You are on page 1of 35

European Journal of Medicinal Chemistry 123 (2016) 596e630

Contents lists available at ScienceDirect

European Journal of Medicinal Chemistry


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

Review article

Synthetic strategy with representation on mechanistic pathway for


the therapeutic applications of dihydroquinazolinones
K. Hemalatha, G. Madhumitha*
Chemistry of Heterocycles & Natural Product Research Laboratory, Department of Chemistry, School of Advanced Sciences, VIT University, Vellore, Tamil
Nadu 632014, India

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

Article history: Dihydroquinazolinones is an important core structure reported with a wide variety of pharmacological
Received 18 May 2016 activities. They are capable of undergoing various transformations because of its reactivity towards
Received in revised form various reagents. The synthetic strategies for the functionalization and derivatization of the nucleus were
1 August 2016
explained. The diversified pharmacological actions of this moiety were illustrated through various
Accepted 1 August 2016
Available online 3 August 2016
biochemical pathways. The structural-activity relationship study of dihydroquinazolinones anticipated
the relationship between the various substituents and its role in the pharmacological action. The main
objective of this review is to summarize the importance of dihydroquinazolinones in the field of chemical
Keywords:
Dihydroquinazolinones
biology.
Synthetic strategy © 2016 Elsevier Masson SAS. All rights reserved.
Functionalization
Pharmacological action
Biochemical pathway

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 597
2. Synthesis and therapeutic applications of dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 597
2.1. M1/M4 mAchR agonist . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 597
2.2. MPO inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 598
2.3. Antibacterial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 598
2.4. IRAP inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 599
2.5. Antimalarial activity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 601
2.6. Chorismate mutase inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 602
2.7. Cyclooxygenase inhibitors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 604
2.8. Antitumor agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 606
2.9. 5-HT2c receptor agonist . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 607
2.10. Melanoma inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 608
2.11. T-type calcium channel antagonist . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 608
2.12. IMPDH inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
2.13. Sodium/calcium exchange inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 610
2.14. PDE7 inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 613
2.15. p38 MAPK inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 615
2.16. CDK5 inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 617
2.17. RyR agonist . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618
2.18. Non-nucleoside reverse transcriptase inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 618
2.19. Aldosterone synthase inhibitor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621
3. Miscellaneous synthesis and applications of dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621

* Corresponding author.
E-mail addresses: dr.g.madhumitha@gmail.com, madhumitha.g@vit.ac.in
(G. Madhumitha).

http://dx.doi.org/10.1016/j.ejmech.2016.08.001
0223-5234/© 2016 Elsevier Masson SAS. All rights reserved.
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 597

3.1. Synthesis of 2, 3-dihydroquinazolin-4(1H)-ones from N-heterocyclic carbenes of indazole . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621


3.2. Copper oxide nanoparticle mediated synthesis of dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621
3.3. Y(OTf)3 mediated synthesis of dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 621
3.4. Boric acid mediated synthesis of dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 622
3.5. Visible light mediated synthesis of dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 623
3.6. Importance of 7-fluoro-2, 2-dimethyl-2, 3-dihydroquinazolin-4(1H)one . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 625
3.7. Bromination of 2, 3-dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627
3.8. Suzuki Miyaura coupling of 2, 3-dihydroquinazolinones . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627
4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 627

1. Introduction reuptake. Quinethazone [34], fenquizone and metolazone are


thiazide-like diuretics [35] and they are used to treat hypertension.
Medicinal chemistry is an interdisciplinary area which links BIBN 4096 BS [36] is an antagonist of CGRP and it is used for
both organic chemistry and biology [1]. The compounds isolated migraine headaches. DPC083, DPC961, and DPC963 are NNRTIs and
from the natural resources [2,3] and by the synthesis [4e6] were they act as a synergistic agent in the treatment of HIV-1 patient
identified with numerous applications in various fields. In some [37]. Due to the tremendous increase in the number of research
cases, the extracts from the natural resources were used as such outputs related to quinazolinone nucleus, the separate collections
and the active components present in it either induce chemical of dihydroquinazolinones will be an efficient tool to assess the
reaction [7e9] or biological response [10e16]. Nitrogen-rich het- properties of the nucleus. Recently enantioselective synthesis of
erocycles are present ubiquitously in natural products, pharma- dihydroquinazolinone based anti-HIV agents were reviewed [38].
ceutical drugs and agrichemicals [17]. Among the various The main scope of this review is to elaborate the synthetic scheme
heterocycles [18e22], quinazolinones [23e25] are of prime and the various reagents utilized in the functionalization pathway
importance because of its ease in synthetic feasibility from less of dihydroquinazolinones. The review also explains the diverse
expensive chemicals [26]. Quinazolinones can be synthesized from pharmacological actions mechanistically portrayed through the
a wide variety of starting materials and also widely distributed in different biochemical pathways.
natural sources [27,28]. The reviews concentrating on the synthetic
and isolated quinazolinones are maintained up to date [29,30]. The 2. Synthesis and therapeutic applications of
quinazolinones can also exist in the form of 2, 3- dihydroquinazolinones
dihydroquinazolinones [31] and 3, 4-dihydroquinazolinones [32]
derivatives. The dihydroquinazolinones were found incorporated 2.1. M1/M4 mAchR agonist
in the marketed drugs and in the drugs undergoing clinical trial
(Fig. 1). Evodiamine [33] is an alkaloid isolated from the fruits of The mAchR subtypes M1 and M4 can regulate the cognitive
Evodia rutaecarpa. It acts as a fat burner by increasing the number of deficits and psychosis disorders and thereby its stimulation can
serotonin transporters in the brain and enhancing the serotonin improve the symptoms of schizophrenia. The markedly available

Fig. 1. Drug moieties containing dihydroquinazolinones.


598 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

M1 and M4 mAchR agonist, xanomeline produces unwanted acid (Fig. 3) [41]. Hypochlorous acid also causes oxidative damage
gastrointestinal stress because of its action through the M3 receptor to the host tissue. Endothelial-derived NO was utilized by MPO and
(Fig. 2) [39]. The importance of the M1 and M4 mAchR agonistic thereby reducing its bioavailability. Reduction in NO bioavailability
activity was identified and the SAR study was performed for the impairs its vasodilating and anti-inflammatory property [42]. Li
series of dihydroquinazolinone derivatives [40]. Based on the high et al. [43] designed three series of compounds (12, 16 and 18) and
throughput screening, a hit compound 1 with weak M4 mAchR they were tested for their activity against MPO inhibition. The
agonist was selected as a potential compound. Through the earlier synthesis of compound series 12 was carried out from the reaction
reports, the pharmacophore N-carbethoxy-piperidine moiety was of isatoic anhydride 10 with various substituted alkyl amines and
thought to be essential for the M4 mAchR agonistic activity. The carbon disulfide (Scheme 2). Another series of compounds 16 were
change of the benzyl group of 1 by N-carbethoxy-piperidine moiety obtained from the reaction between benzamine 13 and thio-
2 activated the M2 agonist activity along with M1 and M4 mAchR phosgene 14 followed by reflux with alkyl hydrazine. The treatment
agonist activity. The removal of the methylene group 3 potentiated of thiocyanate 15 with hydrazine and chlorocarbonyl reagent
the activity towards all the (M1, M2, M3, M4, M5) receptors. There- resulted in the formation of another series of compounds 18. The
fore, the importance of the methylene group in increasing the SAR study concluded that halogen in 6th position and free amino
selectivity towards the receptor subtype was identified. The group in the 3rd position of quinazolinone were stringent for the
replacement of piperidine as a linker to the various cyclic amine activity. Either change in the position of halogen/alteration of the
linkers affected the M1 and M4 mAchR agonistic activity without substituent in the place of halogen decreased the potency. Also, the
receptor subtype selectivity. The increase and decrease in the replacement of hydrogen in the amine group by other substituents
agonist activity of the M1 and M4 mAchR were resulted for the R- weakened the potency. An aromatic ring attached to NeN linkage of
substituted 4 pyrrolidine linker and the S-substituted 5 pyrrolidine compound 16, 18 was more active than the aliphatic chain con-
linker respectively. The presence of azetidine 6 lin ker also poten- taining compounds. The water solubility of the compound was
tiated the activity. The M4-mAChR partial agonistic activity was increased by introducing the methylene piperidinyl group 19. In
obtained through the replacement of N-carbethoxy piperidine by short, thioxo-dihydroquinazolinone derivatives were proved to be
N-carbethoxy tropane 9 moiety. This highly active compound was effective inhibitors of MPO and can be utilized in the treatment of
tested for its pharmacokinetic profile. This compound is less inflammation-related neurodegenerative diseases and atheroscle-
affected by liver enzymes, high cell permeability, effective brain/ rosis. These compounds act reversibly and do not trap the enzyme
blood penetration and good affinity for G-protein coupled re- which showed its superiority over the existing inhibitors.
ceptors. The in vivo pharmacological profile resulted in the reversal
of METH-induced hyperlocomotion in rats and produced a potent 2.3. Antibacterial activity
antipsychotic effect than the standard drug xanomeline. Finally, the
active compound was synthesized from sodium triacetoxyborohy- Ma et al. [44] reported the synthesis of 2-substituted-3-(phe-
dride mediated reduction reaction between the readily available nylamino)-dihydroquinazolin-4(1H)-ones through cascade reac-
starting material, (S)-3-(pyrrolidin-3-yl)-3, 4-dihydroquinazolin- tion from isatoic anhydride 20 and phenyl hydrazine 21 (Scheme 3).
2(1H)-one 7 and ethyl 3-formyl-8-azabicyclo [3.2.1] octane-8- The reaction was carried out using different acid catalyst and the
carboxylate 8 (Scheme 1). most efficient catalyst was found to be bentonite. Water was found
to be the best solvent and the reaction was carried out in the
2.2. MPO inhibitor presence of ultrasonic irradiation. Optimizing the amount of cata-
lyst concluded that the change in the concentration (1e7 mol %)
MPO is an enzyme from heme peroxidase superfamily. MPO acts displayed no significant change in the yield. However reduction in
a catalyst in the production of endogenous oxidant, hypochlorous the concentration of bentonite (0.5 mol %) decreased the

Fig. 2. Mechanism of action of M1/M4 muscarinic acetylcholine receptor agonist.


K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 599

Scheme 1. Synthesis of dihydroquinazolinones as an M1/M4 mAChRs agonist.

