, 126 MHz): δ = 165.0 (C-2), 152.7 (C-14), 7.40 (d, J = 8.30 Hz, 1H, H-5), 7.24 (t, J = 4.15 Hz, 1H,
148.8 (C-16), 144.3 (C-5), 136.7 (C-9), 135.9 (C-12), 135.7 H-6), 5.78 (s, 1H, H-18); 13C NMR (DMSO-d6,
(C-7), 135.2 (C-1), 129.8;(C-13), 123.6 (C-6), 120.1 (C-10), 126 MHz): δ = 160.1 (C-10), 150.3 (C-1), 140.1 (C-4),
118.8 (C-17), 106.1 (C-8) Anal. Calcd for C13H8ClN3O 139.4 (C-15), 132.5 (C-19), 132.1 (C-8), 131.3 (C-17),
(257.67): C, 60.60; H, 3.13; Cl, 13.76; N, 16.31; O, 6.21; 129.6 (C-18), 128.7 (C-6), 128.2 (C-9), 120.4 (C-5),
found C, 60.56; H, 3.17; Cl, 13.71; N, 16.28; O, 6.24. 118.2 (C-7); Anal. Calcd for C12H10ClN3O2S (295.74):
C, 48.74; H, 3.41; Cl, 11.99; N, 14.21; O, 10.82; S,
General procedure for Synthesis of N-(3-chloro-4-(N- 10.84; found: C, 48.79; H, 3.53; Cl, 12.12; N, 14.20; O,
hydroxycarbamimidoyl)phenyl)thiophene-2- 10.89; S, 10.74.
carboxamide (4a) and N-(3-chloro-4-(N-
hydroxycarbamimidoyl)phenyl)nicotinamide (4b) General procedure for Synthesis of N-(3-chloro-4-(5-
aryl-1,2,4-oxadiazol-3-yl)phenyl)thiophene-2-
N-(3-chloro-4-cyanophenyl)thiophene-2-carboxamide carboxamide (6a-f) and N-(3-chloro-4-(5-alkyl-1,2,4-
(0.9 g, 3.5 mmol) or N-(3-chloro-4-cyanophenyl)nicotina- oxadiazol-3-yl)phenyl)nicotinamide (7a-f)
mide (0.9 g, 3.5 mmol) was suspended in ethanol (100 mL),
and the mixture was added to Hydroxylamine (20 mL, 50% To a suspension of N-(3-chloro-4-(N-hydroxycarbamimidoyl)
w/w in H2O) solution. The resulting mixture was refluxed phenyl)thiophene-2-carboxamide (150 mg, 0.5 mmol) or
overnight. On completion of reaction, the solution was N-(3-chloro-4-(N-hydroxycarbamimidoyl)phenyl)nicoti-
allowed to cool down to room temperature and then up to namide (145 mg, 0.5 mmol) and corresponding car-
0 °C. The separated white crystals were filtered, washed boxylic acid (0.55 mmol) in acetonitrile (1.5 mL) was
with cold ethanol and dried. (0.88 g, yield: 86%). added and the resulting mixture was heated under
microwave reactor at 150 °C for 20 min. The mixture was
N-(3-chloro-4-(N-hydroxycarbamimidoyl)phenyl)thiophene- cooled to room temperature, concentrated and residue
2-carboxamide (4a) was purified by column chromatography (DCM: Acet-
one, 5–20%).
Pale yellow solid (MeOH) (This compound was prepared
by the general procedure mentioned for the synthesis of N-(3-Chloro-4-(5-cyclopropyl-1,2,4-oxadiazol-3-yl)phenyl)
Compound 4a & 4b); Yield = 67%; mp: 228–230 °C; IR thiophene-2-carboxamide (6a)
(KBr) νmax: 1630 (amide, C=O), 3140–3260 (NH), 1640
(C = N) (cm−1); 1H NMR (DMSO-d6, 500 MHz): δ = 10.64 Yellow solid (MeOH) (This compound was prepared by
(s, 1H, O-H), 9.47 (s, 1H, N-H), 9.12 (s, 1H, H-6), 8.78 (d, the general procedure mentioned for the synthesis of
J = 4.15 Hz, 1H, H-4), 8.30 (d, J = 7.78 Hz, 1H, H-2), 7.98 Compound 6a–f); Yield = 73%; mp: 241–243 °C; IR
(s, 1H, H-18), 7.71 (d, J = 8.82 Hz, 1H, H-15), 7.59 (dd, J (KBr) νmax: 1660 (amide, C=O), 3120–3220 (NH)
= 5.45, 7.53 Hz, 1H, H-3), 7.41 (d, J = 8.30 Hz, 1H, H-11), (cm−1); 1H NMR (DMSO-d6, 400 MHz): δ = 10.57 (br.