Fig. 3. Mechanism of action of myeloperoxidase inhibitor.

percentage yield. These optimized reaction conditions were uti- requires three substrates: ATP, BCCP and CO2 and they are con-
lized in the synthesis of various 2-substituted-3-(phenylamino)- verted into ADP, phosphate and carboxybiotin-carboxyl-carrier
dihydroquinazolin-4(1H)-ones 22. The derivatives of dihy- protein (Fig. 4). The active compounds displayed hydrogen
droquinazolin-4(1H)-ones with electron-donating substituents bonding interactions with the amino acid residues such as Leu204,
resulted in good yields when compared to the electron- Lys159, and Gly166 of biotin carboxylase enzyme.
withdrawing substituents. The in vitro antibacterial activity
against the gram-negative bacteria Escherichia coli (E. coli) was 2.4. IRAP inhibitor
carried out for all the synthesized derivatives. The antibacterial
potency of the highly active compounds of dihydroquinazolin- The spiro-oxindole containing dihydroquinazolinones were
4(1H)-ones was supported through the docking studies with the synthesized from 5-bromo-1-methyl isatin 23, isatoic anhydride 20
biotin carboxylase enzyme. Biotin carboxylase (E. coli) is an and substituted aniline (Scheme 4) [47]. The time taken for the
important enzyme involved in the fatty acid biosynthesis [45]. This conventional synthesis can be reduced from 2 h to 10 min by car-
pathway is utilized by the bacteria for the membrane biogenesis. rying out the reaction under MW irradiation. The reaction carried
Enzymes involved in this pathway were proved to be a valuable out in the borosilicate glass reactor by MW batch synthesis end up
target for the antibacterial drug discovery [46]. This enzyme with the unreacted starting material along with unidentified
600 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 2. Synthesis of thioxo-dihydroquinazolinone derivatives.

Scheme 3. Synthesis of 2-substituted-3-(phenylamino)-dihydroquinazolin-4(1H)-ones.

byproducts. The flow rate of 1 mL/min and the temperature of equivalence of isatoic anhydride and p-toluidine with the flow rate
200  C were optimized to get an isolated yield of 40% for the re- maintained at 0.5 mL/min at a temperature of 160  C resulted in the
action mixture collected after 5 min. The flow rate of 0.5 mL/min completion of the reaction with the highest yield (82%). The other
and the temperature of 120  C were utilized in the SiC glass reactor derivatives of dihydroquinazolinones were synthesized using the
with the CF of MW irradiation to obtain an isolated yield of 55%. The optimized conditions of MW-SiC-CF method. The aliphatic amines
experimentation was carried out in borosilicate reactor using the were not utilized in the reaction because of the drawback of pro-
same reaction condition and the yield was only 8%. The SiC glass tonation. IRAP is a transmembrane protein and it belongs to the
reactor was advantageous over the borosilicate glass reactor in family of aminopeptidases. It is also known as an AT4 receptor, a
terms of low temperature, less complex reaction mixture and binding site for the peptide angiotensin IV. Angiotensin IV is the
thereby it leads to the easy way of purification. Both in the batch metabolite obtained from the biologically active peptide fragment,
synthesis and CF, there is an existence of 5-bromo-1-methyl isatin angiotensin II. The role of angiotensin IV is to enhance the memory
in the reaction mixture. The reaction proceeded to completion by and learning. Therefore, IRAP based inhibitors are useful for
increasing the equivalence of p-toluidine. Increasing the equiva- improving cognition and develop an effective treatment for de-
lence of p-toluidine 24 lead to substantial increase in the yield of mentia (Fig. 5) [48,49]. The IRAP inhibitory activity of spiro-
spiro-oxindole dihydroquinazolinones 25. The reaction mixture oxindole dihydroquinazolinones concluded that the 5th position
containing 1 equivalence of 5-bromo-1-methyl isatin, 1.5 bromine atom is essential for the activity. The absence of bromine
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 601

Fig. 4. The role of biotin carboxylase in E. coli fatty acid biosynthesis.

Scheme 4. Synthesis of spiro-oxindole containing dihydroquinazolinones.

atom decreased the activity whereas the changes in the position of Derbyshire et al. reported the effectiveness of various compounds
the bromine atom lead to the inactive compound. In order to carry that acts as an inhibitor against both the liver and blood stage of
out the SAR study, spiro-oxindole dihydroquinazolinones were malaria [54]. The malarial drugs act through different mechanism
subjected to Suzuki Miyaura coupling reaction. The MW batch because of the difference in the transcriptomic and proteomic data
synthesis was carried out using this optimized reaction condition. of the liver and blood stage parasites. Preliminary high-throughput
The result concluded that 1 equivalence of halide 25, 2 equivalence phenotypic blood stage malarial screening identified compounds of
of boronic acid 26, 1.5 equivalence of DBU, 2 mol% PdCl2(dppf), different chemotype which showed more than 80% inhibition
acetonitrile with 2% water was appropriate to carry out the reac- against the malarial parasites. The compounds on further screening
tion. Using this optimized condition, the coupling reaction was identified hit with inhibition against blood stage parasites. These
carried out in the borosilicate reactor using CF mode. The reactants selected libraries of compounds were tested for its effectiveness
got consumed at the flow rate of 0.5 mL/min at 220  C with an against liver stage parasites in infected human hepatoma HepG2
isolated yield of 71%. Because of the inactive nature of Suzuki cells. The results explored that most of the compounds were not
Miyaura product 27 towards the IRAP inhibition further optimiza- effective against liver stage parasites. This difference in activity was
tion on the reaction methodology was not carried out. assumed to be either due to the process such as hemozoin forma-
tion or because of the metabolic instability of the compounds to-
2.5. Antimalarial activity wards the liver cell. Among the hits, 32 compounds were active
against the liver stage parasites and 22 compounds displayed EC50
Malaria is one of the deadliest diseases and the treatment value less than 10 mM. The potential hit containing dihy-
mainly relies on the chemotherapy [50e52]. Even though there are droquinazolinone 28 (EC50 ¼ 1.4 mM) were further subjected to SAR
potential drugs for malaria, the treatments are no longer effective analysis (Fig. 7). The modification in the 2nd position of dihy-
due to the evolution of resistance. The present scenario is to focus droquinazolinone using bulky ring system revealed no change in
primarily on the complicated life cycle of the parasite [53] (Fig. 6) activity. This confirmed that the binding site residues are not ste-
and develop molecules that reduce the mortality rate effectively. rically hindered. The alteration of substituents on the phenyl ring of
602 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Fig. 5. Mechanism of action of IRAP inhibitor as cognitive enhancers.

Fig. 6. The life cycle of the malarial parasite and the site of action of dihydroquinazolinones.

the compound 28 changed the activity. The elimination of liver was essential for activity, whereas the compounds with other group
stage activity was reported in the ortho substituents whereas the were less potent. The presence of 3-dialkyl aminoethyl group was
para and meta substituents were well tolerated. Variation in the 3rd found to be lethal for the blood stage parasite. Any factor which
position of compound 28 indicated that 3-dialkyl aminoethyl group increases the liver stage activity correspondingly increased the
blood stage activity. Through the SAR study, the compound 29 was
found to be an ideal candidate for both liver and blood stage ma-
laria parasites.

2.6. Chorismate mutase inhibitor

CM is an important enzyme that catalyzes the isomeric con-


version of chorismate to prephenate (Fig. 8) [55]. This is the key
reaction involved in the biosynthesis of aromatic amino acids
essential for the survival of microorganisms and plants. CM is the
Fig. 7. Quinazolinones for blood and liver-stage malarial parasites. only naturally occurring enzyme catalyzing pericyclic reaction.
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 603

Fig. 8. The role of chorismate mutase in amino acid synthesis.

Scheme 5. Synthesis of spiro 2, 3-dihydro-quinazolin-4(1H)-ones.

Since this pathway is not present in the mammals, it can be used a in the presence of oxygen (Scheme 5). The standard CM inhibitor, 4-
selective pathway for the treatment of tuberculosis [56]. Rambabu (3, 5-dimethoxyphenethylamino)3-nitro-5-sulfamoylbenzoic acid
et al. synthesized the series of spiro 2, 3-dihydro-quinazolin-4(1H)- displayed IC50 value less than 10 mM. Among the synthesized series,
ones 30 using Amberlyst-15 as a catalyst and ultrasound irradiation compound 31 and 32 were reported with 30e35% of CM inhibition

Fig. 9. Mechanism of action of anti-inflammatory agents.


604 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 6. Synthesis of 2, 3-diaryl 2, 3-dihydro-1H-quinazolin-4-ones.

Scheme 7. Synthesis of 3-aryl-2-substituted 1, 2- dihydroquinazolin-4(3H)-one metal complexes.

when compared to the other derivative. Because of very few reports 2.7. Cyclooxygenase inhibitors
on CM inhibition and increasing resistance to antitubercular ther-
apy, these derivatives are of pharmaceutically important [57]. The reactions involving the conversion of arachidonic acid to

Fig. 10. The effect of an antitumor agent on the cell cycle.


K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 605

Scheme 8. Synthesis of isoxazoline/oxazoline and dihydroquinazolinone derivatives. (i)Benzamide, p-toluene sulfonic acid monohydrate, DMA; (ii) Br(CH2)nBr, K2CO3, DMF.

prostaglandins were catalyzed by the COX enzyme. There are two whereas highly active towards the COX-2 enzyme inhibition. The
isoforms of COX referred to as COX-1 and COX-2. The COX-1 is a in vivo anti-inflammatory activity of the compounds was evaluated
constitutive enzyme and it is involved in the various physiological by carrageenan-induced rat paw edema method. The compounds
processes for maintaining the homeostasis. The COX-2 is induced in which showed positive results in the in vitro assay were correlated
case of the inflammatory condition and it is the target for the se- with the in vivo methodology. Among the active series except 39, all
lective NSAIDS (Fig. 9). The drugs selective for the COX-2 enzyme the other compounds belong to methyl sulfonyl series and they are
can be designed on the basis of difference in the amino acid selective towards the COX-2 enzyme [59].
sequence of both the enzymes [58]. Manivannan et al. designed the Hoonur et al. evaluated the analgesic and anti-inflammatory
molecule by applying the analog based pharmacophore design. The activity of the 3-aryl-2-substituted 1, 2- dihydroquinazolin-4(3H)-
quinazolinone nucleus was selected from the two well known one derivatives and its various transition metal complexes. The
natural products, rutaecarpine, and tryptanthrin. The diaryl and the dihydroquinazolin-4(3H)-one ligand 43 was prepared by the
methyl sulfonyl pharmacophore were chosen from the selective condensation of o-aminobenzoylhydrazine 41 with 2-
COX-2 inhibitors, celecoxib, and rofecoxib. The designed com- acetylpyridine 42 and followed by the treatment with benzalde-
pounds were categorized into methyl sulfanyl and methyl sulfonyl- hyde. The transition metal complexes 44 were obtained by
containing series. The various substituted anilines 33 were refluxing the ligand with the transition metal (II) chloride (Cu, Zn,
condensed with 4-thiomethyl benzaldehyde 34 to yield 4- Mn, Ni, Co and Cd) in ethanol (Scheme 7). The analgesic activity was
methylsulfanyl-benzylidene)-phenyl-amine 35. The oxidation of evaluated by acetic acid induced writhing method. The induction of
4-methylsulfanyl-benzylidene)-phenyl-amine 35 into (4-methyl acetic acid generated the visceral type of pain and produced
sulfonyl-benzylidene)-phenyl-amine 37 was carried out in the stretching of hind limbs and bending of trunks. The anti-
presence of oxone medium. The intermediate azomethines 35 & 37 inflammatory activity was carried out by the carrageenan-
were microwave irradiated with isatoic anhydride resulted in the induced rat paw edema method. The copper complex displayed
formation of 2, 3-diaryl 2, 3-dihydro-1H-quinazolin-4-ones 36 & 38 significant analgesic and anti-inflammatory activity than the ligand
(Scheme 6). The compounds were screened for in vitro anti- and other metal complexes. The effectiveness of the metal complex
inflammatory activity using ovine cyclooxygenase enzyme. The may be due to the metal chelation which increased the lipophilic
compound 40 was less active to inactive towards COX-1 enzyme character [60].
606 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 9. Synthesis of dihydroquinazolinones containing anticancer activity.

2.8. Antitumor agents (Fig. 10) are useful in the cancer chemotherapy [61]. The hybrid
molecules that inhibit microtubule polymerization were synthe-
Microtubules are the principle component of cytoskeleton and sized through the formation of ether linkage between the hydroxyl
function in the separation of chromosomes during mitosis. The two group of 3, 5-diaryl isoxazoline/3, 5-diaryl oxazoline derivatives
closely related polypeptides a-tubulin and b-tubulin are polymer- and dihydroquinazolinones [62]. The synthesis of 2, 3-
ized to form the microtubules. Microtubules play a major role in dihydroquinazolinones 46 was carried out by the condensation of
mitosis and the drugs which inhibit microtubule polymerization aldehydes 45 and benzamide in the presence of p-toluene sulfonic

Fig. 11. Mechanism of action of antiobesity drugs.


K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 607

Scheme 10. Synthesis of 6, 6, 6- tricyclic dihydroquinazolinones. (i) (COCl)2/CH2Cl; (ii) NH3/MeOH-THF; (iii) H2/10% palladium on carbon/MeOH; (iv) N-boc-aminoacetaldehyde/
pTsOH/toluene; (v) Na2CO3 or K2CO3/allyl bromide/DMF or DMA/140  C; (vi) OsO4/NaIO4/THF-H2O; (vii) TFA/Et3SiH/CH2Cl2.

acid monohydrate. The hydroxyl group of phenyl ring in the 2nd lengthiest N-alkyl substituents such as cyclopropyl, isopropyl and
position of dihydroquinazolinones was reacted with dibro- sec-butyl showed favorable interaction in the binding pocket and
moalkanes for further condensation reaction to yield the product thereby it leads to the highest cytotoxic potency of the compounds
47. The condensation reaction between 3, 5-diaryl isoxazoline/3,5- [65].
diaryl oxazoline 48 and 2, 3-dihydroquinazolinones 49 using po-
tassium carbonate afforded the hybrid compounds 50 (Scheme 8). 2.9. 5-HT2c receptor agonist
The compound was effective against nine types of cancer and 60
different cell lines. Based on the potency of the compound, the The drugs targeting central 5-HT system has been implicated in
other derivatives were also tested for their activity against the cell the treatment of various ailments. The 5-HT2C receptor agonism is
lines. Most of the derivatives displayed significant activity against the valid target for the treatment of obesity [66]. This was
MCF-7 (breast cancer) and PC3 (prostate cancer) cell lines. Through confirmed through the 5-HT2C receptor knockout mice, which
the series of evaluation, the plausible mechanism of action for the displayed hyperphagia and developed obesity. There are three
anticancer activity of the potent compound 51 was explored. The members of the 5-HT2 receptor subclass (5-HT2A, 5-HT2B, 5-HT2C)
inhibition of tubulin polymerization caused downregulation of sharing close sequence homology. The wide distribution of 5-HT2B
CDK1 and cyclin B1 protein which in turn arrested the cell cycle subtype in the vascular and cardiac tissues causes valvulopathy. The
progression in G2/M phase. drugs acting through the 5-HT2A receptor subtype results in the
Roopan et al. [63] investigated the three-component reaction of unwanted CNS related disorders [67,68]. To overcome these
isatoic anhydride 20, heterocyclic aldehyde 52 and ammonium adverse effects the molecules which are selective towards the 5-
acetate using different catalysts (Scheme 9). The montmorillonite HT2C receptor subtype have to be designed. The activation of hy-
K10 was superior in terms of time and yield. Further, the cytotox- pothalamic 5-HT2C receptors results in the stimulation of POMC, a
icity of the compounds was tested against the Ehrlich Ascites Car- precursor of a-MSH. The binding of a-MSH to the MC4R stimulates
cinoma tumor cells. They displayed comparative cytotoxicity to that the feeling of fullness (Fig. 11). Three compounds series were
of the standard drug 5-fluorouracil. designed on the basis of recently approved 5-HT2C receptor agonist,
The Schiff bases [64] of 2, 3- dihydroquinazolinone derivatives Lorcaserin. The 2-nitrobenzoic acids 60 are converted into their
55 were obtained by refluxing one equivalent of amino- corresponding acid chlorides by the treatment with oxalyl chloride.
benzyhydrazide 53 with two equivalent of substituted aromatic Further reaction with ammonia followed by reduction using
salicylaldehyde 54 in methanol for 2 h (Scheme 9). The compounds hydrogen and palladium on carbon afforded 2-aminobenzamides
were screened for their in vitro anticancer activity against MCF-10 61. The condensation of 2-aminobenzamides with N-boc-amino-
breast cell line, normal WRL-68 hepatic cell line, and human acetaldehyde in the presence of p-toluenesulfonic acid resulted in
MCF-7 breast adenocarcinoma cell line by MTT assay. The com- the cyclized product 62. The N1 of the cyclized product was sub-
pounds showed suppressive action against only MCF-7 breast jected to allylation using allyl bromide to the yield N-allyl
adenocarcinoma cell line. substituted dihydroquinazolinones 63. In another reaction, 2-
Mahdavi et al. synthesized the trans-stilbene scaffold containing fluorobenzoyl chlorides 65 are converted into their corresponding
N-substituted 2-arylquinazolinones with anticancer property. Isa- 2-fluorobenzamides and allylated 66 in the presence of ammonia
toic anhydride 20 was used as a starting material and it was con- and allylamine. The cyclized product 67 was obtained using N-
verted into anthranilamide 56 using primary amines. The protected-3-amino-1-propanal. All these resulting olefins 63 and
condensation of anthranilamide 56 with trans-stilbene 57 deriva- 67 were converted into intermediate aldehydes in the presence of
tive using potassium carbonate resulted in the formation of 2, 3- sodium periodate and a catalytic amount of osmium tetroxide. The
dihydroquinazolinone derivative 58. Dihydroquinazolinones 58 cyclization of these intermediate aldehydes with TFA and trie-
were converted into quinazolinones 59 using TBAB mediated re- thylsilane resulted in the formation of 6, 6, 6-tricyclic dihy-
action (Scheme 9). The synthesized series were tested for their droquinazolinones 64 (Scheme 10) and 6, 6, 7-tricyclic
anticancer activity against three cancer cell lines (MCF-7, MDA-MB- dihydroquinazolinones 68 (Scheme 11). The compounds containing
231, T-47D). Among the tested compounds, N-alkyl substituted halogens 69 & 70 were subjected to Stille-type coupling reaction 71,
derivatives displayed higher anticancer activity than the corre- Suzuki Miyaura coupling 72 and Negishi coupling 73 with tetra-
sponding N-aryl and N-benzyl derivatives. The bulkiest and methyltin, trimethylboroxine, and dimethyl zinc respectively. The
608 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 11. Synthesis of 6, 6, 7-tricyclic dihydroquinazolinones. (i) NH3/MeOH-THF; (ii) allylamine/K2CO3/DMA; (iii) N-cbz-3-amino-1-propanal/N-boc-3-amino-1-propanal/
pTsOH/dioxane; (iv) OsO4/NaIO4/THF-H2O; (v) Et3SiH/TFA/CH2Cl2; (vi) Boc anhydride/Et3N/THF; (vii) Me4Sn/Pd(Ph3P)4/LiCl/DMF; (viii) TFA/CH2Cl2; (ix)H2/10% Pd/C/MeOH; (x)
Pd(Ph3P)4/trimethylboroxine/K2CO3,H2O-Dioxane; (xi) PdCl2(dppf)2/Me2Zn/dioxane; (xii) BH3eMe2S/Et2O, H2O2/NaOH; (xiii) Dess-Martin periodinane/CH2Cl2.