5.80 (s, 1H, H-12); 13C NMR (DMSO-d6, 126 MHz): δ = s., 1H, NH), 8.12 (d, J = 1.56 Hz, 1H, Ar-H), 8.06 (d,
163.2 (C-7), 162.7 (C-16), 148.2 (C-6), 147.8 (C-4), 139.8 J = 3.91 Hz, 1H, Ar-H), 7.83–7.94 (m, 3H, Ar-H), 7.25
(C-8), 134.9 (C-2), 130.4 (C-1 and 14), 127.1 (C-12), 124.7 (t, J = 4.30 Hz, 1H, Ar-H), 2.35–2.46 (m, 1H, CH),
(C-3), 121.8 (C-15), 119.1 (C-11), 117.8 (C-13); Anal. 1.24–1.33 (m, 2H, CH2), 1.18 (td, J = 3.86, 7.53 Hz, 2H,
Calcd for C13H11ClN4O2 (290.70): C, 53.71; H, 3.81; Cl, CH2); 13C NMR (DMSO-d6, 101 MHz): δ = 181.0 (C-
12.19; N, 19.27; O, 11.01 found: C, 53.73; H, 3.89; Cl, 16), 166.0 (C-10), 160.3 (C-1), 142.0 (C-15), 139.1 (C-
12.39; N, 19.17; O, 10.88. 4), 132.7 (C-7), 132.2 (C-8), 132.0 (C-20), 129.9 (C-22),
128.2 (C-21), 121.1 (C-6), 120.1 (C-9), 118.4 (C-5), 10.1
N-(3-chloro-4-(N-hydroxycarbamimidoyl)phenyl) (C-17), 7.1 (C-18 and 19); LC-MS: m/z (pos): 346.21 [M +
nicotinamide (4b) H] + C16H12ClN3O2S (345.80); Anal. Calcd for
C16H12ClN3O2S: C, 55.57; H, 3.50; Cl, 10.25; N, 12.15; O,
Pale yellow solid (MeOH) (This compound was prepared 9.25; S, 9.27; found: C, 55.66; H, 3.61; Cl, 10.35; N, 12.21;
by the general procedure mentioned for the synthesis of O, 9.32; S, 9.32.
Medicinal Chemistry Research (2020) 29:538–548 545

N-(3-chloro-4-(5-methyl-1,2,4-oxadiazol-3-yl)phenyl) (DMSO-d6, 126 MHz): δ = 173.3 (C-16), 167.0 (C-10),


thiophene-2-carboxamide (6b) 160.4 (C-1), 142.4 (C-15), 139.1 (C-4), 133.5 (C-7), 132.8
(C-8), 132.3 (C-23), 132.2 (C-25), 130.1 (C-20), 128.7 (C-
Yellow solid (MeOH) (This compound was prepared by the 19), 128.2 (C-26), 127.1 (C-22), 121.2 (C-17), 119.5 (C-9),
general procedure mentioned for the synthesis of Com- 118.6 (C-5), 117.9 (CN), 115.4 (C-24); LC-MS m/z (pos):
pound 6a–f); Yield = 79%; mp: 247–249 °C; IR (KBr) 407.09 [M + H] + C20H11ClN4O2S (calcd. 406.84); Anal.
νmax: 1665 (amide, C=O), 3160–3240 (NH) (cm−1). 1H Calcd for C20H11ClN4O2S: C, 59.04; H, 2.73; Cl, 8.71; N,
NMR (DMSO-d6, 500 MHz): δ = 10.58 (s, 1H, NH), 8.12 13.77; O, 7.86; S, 7.88; found: C, 58.79; H, 2.79; Cl, 8.86;
(s, 1H, H-18), 8.07 (s, 1H, H-9), 7.85 - 7.96 (m, 3H, H-5, 6, N, 13.92; O, 7.91; S, 7.63.
20), 7.26 (s, 1H, H-19), 2.68 (s, 3H, –CH3); 13C NMR
(DMSO-d6, 126 MHz): δ = 176.6 (C-16), 166.1 (C-10), N-(4-(5-(tert-butyl)-1,2,4-oxadiazol-3-yl)-3-chlorophenyl)
160.3 (C-1), 142.0 (C-15), 139.1 (C-4), 132.8 (C-7), 132.2 thiophene-2-carboxamide (6e)
(C-8), 132.0 (C-18), 129.9 (C-20), 128.2 (C-19), 121.2 (C-
6), 120.1 (C-9), 118.5 (C-5), 11.9 (C-17); LC-MS m/z Yellow solid (This compound was prepared by the general
(pos): 320.20 [M + H] + C14H10ClN3O2S (calcd. 319.76); procedure mentioned for the synthesis of Compound
Anal. Calcd for C14H10ClN3O2S: C, 52.59; H, 3.15; Cl, 6a–f); Yield = 63%; mp: 259–261 °C; IR (KBr) νmax:
11.09; N, 13.14; O, 10.01; S, 10.03; found: C, 52.63; H, 1670 (amide, C=O), 3130–3210 (NH) (cm−1). 1H NMR
3.21; Cl, 11.13; N, 13.03; O, 9.91; S, 10.15. (DMSO-d6, 500 MHz): δ = 10.58 (br. s., 1H, NH), 8.15
(s, 1H, H-18), 8.07 (br. s., 1H, H-9), 7.85–7.99 (m, 3H, H-
N-(3-chloro-4-(5-(4-nitrophenyl)-1,2,4-oxadiazol-3-yl) 5, 6, 20), 7.25 (s, 1H, H-19), 1.45 (s, 9H, 3CH3); 13C
phenyl)thiophene-2-carboxamide (6c) NMR (DMSO-d6, 126 MHz): δ = 185.4 (C-16), 165.8 (C-
10), 160.3 (C-1), 142.0 (C-15), 139.1 (C-4), 132.7 (C-7),
Yellow solid (This compound was prepared by the general 132.2 (C-8), 132.0 (C-18), 129.9 (C-20), 128.2 (C-19),
procedure mentioned for the synthesis of Compound 6a–f); 121.2 (C-9), 120.1 (C-5), 118.5 (C-6), 33.2 (C-17), 27.9
Yield = 80%; mp: 255–257 °C; IR (KBr) νmax: 1650 (C-22, 23, 24); LC-MS m/z (pos): 362.26 [M + H] +
(amide, C=O), 3150–3240 (NH) (cm−1); 1H NMR C17H16ClN3O2S (calcd. 361.84); Anal. Calcd for
(DMSO-d6, 400 MHz): δ = 10.61 (s, 1H, NH), 8.83 (s, 1H, C17H16ClN3O2S: C, 56.43; H, 4.46; Cl, 9.80; N, 11.61; O,
H-18), 8.55 (d, J = 8.22 Hz, 1H, H-22), 8.58 (d, J = 8.84; S, 8.86; found: C, 56.57; H, 4.62; Cl, 9.69; N, 11.65;
7.82 Hz, 1H, H-23), 8.17 (d, J = 1.56 Hz, 1H, H-25), O, 8.83; S, 8.71.
8.06–8.11 (m, 2H, H-9,26), 7.96 (t, J = 8.02 Hz, 1H, H-
20), 7.90–7.94 (m, 2H, H-5,6), 7.26 (t, J = 4.30 Hz, 1H, H- N-(3-chloro-4-(5-(4-methoxyphenyl)-1,2,4-oxadiazol-3-yl)
19); 13C NMR (DMSO-d6, 126 MHz): δ = 172.7 (C-16), phenyl)thiophene-2-carboxamide (6f)
166.8 (C-110), 160.2 (C-1), 148.1 (C-24), 142.3 (C-15),
139.0 (C-4), 133.7 (C-7), 132.5 (C-8), 132.3 (C-18), 132.0 Yellow solid (This compound was prepared by the general
(C-22), 131.3 (C-26), 129.8 (C-17), 128.0 (C-20), 127.4 procedure mentioned for the synthesis of Compound 6a–f);
(C-6), 124.5 (C-19), 122.3 (C-23), 121.1 (C-25), 119.3 (C- Yield = 59%; mp: 242–244 °C; IR (KBr) νmax: 1645
9), 118.3 (C-5); LC-MS m/z (pos): 427.36 [M + H] + (amide, C=O), 3160–3250 (NH) (cm−1). 1H NMR (DMSO-