deprotection was carried out by using TFA. The reduction in the expressed out. This enzyme is responsible for the formation of
presence of hydrogen and palladium on carbon afforded unsub- melanin pigment through the series of reactions. If there is any
stituted dihydroquinazolinones 74 & 75 (Scheme 11). The another change in the melanogenetic pathway, it will affect the pigmenta-
isomeric 6, 6, 7-tricyclic dihydroquinazolinone 77 was obtained tion process (Fig. 12). Thangamalai [71] et al. reported the synthesis
from the alcohol containing dihydroquinazolinone 76 in which the of quinazolinones and quinazoline-2-thiones with its activity pro-
alcohol group was introduced by means of borane reagent followed file against a-MSH induced melanin production. The amino com-
by hydrogen peroxide treatment. The alcohols are converted into pounds 78 on treatment with CDI in the presence of triethylamine
aldehydes using Dess-Martin periodinane as an oxidizing agent. yielded dihydroquinazolinone derivatives 79. The quinazolinones
The TFA and triethylsilane mediated cyclization afforded the were converted into quinazoline-2-thione 80 by refluxing over-
isomeric 6, 6, 7-tricyclic dihydroquinazolinone 77 (Scheme 11) [67]. night with LR (Scheme 12). Quinazolinones displayed high IC50
All the synthesized series for tested for their agonism against 5- value (>10 mM) when compared to the quinazoline-2-thione in a-
HT2C receptors and also its functional selectivity towards 5-HT2A MSH induced melanin production in B16 melanoma cells. The
and 5-HT2B. The functional selectivity towards 5-HT2c receptors and smaller substituent does not affect the activity whereas the sub-
the absence of off-target effects was achieved using 6, 6, 6-tricyclic stitution of chlorine in the quinazoline-2-thione ring system
dihydroquinazolinones. The 6, 6, 6-tricyclic dihydroquinazolinones resulted in slight variation. The presence of bulky ring system and
displayed suboptimal functional selectivity and reduced brain to the addition of substituents in the side chain phenyl ring decreased
plasma concentration. the activity. The number of methylene unit displayed marginal
variation in activity but one methylene unit was found to be opti-
mum. While predicting the mechanism of action, quinazoline-2-
2.10. Melanoma inhibitor thione inhibited the a-MSH induced melanin production in B16
melanoma cells whereas the normal catalytic activity of the
Melanogenesis is a complex pathway and several proteins are tyrosinase enzyme was not affected.
involved in this pathway [69,70]. The process is initiated by the
paracrine stimulator, POMC. It is the precursor of a-MSH, an
endogenous peptide hormone of melanocortin family. Due to the 2.11. T-type calcium channel antagonist
induction of UV rays, the expression of POMC occurs in keratino-
cytes. The a-MSH cleaved from POMC acts through the MC1R. The The role of calcium ions has been implicated in various phys-
remaining processes of melanogenesis are carried out by the sec- iopathological processes [72]. There are various types of calcium
ond messenger, cAMP. The transcriptional activity was regulated channels in which T-type calcium channel plays a major role in the
through the phosphorylation of CREB and activation of the regu- regulation of sleep, nociception, epilepsy, hypertension and cell
latory protein, MITF. Through the activation of the melanogenesis- cycle pathway. Because of the distribution of T-type calcium
related protein, the melanogenetic enzyme tyrosinase was channel in the thalamus and cortex region, they are identified as
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 609

Fig. 12. Mechanism of action of melanoma inhibitor.

Scheme 12. Synthesis of quinazolinone and quinazoline-2-thione.

effective targets in the disorders of thalamocortical signaling the quinazolinone ring system 90. The presence of trifluoro ethyl
pathway [73]. As per the reports of FLIPR tetra high-throughput group in the 3rd position of quinazolinone is maintained constant
cellular screening system, two piperidines 81, 82 and one quina- because of its potency and metabolic stability. The halogen
zolinone analogs 83 were identified as T-type calcium channel replacement in the 4th position phenyl ring 91 was carried by using
antagonists for the treatment of epilepsy with minimal cardiovas- either Suzuki Miyaura coupling reaction or by using cuprous iodide
cular side effects. Barrow et al. [74] designed the series of 4, 4- mediated coupling with morpholine. The Suzuki coupling involves
disubstituted quinazolin-2-ones based on the earlier reports and the reaction of bromo derivative with boronic acid in the catalyst
screened them for its antagonistic activity against the T-type cal- medium containing PCy3 and Pd2dba3 using potassium phosphate
cium channel. The amino benzophenone 84 was cyclized in the as a base. The amino benzophenone 92 containing fluorine in the
presence of CDI in DCM followed by subsequent treatment with 5th and 6th position follows the alternative way to form the qui-
various primary amines. The cyclized product 85 was then heated nazolinone nucleus. The quinazolinone obtained from the grignard
with toluene in Dean-Stark apparatus to obtain the dehydrated reaction 93 and allyl magnesium bromide 94 was subjected to
product 86. The treatment of either the intermediates with ozonolysis yields the aldehyde 95. The dimethyl sulfide acts as a
Grignard reagent in THF resulted in the formation of dihy- reducing agent in the ozonolysis reaction. The aldehydes are
droquinazolinones 87 (Scheme 13). The compounds were tested for directly converted into geminal difluorides 96 with DAST. The alkyl
their potency against two types of FLIPR assays to identify their fluorides 97 are obtained after the reduction of the aldehyde into
state dependent inhibition. Type 1 is based on measuring the po- alcohols and then treated with DAST. The selection of various amino
tency in the inactivated state of the channel (depolarized assay) and ketones as the starting material leads to the different substituents
type 2 is based on the resting state of the channel (hyperpolarized in the aromatic ring of the quinazolinone (Scheme 14). The racemic
assay). All the compounds are selective towards the T-type calcium mixtures obtained through the synthetic routes were resolved into
channel and the reading in the EEG displayed a reduction in the enantiomers using chiral HPLC. All these trifluoroethyl containing
seizure time. The compound with the trifluoroethyl group in the derivatives were subjected to preliminary evaluation of calcium
3rd position and fluorine in the 4th position of the phenyl ring channel antagonistic activity in the depolarized FLIPR assay. The
displayed weak time dependent inhibition in the metabolic assay. compounds ability to activate the PXR was also evaluated. The
Also, the compound acts as a weak substrate for the PXR which is metabolic stability of the compounds was found out by incubating
responsible for the induction of metabolic enzyme (CYP3A4). In the compounds with human liver microsomes. The potent com-
short, the compounds 88 and 89 are effective against harmaline pounds were studied for their pharmacokinetic profiling in the rat,
induced tremor, active wake suppression, and epileptic seizures dog, and monkey. The compound which is potent in the FLIPR assay,
through the T-type calcium channel blockade. metabolically stable and has reduced PXR activation was subjected
Schlegel et al. [75] extended the work on 4, 4-disubstituted to further studies. The compound resulted in 70% inhibition against
quinazolin-2-ones as T-type calcium channel antagonists and seizure, decreased the active wake and REM sleep. Based on these
increased the number of derivatives by varying the substituents on findings, the compound with T-type calcium channel antagonist
610 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 13. Synthesis of 4, 4-disubstituted quinazolin-2-ones-I.

activity could be a valuable agent for the treatment of CNS improved the potency in the PBMC proliferation assay. Many of the
disorders. spiro-centered compounds generated chiral centre and the activity
was reported in one of the enantiomers. The SAR of spiro-
2.12. IMPDH inhibitor pyrrolidine containing compounds were studied by replacing the
t-butyloxy group 103 with different electrophiles. The oxy-urea
IMPDH is an enzyme which catalyzes the conversion of IMP to substituted spiro-pyrrolidine 104 displayed reduction in the he-
XMP. It is an example of oxidation reaction using NADþ as a cofactor patic clearance (CLint) with promising enzyme and cellular activ-
in the biosynthesis of guanine nucleotides (GMP, GDP, and GTP). ities. Another set of experimentation was performed to rectify the
There are two isoforms, IMPDH I and IMPDH II. The upregulation of increase in hepatic clearance rate. From the in vivo pharmacokinetic
IMPDH II is expressed in highly proliferating cells. This acts as a key study, the major site of metabolism was identified in the pyrroli-
mechanism for the IMPDH inhibitors (Fig. 13). Therefore, the inhi- dine core. The pyrrolidine moiety was replaced by proline core 105
bition of IMPDH has been exploited in the therapeutic intervention to create a metabolically stable structure. A pair of diastereomers
of anticancer, antiviral, antiparasitic and immunosuppressive was produced and they were purified by HPLC. The compound 105
therapy [76,77]. From the earlier reports [78], 7-methoxy-6-oxazol- having S stereochemistry at the spiro-centre reported less CLint
5-yl]-1H-quinazoline-2, 4-diones 100 were identified as IMPDH II value (18 mL/min/mg) with significant results aganist IMPDH II in-
inhibitors with poor physical properties. A slight modification of hibition and PBMC proliferation assay.
the structure resulted in a series of 7-methoxy-6-oxazol-5-yl-2, 3-
dihydro-1H-quinazolin-4-ones 99 with potent IMPDH II inhibition. 2.13. Sodium/calcium exchange inhibitor
The compounds were prepared by EDC coupling of amino-acid 98
with appropriate amines (Scheme 15). These derivatives were NCX is a membrane protein responsible for the maintenance of
designed with the objective of improving its potency along with the calcium ions (Ca2þ) in the cardiac myocytes [79]. The role of Ca2þ is
favorable DMPK properties. The replacement of 2-oxo moiety by to control the contraction in myocytes. Any impairment in the ex-
gem-dimethyl group 101 resulted in a moderate decrease in activity change of Naþ and Ca2þ can affect the function of cardiac myocytes.
with improved solubility. The substitution of one of the methyl The increase in Ca2þ overload causes an increase in the force of
group of the gem-dimethyl series by styrene moiety 102 improved contraction of the heart thereby it leads to myofibril hyper
the potency along with solubility. A range of spiro analogues was contracture. The NCX inhibitor regulates the improper functioning
synthesized which showed comparable activity to that of dione and of NCX and provides a valuable treatment in the case of reperfusion
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 611

Scheme 14. Synthesis of 4, 4-disubstituted quinazolin-2-ones-II. (i) AreB(OH)2, R ¼ Ar/P(Cy)3/Pd2dba3/K3PO4/Dioxane; (ii) Morpholine, R ¼ Morpholine/CuI, proline/K2CO3/DMSO;
(iii) Triphosgene/Et3N/CF3CH2NH2/ether/0  C; (iv) Et3N/THF; (v) SOCl2/THF; (vi) RMgBr/THF; (vii) DAST/0  C/CH2Cl2; (viii) NaBH4/MeOH/0  C.

Fig. 13. Mechanism of action of IMPDH inhibitor.


612 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 15. Synthesis of 7-methoxy-6-oxazol-5-yl-2, 3-dihydro-1H-quinazolin-4-ones. (i) EDC/CH2Cl2/R1-NH2; (ii)PTSA/ClCH2CH2Cl/ketone.

injury [80]. Hasegawa et al. found that the derivatives of 3, 4- quinazolinone 112 can be methylated 113 with iodomethane in the
dihydro-2(1H)-quinazolinone exhibited significant activity against presence of NaH. NaH is a strong base and it acts as a deprotonating
the NCX inhibition (Fig. 14). agent for the NeH bond. The carbonyl group of quinazolinone 112
The SAR study was carried out to evaluate the effect of sub- can be converted into thiocarbonyl group 114 by treating with
stituents on the various positions of 3, 4-dihydro-2(1H)-quinazo- phosphorous pentasulfide in xylene. The introduction of piperidine
linone nucleus. The reaction of 2-amino benzophenones 106 with ring is essential for the further derivatization of the dihy-
trichloroacetyl chloride resulted in the formation of tri- droquinazolinones. The cyclized product 116 was obtained by
chloroacetamide 107 which on subsequent reaction with primary refluxing the aniline derivative 115 with CDI. Removal of benzyl
amines gave cyclized products 108. Further treatment with NaBH4 group 117 was effected through the treatment with ammonium
resulted in the loss of trichloromethyl group and formed 3, 4- formate in the presence of palladium on carbon. The reductive
dihydro-2(1H)-quinazolinone 109. Debenzylation 111 of the com- amination 118 was favored when the aldehyde and sodium cya-
pound 110 can be achieved by the reaction with ammonium noborohydride reacted with an amine. Alternatively, the amine can
formate in the presence of Pd/C in methanol. The free NH group of be N-alkylated 118 using alkyl halide and potassium carbonate

Fig. 14. Mechanism of action of sodium/calcium exchange inhibitor.