C19H11ClN4O4S (calcd. 426.83); Anal. Calcd for d6, 500 MHz): δ = 10.40 (br, s, 1H, NH), 8.75 (d, J =
C19H11ClN4O4S: C, 53.47; H, 2.60; Cl, 8.31; N, 13.13; O, 4.15 Hz, 1H, H-18), 8.29 (d, J = 7.79 Hz, 1H, H-20), 8.10
14.99; S, 7.51; found: C, 53.57; H, 2.65; Cl, 8.37; N, −8.19 (m, J = 8.82 Hz, 2H, H-9, 22), 8.12 (s, 1H, H-26),
13.19; O, 15.09; S, 7.64. 7.79 (d, J = 8.30 Hz, 1H, H-6), 7.60 (dd, J = 5.20, 7.30 Hz,
1H, H-5), 7.49 (d, J = 8.25 Hz, 1H, H-19), 7.00–7.06 (m,
N-(3-chloro-4-(5-(4-cyanophenyl)-1,2,4-oxadiazol-3-yl) J = 8.80 Hz, 1H, H-23), 6.91 (s., 1H, H-25), 3.76 (s, 3H,
phenyl)thiophene-2-carboxamide (6d) OCH3); 13C NMR (DMSO-d6, 126 MHz): δ = 165.3 (C-
16), 162.0 (C-10), 161.9 (C-1), 157.7 (C-24), 155.3 (C-15),
Yellow solid (This compound was prepared by the general 149.6 (C-4), 139.9 (C-7), 136.4 (C-8), 133.2 (C-18), 130.5
procedure mentioned for the synthesis of Compound 6a–f); (C-20), 129.3 (C-19), 128.0, (C-6), 127.8 (C-9), 125.4 (C-
Yield = 61%; mp: 251–253 °C; IR (KBr) νmax: 1640 5), 122.6 (C-17), 121.5 (C-22), 117.3 (C-26), 114.4 (C-23,
(amide, C=O), 3160–3250 (NH) (cm−1); 1H NMR (DMSO- 25), 55.9 (C-28); LC-MS m/z (pos): 412.56 [M + H] +
d6, 500 MHz): δ = 10.71 (br. s., 1H, NH), 8.33 (d, J = C20H14ClN3O3S (calcd. 411.86); Anal. Calcd for
7.78 Hz, 2H, H-18, 19), 8.19 (s., 1H, H-20), 8.10–8.16 (m, C20H14ClN3O3S: C, 58.33; H, 3.43; Cl, 8.61; N, 10.20; O,
2H, H-26, 22), 8.05 (d, J = 8.30 Hz, 1H, H-6), 7.93 (s, 1H, 11.65; S, 7.78; found: C, 58.47; H, 3.33; Cl, 8.56; N, 10.20;
H-23), 7.96 (s, 1H, H-21), 7.26 (s., 1H, H-19); 13C NMR O, 11.66; S, 7.62.
546 Medicinal Chemistry Research (2020) 29:538–548

N-(3-chloro-4-(5-cyclopropyl-1,2,4-oxadiazol-3-yl)phenyl) 8.02 Hz, 1H, H-17); 13C NMR (DMSO-d6, 101 MHz): δ =
nicotinamide (7a) 173.4 (C-21), 167.4 (C-10), 165.0 (C-1), 152.9 (C-20),
150.4 (C-18), 149.1 (C-4), 142.8 (C-16), 136.0 (C-7), 132.7
Yellow solid (This compound was prepared by the general (C-8), 132.6 (C-15), 130.3 (C-23), 129.8 (C-27), 128.9 (C-
procedure mentioned for the synthesis of Compound 7a–f); 6), 125.0 (C-24), 123.9 (C-26), 121.7 (C-17), 120.1 (C-9),
Yield = 57%; mp: 237–239 °C; IR (KBr) νmax: 1640 119.0 (C-5); LC-MS m/z (neg): 420.41 [M-H]-
(amide, C=O), 3130–3260 (NH) (cm−1). 1H NMR (DMSO- C20H12ClN5O4 (calcd. 421.79); Anal. Calcd for
d6, 500 MHz): δ = 10.85 (s, 1H, NH), 9.14 (br. s., 1H, H- C20H12ClN5O4: C, 56.95; H, 2.87; Cl, 8.40; N, 16.60; O,
20), 8.79 (br. s., 1H, H-18), 8.34 (d, J = 7.79 Hz, 1H, H- 15.17; found: C, 57.02; H, 2.93; Cl, 8.37; N, 16.50;
16), 8.18 (s, 1H, H-9), 7.91 (s, 2H, H-6, 5), 7.61 (br. s., 1H, O, 15.18.