K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 613

Scheme 16. Synthesis of dihydroquinazolinone as sodium/calcium exchange inhibitor. (i) CDI, THF; (ii) HCO2NH4, Pd/C, MeOH; (iii) NaBH3CN, HCl, MeOH/R-X, K2CO3, DMF.

(Scheme 16). The compounds were evaluated for their fura 2 should be preferred. The inclusion of piperidine ring with benzyl
fluorescence ratio, an index for measuring the calcium ion con- group in the 3rd position and 3-methoxy substituent 120 in the
centration in an isolated left atria from guinea pigs. The inhibition phenyl ring was found to be essential for the inhibition [81,82]. The
against Naþ and Kþ free contracture was calculated as IC30 value. same author extended the work by synthesizing the citrate form of
The removal of chlorine atom from the 6th position does not compound 112 and 116 using citric acid in methanol medium [83].
change the activity. The introduction of a methyl group and the
conversion of carbonyl to thiocarbonyl diminished the activity. The 2.14. PDE7 inhibitor
presence of phenyl ring 119 in the 4th position was found to be
optimum for retaining the activity. The side chain attached to the Phosphodiesterases are a class of enzymes that plays the catalytic
3rd position should have tertiary nitrogen atom. The optimum of role in the degradation of cAMP/cGMP to AMP/GMP. PDE7 is one
three carbon atoms between the ring and side chain nitrogen among the family and it selectively catalyzes the conversion of cAMP
614 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Fig. 15. Mechanism of action of PDE7 inhibitor.

Scheme 17. Synthesis of dihydroquinazolinones as PDE7 inhibitor-I. (i) NIS, TFA, H2SO4; (ii) Pd(PPh3)4/2-pyridylZnBr/THF/RB(OH)2/K2CO3/DMF; (iii) SOCl2/toluene; (iv) R3R4NH/
toluene.
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 615

Scheme 18. Synthesis of dihydroquinazolinones as PDE7 inhibitor-II.

to AMP [84]. Therefore, inhibition of PDE7 leads to increase in the level various alkyl halides (Scheme 18). The functionalization of 5-
of cAMP which affects various signal transduction pathways. The hydroxy-8-chloro-spirocyclohexane-quinazolinone 137 was sum-
expression of PDE7 is highly upregulated in T-cells and B-lympho- marized in Scheme 19. The carboxylic acid 138 was introduced
cytes. The inhibitors of PDE7 have been considered as a valuable through the treatment of phenol 137 with ethyl bromoacetate
therapeutics in the treatment of T-cell related diseases, autoimmune followed by the hydrolysis of the ester with KOH. The nitrile 141
diseases, CNS disorders and airway disease (Fig. 15) [85]. was synthesized using bromoacetonitrile and it reacted with azi-
The synthesis of dihydroquinazolinones was initiated by the dotrimethyltin (IV) to form tetrazole 139. The nitrile 141 was also
reaction between ureas and different ketones in the presence of used in the synthesis of hydroxy-oxadiazole 140 through oximes
PPA. The mono substituted (2/4- substitution) ureas 124 resulted in 142 as an intermediate. The introduction of alcohol in the side chain
the formation of single type cyclized product 125. The mixture of 143 was obtained through the reaction with 2-(2-bromoethoxy)
isomers 122, 123 (5, 6 and 6, 7-dichloro substituted quinazolinones) tetrahydro-2H-pyran followed by hydrolysis with HCl. The mesy-
was obtained from 3-chloro substituted or 3, 4-dichloro substituted late group 144 can be replaced either with morpholine 145 or ethyl
phenyl ureas 121. The chlorination reaction using NCS converted 6- glycinate 146 to form their corresponding derivatives.
substituted derivatives 125 into 6, 8-disubstituted derivatives 126.
The 8-chloro intermediates 127 were subjected to iodination re- 2.15. p38 MAPK inhibitor
action to afford 6-chloro-8-iodo intermediates 128. The iodide
atom present in the intermediate was replaced with phenyl ring The p38 MAPK pathway involves the sequential activation of
using Negishi/Suzuki coupling 129 reactions. The iodide atom three protein kinases: MAPK, MAPKK, and MAPKKK. p38 MAPK is
reacted with 2-pyridyl zinc bromide and phenyl boronic acid in the responsible for regulating the biosynthesis of pro-inflammatory
negishi and Suzuki coupling reaction respectively. The carboxylic cytokines, namely IL-1 and TNFa [88]. The interruption of this
group present in the intermediate was transformed into acid pathway through p38 inhibitors was responsible for the treatment
chloride using thionyl chloride. The amide derivatives 130 were of rheumatoid arthritis (Fig. 16) [89]. Stelmach et al. [90] reported
obtained by the reaction between an acid chloride and respective p38a MAP kinase inhibitory activity and pharmacokinetic profile of
amines (Scheme 17). The compounds were screened for their dihydroquinazolinones and they were designed on the basis of the
effectiveness against PDE7A subtype inhibitory activity. The reported structures. VX-745, 147 was reported as the phase II
unsubstituted 4th position spirocyclohexyl ring, 6th position clinical trial undergoing drug for rheumatoid arthritis and they are
phenyl ring, and dihalogen substitution were the features essential selective for p38a MAP kinase. The compound 148 is the reduced
for the potent action. The presence of carboxyl group increased the isomeric analog of VX-745 with polar amine substituents in the C-7
activity but the lack of selectivity towards the enzyme subtypes. position for increasing the potency and physicochemical properties.
Whereas the introduction of amide group with the linker con- The thioether group in C-6 position can be replaced with C-5
taining 2e3 carbon atom displayed effective inhibition score and phenyl ring for occupying the p38a hydrophobic pocket. The de-
good selectivity against PDE7A enzyme. The amide containing rivatives of dihydroquinazolinones containing C-5 phenyl ring 149
piperazine derivatives displayed similar potency but decreased were prepared as per Scheme 25. The Ullmann type coupling re-
solubility [86]. action of 150 afforded cyclized product 151. The deprotection of
Bernardelli et al. [87] extended the work on spiroquinazolinones PMB group was effected with TFA. The C-5 bromine was subjected
by screening the potency and selectivity of various 5, 8- to Suzuki coupling 152 with aryl boronic acids in the presence of
disubstituted derivatives towards the PDE enzyme. The Pd(PPh3)4. As a demethylating agent, boron tribromide converted
substituted anilines 131 reacted with potassium cyanate and C-7 methoxy group 153 into phenolic hydroxyl group 154. The
transformed the amine derivatives into urea derivatives 132. The conversion of phenolic OH into its triflates 155 was carried out by
urea derivatives were cyclized into spiroquinazolinones 133 by phenyl triflimide. The triflates reacted with vinyl trimethyltin in the
reacting with cyclohexanone using PPA. The phenol containing presence of Pd(PPh3)4 and lithium chloride resulted in Stille type
quinazolinone 135 was obtained from the methoxy derivative 134 coupling product 156. The presence of lithium salt is to increase the
by using boron tribromide as a demethylating agent. The O-alkyl- polarity of the solvent for the easy removal of triflates. The plat-
ation 136 of phenol intermediate was carried out in the presence of inum dioxide also known as Adam's catalyst was used for the
616 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 19. Synthesis of dihydroquinazolinones as PDE7 inhibitor-III. (i) Ethyl bromoacetate/K2CO3/DMF; (ii) KOH/THF; (iii) Bromoacetonitrile/K2CO3/DMF; (iv) NH2OH$HCl/NaOH,
EtOH; (v)ClCO2Et/CHCl3/Et3N; (vi)DBU/ACN; (vii) Me3SnN3/Toluene; (viii)2-(2-bromoethoxy)tetrahydro-2H-pyran/K2CO3/DMF; (ix) HCl/THF/H2O; (x) Methanesulfonyl chloride/
Et3N/CH2Cl2/0  C; (xi) Morpholine/EtOH; (xii) Ethyl glycinate/CH3CN; (xiii) 6 N HCl/1, 4-dioxane.

hydrogenation of vinyl group and N-boc can be deprotected using dibromo-2-methyl-5-nitrobenzene 166. The reaction up to the
TFA (Scheme 20). The triflates 157 were also subjected to synthesis of the intermediate 152 was followed as per the report of
Buchwald-Hartwig coupling reaction 158 in the presence of the previous author except deprotection of PMB group 167. The
amines. The Mitsunobu reaction was carried out to convert the amine group 168 was introduced by means of reduction with
phenolic OH 159 into ether 160 using DEAD and triphenylphos- hydrogen in the presence of palladium on carbon. The reductive
phine. The dihydroquinazolinones containing methylene linker 162 alkylation of the amine with ketone was carried out by using the
and carbonyl linker 163 at C-7 were obtained either by reductive mild reducing agent, sodium triacetoxyborohydride. Finally, the
amination and CDI coupling of ester 161 respectively (Scheme 21). PMB group was deprotected by TFA to yield the dihy-
The compounds containing piperazine/piperidine at C-7 position droquinazolinones 169 (Scheme 22). The three series of C7-
exhibited significant inhibition against p38a enzyme assay. They piperidine and 4-aminopiperidine substituted naphthyridinones
also inhibited the TNFa production. The rat pharmacokinetic profile 164, quinolinones 165 and dihydroquinazolinones 169 were
displayed that these compounds resulted in low oral bioavailability screened for their inhibition against p38a enzyme assay and TNFa
and rapid clearance. The compound containing t-butyl group in the production. The naphthyridinone and quinolinone derivatives were
C-7 piperidine ring increased the oral bioavailability and decreased more potent and exhibited improved pharmacokinetic profile in
the clearance rate. the rat. The comparatively less potency of the dihy-
Hunt et al. [91] investigated the p38 inhibitory activity of droquinazolinone derivatives was due to the difference in the C-4
dihydroquinazolinones in addition to naphthyridinones 164 and position. The C-4 position was stabilized because of the sp2 hy-
quinolinones 165. The reaction scheme was initiated with 1, 3- bridized centre, the clearance rate was lowered and oral
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 617

Fig. 16. Mechanism of action of p38 inhibitor.

bioavailability was increased. This is the reason for the enhanced 2.16. CDK5 inhibitor
pharmacokinetic profile of the naphthyridinone derivatives.
Schlapbach et al. [92] synthesized the dihydroquinazolinone CDK5 is a proline-directed serine/threonine kinase and its acti-
derivatives from diclofenac and tested its inhibition score against vation requires various cyclin (cyclin I, E, and G1) and non-cyclin
the p38a enzyme. The a-arylamino phenylacetic acid 170 was activators (p35, p39, and p67). This protein plays a vital role in the
subjected to curtius rearrangement in the presence of DPPA. The normal development of CNS and they have been involved in con-
intramolecular ring closure of the isocyanate intermediate led to trolling several neuronal processes. The role of CDK5 has also been
the formation of 1-(2, 6-dichlorophenyl)-3, 4-dihydroquinazolin- implicated in several non-neuronal functions (Fig. 17) [93]. The
2(1H)-one 171. The dihydroquinazolinone are subjected to C-6 deregulation of CDK5 has been corroborated in several neurode-
functionalization by various reactions. The bromination and further generative diseases such as Alzheimer's disease, Huntington's
coupling at the C-6 position were preceded via Buchwald-Hartwig chorea, stroke, Parkinson's disease and Lou Gehrig's disease [94].
reaction 172. The 2, 4-difluoro-thiophenol reacted with the Rzasa et al. [95] designed a series of 3, 4-dihydroquinazolin-2(1H)-
bromine intermediate in the presence of tetrakis palladium and ones as CDK5 inhibitors. The molecule 183 was designed on the basis
sodium tert-butoxide. The C-6 position was nitrated using potas- of intramolecular hydrogen bond formation between the two ni-
sium nitrate and reduced in the presence of hydrogen and palla- trogen atoms in the pyridyl urea 182. It was assumed that the
dium on carbon for introducing the amine group 173. The 6-amino- intramolecular hydrogen bond between N1 and hydrogen atom
1-(2, 6-dichlorophenyl)-3,4-dihydroquinazolin-2(1H)-one was uti- from N3 occupies the active site of CDK5 enzyme. This assumption
lized in the synthesis of arylamines, benzylamines, carboxamides was confirmed from the cocrystallographic data of pyridyl urea 182
or sulfonamides. The Buchwald-Hartwig coupling of the amine and CDK2, an analog of CDK5. Various modifications have been
with 2, 4-difluoro-bromobenzene was carried out using Pd2dba3 made in the A, B, C and D ring systems of the structure 183. From the
and DPEphos afforded arylamine 174. The formation of benzyl- SAR study, it was found that the compounds containing dihy-
amines 175 through the reductive amination of amine and benz- droquinazolinones ring system (B ring: U]CH2) were more potent
aldehyde was carried out with the aid of sodium cyanoborohydride. than the quinazolinedione and benzimidazolone ring. It is because
The reaction of amine with benzoyl chloride and the of the van der waals contact with the amino acid residues found
benzenesulfonyl-chloride resulted in the formation of carboxamide within the active site of the enzyme. The derivatives with the range
176 and sulfonamide 177 respectively (Scheme 23). The phenyl ring of substituents were tolerated in the ring A, thiazolo moiety in the
at the N-1 position of dihydroquinazolinones replaced with various place of C ring and 4- pyridyl moiety in the ring D showed significant
substituents as a p38a inhibitor was synthesized. The urea de- activity against HTRF human CDK5/p25 assay. The importance of 4-
rivatives 178 were cyclized using palladium complex Pd2dba3 to pyridyl ring was supported by the formation of hydrogen bonding to
afford N-1 substituted dihydroquinazolinones 179. The O-alkylated the Asp145-Lys33 salt bridge.
180 and N-alkylated 181 derivatives of 1-(4-hydroxy-2, 6- The anilines 184, 186, 189, 191 and 194 obtained through the
dimethylphenyl)-6-nitro-3, 4-dihydroquinazolin-2(1H)-one were various methodologies were cyclized using p-nitrophenyl chlor-
obtained from chloropropyl-morpholine and 3-chloro-4-fluoro oformate or CDI mediated reaction to give dihydroquinazolinones
benzenesulfonylchloride respectively (Scheme 24). Because of the 185, 187, 190, 192 and 195. The ester containing dihy-
selectivity towards the p38 MAP kinase, significant inhibition of droquinazolinones 187 was hydrolysed and decarboxylated in the
TNFa release and non-interference with liver enzymes, the com- presence of alkaline and acidic medium to give the final product
pound was identified as a valuable candidate for drug discovery. 188 (Scheme 25).
618 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 20. Synthesis of dihydroquinazolinones as p38 inhibitor-I.

2.17. RyR agonist chloropyridin-2-yl)-1H-pyrazole-5-carboxylate 199 was reduced


into alcohol 200 in the presence of one equivalent of LiAlH4.
Ryanodine is a natural insecticide and the receptor through Dechlorination of pyridine ring 201 was obtained from the treat-
which it acts is called RyR. RyR is an example of non-voltage-gated ment of more than two equivalent of reducing agent. The primary
calcium channel and it causes the regulation of calcium release alcohol 200 & 201 was oxidized using PCC into aldehyde 202. The
from intracellular storage [96]. Ryanodine type agonist causes the amide and the aldehyde were refluxed in the presence of PTSA and
continuous release of calcium from the sarcoplasmic reticulum. toluene medium to afford the dihydroquinazolinones 203. When
Thereby the concentration of calcium in the myoplasm of insect the same reaction was carried out in the presence of ethanol, qui-
increases and the muscles remain in the state of continuous nazolinones 204 were obtained (Scheme 26). The compound 205
contraction. The massive muscular contraction causes paralysis of exhibited 100% mortality against the larvae of Spodoptera exigua.
muscles leading to the death of the insects (Fig. 18) [97]. Zhou et al. The results of calcium imaging techniques demonstrated that RyR
[98] carried out the synthesis of 2, 3-dihydroquinazolinones and may be the possible site of action for the compounds containing
tested their insecticidal activity against oriental armyworm dihydroquinazolinones.
(Mythimna separata). The 2-aminobenzoic acid 196 was used as a
starting material for the synthesis of various amides. The amides 2.18. Non-nucleoside reverse transcriptase inhibitor
197 & 198 were obtained by the reaction of the 2-aminobenzoic
acid with thionyl chloride/triphosgene/CDI followed by the treat- HIV is a member of retroviridae family and it specifically attacks
ment with corresponding amines. The ethyl 3-bromo-1-(3- the T-helper cells. The T-helper cells are the important component
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 619

Scheme 21. Synthesis of dihydroquinazolinones as p38 inhibitor-II. (i) NBS/Benzoyl peroxide/p-methoxy benzyl amine; (ii) 2, 6-di-Cl-PhNCO; (iii) CuI/K2CO3/DMF; (iv)TFA/(v)
ArB(OH)2/Pd(PPh3)4; (vi) N-boc-4-trimethylstannyl-5, 6-dihydropyridine/Pd(PPh3)4/LiCl, (vii) H2, PtO2; (viii) TFA.

Scheme 22. Synthesis of dihydroquinazolinones as p38 inhibitor-III.


620 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 23. Synthesis of dihydroquinazolinones as p38 inhibitor-IV. (i)Diphenylphosphoryl azide/NEt3/dimethoxyethane; (ii) Br2/CH2Cl2; (iii) 2, 4-difluoro-thio-phenol/Pd(PPh3)4/
NaOtBu/toluene; (iv) KNO3/H2SO4/0  C; (v) H2, Pd/C, ethanol; (vi) 2, 4-di-F-bromobenzene/Pd2dba3/DPEphos/NaOtBu/toluene; (vii) benzaldehyde/NaBH3CN/3% AcOH in DMF; (viii)
Benzoyl chloride/NEt3/THF; (ix) Benzenesulfonyl chloride/DMAP/pyridine.

Scheme 24. Synthesis of dihydroquinazolinones as p38 inhibitor-V. (i) Boc2O/DMAP/CH2Cl2; (ii) Pd2dba3/2-(dicyclohexyl-phosphino)biphenyl/K3PO4; (iii) Chloropropyl-
morpholine/CsI/DMF; (iv) H2/Pd/C/MeOH; (v) 3-chloro-4-fluoro benzenesulfonylchloride/pyridine/DMAP/CH2Cl2.

of WBC helps to coordinate the immune response. HIV enters the categorized into NRTIs and NNRTIs. The development of NNRTI
host cell through binding with host cell CD4 receptors found on the based drug candidate is advantageous because of its potency and
surface of the cell. After entering into the host cell, HIV utilizes the low cytotoxicity [100]. Corbett et al. [101] synthesized dihy-
host cell machinery for the replication process and leaves them as a droquinazolinones from amino ketones 206 as starting materials.
mature virion. Several proteins are involved in the replication The aminols 207 were obtained by the reaction of amino ketones
process and they can act as a valuable target for the treatment of 206 with TMSNCO and TBAF. The trifluoromethyl ketimines 208 are
HIV (Fig. 19) [99]. RT is an attractive target for HIV and they are the dehydrated product of aminols treated in the presence of
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 621

Fig. 17. The role of CDK5 inhibitors in neurodegenerative diseases.

toluene or xylene and the moisture is absorbed by 4 Å molecular and they are synthesized from the wide variety of starting mate-
sieves. Then the ketimines were alkylated with lithiated alkynes rials. Guan et al. [105] developed an efficient protocol for the syn-
using the catalytic amount of lewis acid like boron trifluoride thesis of dihydroquinazolinones from anthranilamide obtained via
etherate to afford quinazolinones 209 (Scheme 27). The analogs indazoles. Indazoles 221 are either prepared by Buchwald-Hartwig
were screened for their in vitro inhibition of HIV-1 RT assay. The 5, reaction (i) or by copper-catalyzed aryl coupling reaction (ii). The
6-dihalogen compounds and the presence of trifluoromethyl group indazole is converted into indazolium salts 222 by methylating
improved the overall resistance of the molecule. The compounds with dimethylsulphate. The indazolium salts are reacted with po-
DPC 961 210, DPC 963 211, DPC 082 212, and DPC 083 213 exhibited tassium tert-butoxide in the presence of toluene and water gives
pharmacokinetic property similar to that of efavirenz. These com- the corresponding anthranilamides 223. The anthranilamides on
pounds also provided superior antiviral potency and safety profile treatment with 50% sulphuric acid and formaldehyde yielded 2, 3-
against the mutant HIV variants (see Scheme 28). dihydro-4(1H)-quinazolinones 224 (Scheme 29).