H-17), 2.42 (br. s., 1H, H-22), 1.30 (d, J = 5.19 Hz, 2H, H-
23), 1.19 (s, 2H, H-24); 13C NMR (DMSO-d6, 126 MHz): δ N-(3-chloro-4-(5-(4-cyanophenyl)-1,2,4-oxadiazol-3-yl)
= 181.1 (C-21), 166.0 (C-10), 164.6 (C-1), 152.4 (C-20), phenyl)nicotinamide (7d)
148.7 (C-18), 142.0 (C-4), 135.8 (C-16), 132.2 (C-7), 132.0
(C-8), 130.1 (C-15), 123.6 (C-6), 121.3 (C-17), 120.4 (C-9), Yellow solid (This compound was prepared by the general
118.6 (C-5), 10.1 (C-22), 7.2 (C-23, 24); LC-MS m/z (pos): procedure mentioned for the synthesis of Compound 7a–f);
341.26 [M + H] + C17H13ClN4O2 (calcd. 340.76); Anal. Yield = 70%; mp: 241–243 °C; IR (KBr) νmax: 1630
Calcd for C17H13ClN4O2: C, 59.92; H, 3.85; Cl, 10.40; N, (amide, C=O), 3180–3270 (NH) (cm−1). 1H NMR (DMSO-
16.44; O, 9.39; found: C, 60.01; H, 3.89; Cl, 10.49; N, d6, 500 MHz): δ = 10.31 (br, s, 1H, NH), 8.32 (d, J =
16.52; O, 9.27. 7.80 Hz, 2H, H-18, 20), 8.20 (s, 1H, 16), 8.06–8.14 (m, 3H,
H-9, 23, 27), 8.10 (d, J = 8.24 Hz, 1H, H-6), 7.95 (s, 1H, H-
N-(3-chloro-4-(5-methyl-1,2,4-oxadiazol-3-yl)phenyl) 24), 7.98 (s, 1H, H-26), 7.28 (s, 1H, H-17); 13C NMR
nicotinamide (7b) (DMSO-d6, 126 MHz): δ = 175.3 (C-21), 169.0 (C-10),
165.4 (C-1), 140.4 (C-18), 138.1 (C-20), 135.5 (C-4), 134.8
Yellow solid (This compound was prepared by the general (C-16), 133.3 (C-7), 131.2 (C-8), 129.1 (C-15), 128.5 (C-22),
procedure mentioned for the synthesis of Compound 7a–f); 128.0 (C-23), 127.8 (C-27), 123.2 (C-17), 118.5 (C-9),
Yield = 71%; mp: 271–273 °C; IR (KBr) νmax: 1650 117.6 (C-5), 116.9 (CN), 114.4 (C-25); LC-MS m/z (pos):
(amide, C=O), 3150–3280 (NH) (cm−1). 1H NMR (DMSO- 402.26 [M + H] + C21H12ClN5O2 (calcd. 401.81); Anal.
d6, 500 MHz): δ = 10.81 (s, 1H, NH), 9.13 (s, 1H, H-20), Calcd for C21H12ClN5O2: C, 62.77; H, 3.01; Cl, 8.82; N,
8.79 (d, J = 3.63 Hz, 1H, H-18), 8.32 (d, J = 8.30 Hz, 1H, 17.43; O, 7.96; found: C, 62.87; H, 2.96; Cl, 8.81; N, 17.53;
H-16), 8.17 (s, 1H, H-9), 7.89–7.99 (m, 2H, H-5, 6), 7.58 - O, 7.96%.