2.19. Aldosterone synthase inhibitor 3.2. Copper oxide nanoparticle mediated synthesis of
dihydroquinazolinones
Aldosterone synthase is an important enzyme involved in the
biosynthesis of aldosterone. Aldosterone is a mineralocorticoid Zhang et al. [106] carried out the one pot condensation of isatoic
hormone essential for the homeostasis of blood volume and elec- anhydride 20, anilines 225 and aldehydes in the presence of various
trolyte balance. The excessive secretion of aldosterone is respon- catalysts to afford dihydroquinazolinones 226 & 227 (Scheme 30).
sible for the pathological conditions such as hypertension, cardiac CuO nanoparticles were found to be superior among the catalyst.
fibrosis and congestive heart failure (Fig. 20) [102,103]. Grombien The experimentation on the effect of ethanol:water ratio on the
et al. [104] carried out the synthesis of moieties designed on the yield of the product was tested. The increase in the amount of water
basis of 3, 4-dihydroquinolinone nucleus in which the methylene substantially increased the yield. The yield was optimised at 1:1
unit of 3, 4-dihydropyridinone were bioisosterically exchanged ratio of ethanol: water and a further increase in the water
with various heteroatoms 214.2-(aminomethyl)aniline 216 was decreased the yield. The 10 mol% of CuO nanoparticles was
cyclized into 3, 4-dihydroquinazolinone 217 in the presence of necessary to increase the yield. When the reaction was stirred at
triphosgene. The bromine atom was introduced in to 3, 4- reflux temperature, it required 3 h for completion. In the presence
dihydroquinazolinone using NBS 218 and it was subjected to of ultrasound irradiation (40 KHz and 250 W), the reaction was
Suzuki Miyaura coupling 219 using heteroarylboronic acid. The completed in 10e30 min. The condensation reaction between isa-
compound 220 showed minor selectivity towards the human toic anhydride, ammonium chloride 228 and various aldehydes in
CYP11B1 gene responsible for encoding of the aldosterone synthase the presence of CuO nanoparticles, quinazolinones 229 were ob-
enzyme. The replacement of lactame 214 by sultame 215 moiety tained as a product. When the ethanol: water system was main-
displayed significant reduction in the biological activity. tained at 3:1 ratio, there is a maximum conversion of
quinazolinone. The reason for the unexpected formation of quina-
3. Miscellaneous synthesis and applications of zolinones may be due to the intramolecular electron transfer and
dihydroquinazolinones rearrangement in the presence of inorganic salts.

3.1. Synthesis of 2, 3-dihydroquinazolin-4(1H)-ones from N- 3.3. Y(OTf)3 mediated synthesis of dihydroquinazolinones


heterocyclic carbenes of indazole
The synthesis of dihydroquinazolinones was carried out by the
Indazoles are medicinally and pharmaceutically important motif condensation reaction between anthranilamide 230 and various
622 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 25. Synthesis of dihydroquinazolinones as CDK5 inhibitor. (i) p-nitophenyl chloroformate/Et3N/toluene/THF; (ii) CDI/NaH/DMF; (iii) NaOH/MeOH/H2SO4; (iv) iron dust/
NH4Cl/aq. EtOH.

aldehydes 231. In the absence of a catalyst, traces of product were quinazolinones 233 along with dihydroquinazolinones 232
obtained even after long time treatment. The comparison of acidic (Scheme 31). The role of oxidants identified dimethyl sulfoxide in
catalyst resulted in improved yields. The uniqueness of the rare providing a satisfactory yield of quinazolinones [107].
earth metal chloride as catalyst displayed effectiveness in yield.
However, Y(OTf)3 was found to be superior among the rare earth 3.4. Boric acid mediated synthesis of dihydroquinazolinones
metals. Ethanol was optimized as the suitable solvent for the re-
action. These optimized reaction conditions yielded traces of Anthranilamide 234 was converted into 3, 5-
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 623

Fig. 18. The role of the ryanodine receptor in insecticidal activity.

Scheme 26. Synthesis of dihydroquinazolinones as an insecticidal agent. (i) a) SOCl2, b) R2NH2/THF; (ii) a) triphosgene, b) CH3NH2/H2O; (iii) CDI/THF; (iv) NH3/H2O; (v) LiAlH4/THF;
(vi)PCC/CH2Cl2; (vii) PTSA/toluene/reflux; (viii) CH3CH2OH/reflux.

dibromoanthranilamide 235 using N-bromosuccinimide in 3.5. Visible light mediated synthesis of dihydroquinazolinones
chloroform-carbon tetrachloride mixture. It was then cyclized with
benzaldehyde derivatives using boric acid as a catalyst in the Hemalatha et al. reported the visible light mediated synthesis of
presence of solvent-free condition to obtain 2-aryl-6,8-dibromo- 2, 3- dihydroquinazolinones from isatoic anhydride and ketones in
2,3-dihydroquinazolin-4(1H)-ones 236. Further derivativatization the presence and absence of acetic acid. The reaction was efficient
of dihydroquinazolin-4(1H)-ones was carried out by subjecting the in the presence of acetic acid. The solvatochromic effect of the
bromine to Suzuki Miyaura coupling reaction. The coupling reac- synthesized series was carried out using various solvents of
tion between 2-aryl-6,8-dibromo-2,3-dihydroquinazolin-4(1H)- increasing polarity. All the compounds exhibited significant quan-
ones and phenylboronic acid in the presence of PdCl2(PPh3)2-Xphos tum yield and they yielded maximum fluorescence in dimethyl
catalyst complex yields biarylated 2, 3-dihydroquinazolin-4(1H)- sulfoxide. Further the antioxidant activity and in vitro anti-
one derivatives 237. The 2, 3-dihydroquinazolin-4(1H)-one de- inflammatory activity of dihydroquinazolinones concluded those
rivatives were oxidized into quinazolin-4(3H)-one 238 derivatives isatin containing derivatives 240 are most effective among the
in the presence of molecular iodine as an oxidant. The formation of synthesized series (Fig. 21) [109]. The biological activity of the
lactam form of quinazolin-4(3H)-one derivative 239 was confirmed compounds was correlated with the docking studies carried out
through the NMR spectra (Scheme 32) [108]. using PARP [110] enzyme.
624 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Fig. 19. Mechanism of action of anti-HIV drugs.

Scheme 27. Synthesis of trifluoromethyl-containing dihydroquinazolinones.

Scheme 28. Synthesis of 3, 4-dihydroquinazolinones as aldosterone synthase inhibitor. (i) Triphosgene/THF; (ii) NBS/DMF/0  C; (iii) Heteroarylboronic acid/Pd(PPh3)4/aq. Na2CO3/
DME/reflux.
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 625

Fig. 20. Mechanism of action of aldosterone synthase inhibitor.

Scheme 29. Synthesis of dihydroquinazolinones from indazoles.

3.6. Importance of 7-fluoro-2, 2-dimethyl-2, 3-dihydroquinazolin- cyclization with acetone in the presence of concentrated HCl to
4(1H)one afford the fluorinated derivative of 2, 3-dihydroquinazolinone 244
(Scheme 33). The interaction between the fluorinated derivative
Due to the importance of fluorinated compounds in the field of and lysozyme was predicted using NMR study in addition to the
drug discovery and development, the fluorine containing 2, 2- various spectrophotometric studies. The difference in the chemical
dimethyl-2, 3-dihydroquinazolin-4(1H)one was synthesized. The shift value of the ligand after interaction with the protein
amide 243 for the synthesis of the fluorinated derivative was pre- confirmed the interaction between them. In silico prediction of the
pared from the condensation reaction between 2-amino-4- metabolic pathway of the fluorinated derivative confirmed that the
fluorobenzoic acid 241 and methylamine 242 in the presence of presence of fluorine atom prevents the oxidation of aromatic ring
EDC and hydroxybenzotriazole. The amide 243 undergoes and makes the molecule metabolically stable. From the MTT assay,
626 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

Scheme 30. CuO nanoparticle-mediated synthesis of dihydroquinazolinones.

Scheme 31. Y(OTf)3 mediated synthesis of dihydroquinazolinones.

Scheme 32. Boric acid mediated synthesis of dihydroquinazolinones.


K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 627

3.8. Suzuki Miyaura coupling of 2, 3-dihydroquinazolinones

Hemalatha et al. extended the work by subjecting the bromi-


nated derivatives of 2, 3-dihydroquinazolinones 248 to Suzuki
Miyaura coupling (Scheme 35). The Suzuki coupled products 249
were screened for antifungal activity against the Aspergillus species.
The interaction of dihydroquinazolinones containing CeO bond
Fig. 21. Isatin containing dihydroquinazolinone derivative. with the amino acid residues of the protein N-myristoyl transferase

Scheme 33. Synthesis of 7-fluoro-2, 2-dimethyl-2, 3-dihydroquinazolin-4(1H)one. (i) EDC. HCl/HOBt/Et3N/DCM; (ii) Acetone/HCl.

Scheme 34. Bromination of 2, 3-dihydroquinazolinones.

was identified to be the reason for the superior antifungal activity


of the compounds. The active compound among the synthesized
series was subjected to lysozyme interaction studies. The interac-
tion between the compound and the lysozyme was justified
through various spectroscopic studies [113].
In another work, optimization of the same reaction using
palladium nanoparticles was carried out. The coupling reaction
carried out in the presence of both microwave and ultraviolet
irradiation was resulted in maximum yield. The toxicity of the
palladium nanoparticles was tested using Artemia salina bioassay
and it was found to be non toxic. Environmental impact of the re-
action conditions was assessed from the E-factor and eco scale
value. The analysis confirmed the ecofriendliness of the reaction
condition [114].
Scheme 35. Synthesis of 6-substituted 2, 3-dihydroquinazolinones via Suzuki Miyaura
coupling. 4. Conclusion

The summation of the synthetic methodology will drive the


the concentration greater than 80 mM was identified as the cyto- interest of the chemist towards the dihydroquinazolinone nucleus.
toxic concentration [111]. The compounds with the same reactivity can also be experimented
using the same reaction condition. The schematic representation of
3.7. Bromination of 2, 3-dihydroquinazolinones the biochemical pathway will be clear cut for even the non-
biologist to understand the mechanism easily. The designing of
Hemalatha and co-worker carried out the bromination of 2, 3- the compounds based on the effects of the substituents will result
dihydroquinazolinones 245 using NBS as the brominating agent. in the development of an effective molecule for the future scenario.
In the presence of 1 equivalent of NBS, bromine was substituted in
the 6th position of 2, 3-dihydroquinazolinones 246. However in the References
presence of 1.1 equivalent of NBS, there may be a chance for the
[1] K. Hemalatha, G. Madhumitha, S.M. Roopan, Indole as a core anti-
substitution of bromine atom in the 5th position of 2, 3-
inflammatory agent- a mini review, Che. Sci. Rev. Lett. 2 (2013) 287e292.
dihydroquinazolinones 247 I n addition to 6th position bromine [2] Y. Chen, C. Tang, Y. Wu, S. Mo, S. Wang, G. Yang, Z. Mei, Glycosmisines A and
atom (Scheme 34). The introduction of hydrophobic bromine atom B: isolation of two new carbazole-indole-type dimeric alkaloids from Gly-
was justified for the significant anthelmintic activity of the com- cosmis pentaphylla and an evaluation of their antiproliferative activities, Org.
Biomol. Chem. 13 (2015) 6773e6781.
pounds. The interaction study with bovine serum albumin was [3] A.J. Singh, J.D. Dattelbaum, J.J. Field, Z. Smart, E.F. Woolly, J.M. Barber,
determined from the spectroscopic studies [112]. R. Heathcott, J.H. Miller, P.T. Northcote, Structurally diverse hamigerans from
628 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

the New Zealand marine spongeHamigera tarangaensis: NMR-directed 193e244.