7.62 (m, 1H, H-17), 2.68 (s, 3H, H-22); 13C NMR (DMSO-
d6, 126 MHz): δ = 176.7 (C-21), 166.1 (C-10), 164.7 (C-1), N-(4-(5-(tert-butyl)-1,2,4-oxadiazol-3-yl)-3-chlorophenyl)
152.5 (C-18), 148.8 (C-20), 142.0 (C-4), 135.6 (C-16), nicotinamide (7e)
132.2 (C-7), 132.0 (C-8), 130.0 (C-15), 123.5 (C-6), 121.3
(C-17), 120.4 (C-9), 118.6 (C-5), 11.9 (C-22); LC-MS m/z Yellow solid (This compound was prepared by the general
(pos): 315.21 [M + H] + C15H11ClN4O2 (calcd. 314.72); procedure mentioned for the synthesis of Compound 7a–f);
Anal. Calcd for C15H11ClN4O2: C, 57.24; H, 3.52; Cl, Yield = 79%; mp: 252–254 °C; IR (KBr) νmax: 1680
11.26; N, 17.80; O, 10.17; found: C, 57.16; H, 3.63; Cl, (amide, C=O), 3130–3240 (NH) (cm−1). 1H NMR
11.16; N, 17.83; O, 10.23. (DMSO-d6, 400 MHz): δ = 10.91 (s, 1H, NH), 9.18 (d, J =
1.96 Hz, 1H, H-20), 8.83 (d, J = 3.52 Hz, 1H, H-18), 8.42
N-(3-chloro-4-(5-(4-nitrophenyl)-1,2,4-oxadiazol-3-yl) (d, J = 7.82 Hz, 1H, H-16), 8.20 (d, J = 1.96 Hz, 1H, H-9),
phenyl)nicotinamide (7c) 7.98 (d, J = 8.61 Hz, 1H, H-6), 7.91–7.95 (m, 1H, H-17),
7.68 (dd, J = 4.89, 8.02 Hz, 1H, H-5), 1.46 (s, 3H, H-22);
Yellow solid (This compound was prepared by the general 13
C NMR (DMSO-d6, 101 MHz): δ = 185.4 (C-21), 165.8
procedure mentioned for the synthesis of Compound 7a–f); (C-10), 163.8 (C-1), 150.3 (C-20), 147.0 (C-18), 142.0 (C-
Yield = 76%; mp: 260–262 °C; IR (KBr) νmax: 1640 4), 138.1 (C-16), 132.2 (C-7), 132.1 (C-8), 130.7 (C-15),
(amide, C=O), 3160–3270 (NH) (cm−1). 1H NMR (DMSO- 124.4 (C-6), 121.4 (C-17), 120.6 (C-9), 118.7 (C-5), 27.9
d6, 400 MHz): δ = 10.85 (br. s., 1H, NH), 9.10–9.16 (m, (C-22); LC-MS m/z (pos): 357.31 [M + H] +
1H, H-20), 8.80 (d, J = 4.70 Hz, 1H, H-18), 8.44 (q, J = C18H17ClN4O2 (calcd. 356.81); Anal. Calcd for
9.00 Hz, 4H, H-23, 24, 26, 27), 8.32 (d, J = 7.82 Hz, 1H, H- C15H11ClN4O2: C, 57.24; H, 3.52; Cl, 11.26; N, 17.80; O,
16), 8.20 (d, J = 1.96 Hz, 1H, H-9), 8.08 (d, J = 8.61 Hz, 10.17; found: C, 57.16; H, 3.63; Cl, 11.16; N, 17.83;
1H, H-6), 7.92–8.00 (m, 1H, H-5), 7.60 (dd, J = 4.89, O, 10.23.
Medicinal Chemistry Research (2020) 29:538–548 547

N-(3-chloro-4-(5-(4-methoxyphenyl)-1,2,4-oxadiazol-3-yl) Publisher’s note Springer Nature remains neutral with regard to


jurisdictional claims in published maps and institutional affiliations.
phenyl)nicotinamide (7f)

Yellow solid (This compound was prepared by the general References


procedure mentioned for the synthesis of Compound 7a–f);
A Rattan (2000) In: BI Churchill (ed), Antimicrobials in laboratory
Yield = 79%; mp: 264–266 °C; IR (KBr) νmax: 1650 (amide, medicine. Livingstone, New Delhi, p 85–108
C=O), 3130–3210 (NH) (cm−1). 1H NMR (DMSO-d6, Bakht MA, Yar MS, Abdel-Hamid SG, Al Qasoumi SI, Samad A
500 MHz): δ = 10.66 (s, 1H, NH), 9.15 (s, 1H, H-20), 8.79 (d, (2010) Molecular properties prediction, synthesis and anti-
microbial activity of some newer oxadiazole derivatives. Eur J
J = 4.15 Hz, 1H, H-18), 8.33 (d, J = 7.79 Hz, 1H, H-16),
Med Chem 45:5862–5869
8.13–8.21 (m, J = 8.82 Hz, 2H, H-23, 27), 8.06 (s, 1H, H-9), Desai NC, Bhatt N, Somani H, Trivedi A (2013) Synthesis, anti-
7.79 (d, J = 8.30 Hz, 1H, H-6), 7.59 (dd, J = 5.19, 7.27 Hz, microbial and cytotoxic activities of some novel thiazole clubbed
1H, H-5), 7.53 (d, J = 8.30 Hz, 1H, H-17), 7.01–6.96 (m, 2H, 1,3,4-oxadiazoles. Eur J Med Chem 67:54–59
Dramowski A, Morsheimer MM, Jordaan AM, Victor TC, Donald PR,
H-24, 26), 3.86 (s, 1H, H-29); 13C NMR (DMSO-d6,
Schaaf HS (2012) Rifampicin-monoresistant Mycobacterium
126 MHz): δ = 164.3 (C-21), 163.0 (C-10), 162.9 (C-1), 155.7 tuberculosis disease among children in Cape Town, South Africa.
(C-25), 152.3 (C-20), 148.6 (C-18), 140.9 (C-4), 135.4 (C-16), Int J Tuberc Lung Dis 16:76–81
132.2 (C-7), 131.5 (C-8), 131.3 (C-15), 130.0 (C-6), 127.1 (C- Hemming K (2001) Recent developments in the synthesis, chemistry
and applications of the fully unsaturated 1,2,4-Oxadiazoles. J
17), 123.4 (C-9), 121.6 (C-5), 120.5 (C-22), 118.3 (C-23, 27),
Chem Res 209–216
113.7 (C-24), 55.4 (C-26); LC-MS m/z: Not supported Karakaya S, Yilmaz-Oral D, Kilic CS, Gur S (2019) Umbelliferone
C21H15ClN4O3 (calcd. 406.82); Anal. Calcd for C21H15ClN4O3 isolated from Zosima absinthifolia roots partially restored erectile
(406.82): C, 62.00; H, 3.72; Cl, 8.71; N, 13.77; O, 11.80; dysfunction in streptozotocin-induced diabetic rats. Med Chem
Res 28:1161–1167
found: C, 62.09; H, 3.92; Cl, 8.59; N, 13.78; O, 11.99.
Karunamoorthi K (2014) Malaria vaccine: a future hope to curtail the
global malaria burden. Int J Prev Med 5:529–538
Kumar B, Kumar A, Beheraand AK, Raj V (2016) Latest update on
Conclusion pharmacological activities of 1,3,4-oxadiazole derivatives. J Cell
Sci Ther 7:233–249
London L, Cox H, Coomans F (2016) Multidrug-resistant TB:
All the novel oxadiazole derivatives bearing thiazole and implementing the right to health through the right to enjoy the
pyridine core have been synthesized by the three-step reaction benefits of scientific progress. Health Hum Rights 18:25–41
process using a conventional method. All the new synthesized Madhu Sekhar M, Nagarjuna U, Padmavathi V, Padmaja A, Reddy
NV, Vijaya T (2018) Synthesis and antimicrobial activity of
targets were screened for their antimalarial, antituberculosis,
pyrimidinyl 1,3,4-oxadiazoles, 1,3,4-thiadiazoles and 1,2,4-tria-
antibacterial and antifungal activities with the expectation of zoles. Eur J Med Chem 145:1–10
ascertained some new structural leads hopeful as potent Malladi S, Isloor AM, Peethambar SK, Fun HK (2014) Synthesis and
antimicrobial, antimalarial and antituberculosis components. biological evaluation newer analogues 2,5-disubstituted 1,3,4-
oxadiazole containing pyrazole moiety antimicrobial agents. Arab
It can be fulfilled that the compounds 6c, 6d, 6e, 6f, 7c, 7d,
J Chem 7:1185–1191
7e, and 7f were found to be mainly skillful members of the NCCLS (National Committee for Clinical Laboratory Standards)
series. Majority of the compounds were found to be active (2002) Performance Standards for Antimicrobial Susceptibility
against C. tetani and B. subtilis. In the antifungal activity, as Testing. Twelfth Informational Supplement, 1-56238-454-6
M100-S12, M7
compared with griseofulvin compounds 6a, 6d and 7f have
Ningaiah S, Bhadraiah UK, Doddaramappa SD, Keshavamurthy S,
shown exceptional activity against C. albicans. While in Javarasetty C (2014) Novel pyrazole integrated 1,3,4-oxadia-
antituberculosis activity, compounds 6f, 7e and 7f displayed zoles: Synthesis, characterization and antimicrobial evaluation.
marvelous antituberculosis activities. Whereas in anti- Bioorg Med Chem Lett 24:245–248
O Bora R, Dar B, Pradhan V, Farooqui M (2014) [1, 2, 4]-oxadiazoles:
malarial activity, four compounds (6c, 6d, 7d, and 7f) have
synthesis and biological applications. Mini-Rev Med Chem
shown admirable activity against strains of P. falciparum as 14:355–369
compared with quinine. The presence of oxadiazole core in Padmavathi V, Sudhakar Reddy G, Padmaja A, Kondaiah P, Ali S
the synthesized compounds are predicted to be liable for the (2009) Synthesis, antimicrobial and cytotoxic activities of 1,3,4-
oxadiazoles, 1,3,4-thiadiazoles and 1,2,4-triazoles. Eur J Med
good biological activity.
Chem 44:2106–2112
Palit R, Saraswat N, Sahoo J (2016) Review on substituted 1,3,4-
Acknowledgements The author gratefully acknowledges the research oxadiazole and its biological activities. Int Res J Pharm 7:1–7.
facilities provided by the P. D. Patel Institute of Applied Sciences, Palomino JC, Martin A (2014) Drug resistance mechanisms in
CHARUSAT. Mycobacterium tuberculosis. Antibiotics (Basel) 3:317–340
Patel NB, Patel JN, Purohit AC, Patel VM, Rajani DP, Moo-Puc R,
Compliance with ethical standards Lopez-Cedillo JC, Nogueda-Torres B, Rivera G (2017) In vitro
and in vivo assessment of newer quinoxaline–oxadiazole hybrids
as antimicrobial and antiprotozoal agents. Int J Antimicrob
Conflict of interest The authors declare that they have no conflict of
Agents 50:413–418
interest.
548 Medicinal Chemistry Research (2020) 29:538–548

Patel RV, Patel PK, Kumari P, Rajani DP, Chikhalia KH (2012) molecular docking studies, antimalarial as DHFR inhibitor and
Synthesis of benzimidazolyl-1,3,4-oxadiazol-2ylthio-N-phenyl antimicrobial activities. Bioorg Med Chem 25:4064–4075
(benzothiazolyl) acetamides as antibacterial, antifungal and anti- Tresse C, Radigue R, Gomes Von Borowski R, Thepaut M, Hanh Le
tuberculosis agents. Eur J Med Chem 53:41–51 H, Demay F, Georgeault S, Dhalluin A, Trautwetter A, Ermel G,
Rane RA, Bangalore P, Borhade SD, Khandare PK (2013) Synthesis Blanco C, van de Weghe P, Jean M, Giard J-C, Gillet R (2019)
and evaluation of novel 4-nitropyrrole-based 1,3,4-oxadiazole Synthesis and evaluation of 1,3,4-oxadiazole derivatives for
derivatives as antimicrobial and anti-tubercular agents. Eur J Med development as broad-spectrum antibiotics. Bioorg Med Chem
Chem 70:49–58 27:115097
Salahuddin, Mazumder A, Yar MS, Mazumder R, Chakraborthy GS, malERA Consultative Group on Drugs (2011) A research agenda for
Ahsan MJ, Rahman MU (2017) Updates on synthesis and biological malaria eradication: drugs. PLOS Med 8:e1000402
activities of 1,3,4-oxadiazole: a review. Synth Comm 47:1805–1847 WHO (2011) The sixteenth global report on tuberculosis. WHO,
Sandhu GK (2011) Tuberculosis: current situation, challenges and over- Geneva, Switzerland
view of its control programs in India. J Glob Infect Dis 3:143–150 WHO (2018) World malaria report. https://www.who.int/malaria/
Singh R, Chouhan A (2014) Various approaches for synthesis of 1,3,4- publications/world-malaria-report-2018/en/
Oxadiazole derivatives and their pharmacological activity. World Zhou L, Wang P-Y, Zhou J, Shao W-B, Fang H-S, Wu Z-B, Yang S
J Pharm Pharm Sci 3, 10:1474–1505 (2017) Antimicrobial activities of pyridinium-tailored pyr-
Thakkar SS, Thakor P, Doshi H, Ray A (2017) 1,2,4-Triazole and azoles bearing 1,3,4-oxadiazole scaffolds. J Saudi Chem Soc
1,3,4-oxadiazole analogues: Synthesis, MO studies, in silico 21:852–860

You might also like