isolation, structure elucidation and antifungal activity, Org. Biomol. Chem. [29] S.B. Mhaske, N.P. Argade, The chemistry of recently isolated naturally
11 (2013) 8041e8051. occurring quinazolinone alkaloids, Tetrahedron 62 (2006) 9787e9826.
[4] J. Palaniraja, S.M. Roopan, UV-light induced domino type reactions: synthesis [30] U.A. Kshirsagar, Recent developments in the chemistry of quinazolinone
and photophysical properties of unreported nitrogen ring junction quina- alkaloids, Org. Biomol. Chem. 13 (2015) 9336e9352.
zolines, RSC Adv. 5 (2015) 37415e37423. [31] V.B. Labade, P.V. Shinde, M.S. Shingare, A facile and rapid access towards the
[5] K. Hemalatha, G. Madhumitha, A. Kajbafvala, N. Anupama, S. Rajesh, synthesis of 2, 3-dihydroquinazolin-4(1H)-ones, Tetrahedron Lett. 54 (2013)
S.M. Roopan, Function of nanocatalyst in chemistry of organic compounds 5778e5780.
revolution: an overview, J. Nanom. 2013 (2013) 1e23. [32] Z. Liu, L. Ou, M.A. Giulianotti, R.A. Houghten, Solid-phase synthesis of N-
[6] A. Bharathi, S.M. Roopan, A.A. Rahuman, G. Rajakumar, (E)-2-Benzylidene-7- substituted 3, 4-dihydroquinazolinone derivatives, Tetrahedron Lett. 52
chloro-9-phenyl-3, 4- dihydroacridin-1(2H)-ones: synthesis and larvicidal (2011) 2627e2628.
activity, Res. Chem. Intermed. 41 (2015) 2453e2464. [33] Y. Kobayashi, Y. Nakano, M. Kizaki, K. Hoshikuma, Y. Yokoo, T. Kamiya,
[7] A. Kalaiselvi, S.M. Roopan, G. Madhumitha, C. Ramalingam, G. Elango, Syn- Capsaicinlike antiobese activities of evodiamine from fruits of Evodia rutae-
thesis and characterization of palladium nanoparticles using Catharanthus carpa, a vanilloid receptor agonist, Planta Med. 67 (2001) 628e633.
roseus leaf extract and its application in the photo-catalytic degradation, [34] J. Rosenberg, F. Gustafsson, S. Galatius, P.R. Hildebrandt, Combination ther-
Spectrochim. Acta Mol. Biomol. Spectrosc. 135 (2015) 116e119. apy with metolazone and loop diuretics in outpatients with refractory heart
[8] S.M. Roopan, A. Bharathi, A. Prabhakarn, A.A. Rahuman, K. Velayutham, failure: an observational study and review of the literature, Cardiovasc. Drug.
G. Rajakumar, R.D. Padmaja, M. Lekshmi, G. Madhumitha, Efficient phyto- Ther. 19 (2005) 301e306.
synthesis and structural characterization of rutile TiO2 nanoparticles using [35] G.C. Roush, R. Kaur, M.E. Ernst, Diuretics: a review and update, J. Cardiovasc.
Annona squamosa peel extract, Spectrochim. Acta Mol. Biomol. Spectrosc. 98 Pharmacol. Ther. 19 (2014) 5e13.
(2012) 86e90. [36] J. Olesen, H.-C. Diener, I.W. Husstedt, P.J. Goadsby, D. Hall, U. Meier,
[9] C. Jayaseelan, A.A. Rahuman, S.M. Roopan, A.V. Kirthi, J. Venkatesan, S.- S. Pollentier, L.M. Lesko, Calcitonin gene-related peptide receptor antagonist
K. Kim, M. Iyappan, C. Siva, Biological approach to synthesize TiO2 nano- BIBN 4096 BS for the acute treatment of migraine, N. Engl. J. Med. 350 (2004)
particles using Aeromonas hydrophila and its antibacterial activity, Spec- 1104e1110.
trochim. Acta Mol. Biomol. Spectrosc. 107 (2013) 82e89. [37] R.W. King, R.M. Klabe, C.D. Reid, S.K. Erickson-Viitanen, Potency of non-
[10] G. Madhumitha, G. Rajakumar, S.M. Roopan, A.A. Rahuman, K.M. Priya, nucleoside reverse transcriptase inhibitors (NNRTIS) used in combination
A.M. Saral, F.R.N. Khan, V.G. Khanna, K. Velayutham, C. Jayaseelan, with other human immunodeficiency virus NNRTIS, NRTIS, or protease in-
C. Kamaraj, G. Elango, Acaricidal, insecticidal, and larvicidal efficacy of fruit hibitors, Antimicrob. Agents Ch 46 (2002) 1640e1646.
peel aqueous extract of Annona squamosa and its compounds against blood- [38] S. Li, J.-A. Ma, Core-structure-inspired asymmetric addition reactions:
feeding parasites, Parasitol. Res. 111 (2012) 2189e2199. enantioselective synthesis of dihydrobenzoxazinone and dihy-
[11] G. Madhumitha, A.M. Saral, Screening of larvicidal activity of Crossandra droquinazolinone based anti-HIV agents, Chem. Soc. Rev. 44 (2015)
infundibuliformis extracts against Anopheles stephensi, Aedes aegypti and 7439e7448.
Culex Quinquefasciatus, Int. J. Pharm. Pharm. Sci. 4 (2012) 485e488. [39] D.J. Foster, D.L. Choi, P.J. Conn, J.M. Rook, Activation of M1 and M4 muscarinic
[12] G. Madhumitha, A.M. Saral, Preliminary phytochemical analysis, antibacte- receptors as potential treatments for Alzheimer's disease and schizophrenia,
rial, antifungal and anticandidal activities of successive extracts of Cross- Neuropsychiatr. Dis. Treat. 10 (2014) 183e191.
andra infundibuliformis, Asian Pac. J. Trop. Med. 4 (2011) 192e195. [40] Y. Uruno, Y. Konishi, A. Suwa, K. Takai, K. Tojo, T. Nakako, M. Sakai,
[13] G. Madhumitha, A.M. Saral, B. Senthilkumar, A. Sivaraj, Hepatoprotective T. Enomoto, H. Matsuda, A. Kitamura, T. Sumiyoshi, Discovery of dihy-
potential of petroleum ether leaf extract of Crossandra infundibuliformis on droquinazolinone derivatives as potent, selective, and CNS-penetrant M1
CCl4 induced liver toxicity in albino mice, Asian Pac. J. Trop. Med. 3 (2010) agonists and M4 muscarinic acetylcholine receptors, Bioorg. Med. Chem. Lett.
788e790. 25 (2015) 5357e5361.
[14] G. Madhumitha, A.M. Saral, Free radical scavenging assay of Thevetia ner- [41] B.S. Rayner, D.T. Love, C.L. Hawkins, Comparative reactivity of
ufolia leaf extracts, Asian J. Chem. 21 (2009) 2468e2470. myeloperoxidase-derived oxidants with mammalian cells, Free Radic. Bio.
[15] K. Velayutham, A.A. Rahuman, G. Rajakumar, S.M. Roopan, G. Elango, Med. 71 (2014) 240e255.
C. Kamaraj, S. Marimuthu, T. Santhoshkumar, M. Iyappan, C. Siva, Larvicidal [42] E. Malle, T. Buch, H.-J. Grone, Myeloperoxidase in kidney disease, Kidney. Int.
activity of green synthesized silver nanoparticles using bark aqueous extract 64 (2003) 1956e1967.
of Ficus racemosa against Culex quinquefasciatus and Culex gelidus, Asian Pac. [43] Y. Li, T. Ganesh, A. Sun, B.A. Diebold, Y. Zhu, J.W. McCoy, S.M.E. Smith,
J. Trop. Med. 6 (2013) 95e101. J.D. Lambeth, Thioxo-dihydroquinazolin-one compounds as novel inhibitors
[16] S.M. Roopan, T.V. Surendra, G. Elango, S.H.S. Kumar, Biosynthetic trends and of myeloperoxidase, ACS Med. Chem. Lett. 6 (2015) 1047e1052.
future aspects of bimetallic nanoparticles and its medicinal applications, [44] Y. Ma, D. Ren, J. Zhang, J. Liu, J. Zhao, L. Wang, F. Zhang, Synthesis, antibac-
Appl. Microbiol. Biotechnol. 98 (2014) 5289e5300. terial activities evaluation, and docking studies of some 2-substituted-3-
[17] C.T. Walsh, Nature loves nitrogen heterocycles, Tetrahedron Lett. 56 (2015) (phenylamino)-dihydroquinazolin-4(1H)-ones, Tetrahedron Lett. 56 (2015)
3075e3081. 4076e4079.
[18] S.M. Roopan, F.R.N. Khan, ZnO nanoparticles in the synthesis of AB ring core [45] K. Janiyani, T. Bordelon, G.L. Waldrop, J.E. Cronan, Function of Escherichia coli
of camptothecin, Chem. Pap. 64 (2010) 812e817. biotin carboxylase requires catalytic activity of both subunits of the homo-
[19] S.M. Patil, S. Kulkarni, M. Mascarenhas, R. Sharma, S.M. Roopan, dimer, J. Biol. Chem. 276 (2001) 29864e29870.
A. Roychowdhury, DMSO-POCl3: a reagent for methylthiolation of imidazo[1, [46] M. Brylinski, G.L. Waldrop, Computational redesign of bacterial biotin
2-a]pyridines and other imidazo-fused heterocycles, Tetrahedron 69 (2013) carboxylase inhibitors using structure-based virtual screening of combina-
8255e8262. torial libraries, Molecules 19 (2014) 4021e4045.
[20] P. Manivel, S.M. Roopan, R.S. Kumar, F.N. Khan, Synthesis of 3-substituted [47] K. Engen, J. Savmarker, U. Rosenstrom, J. Wannberg, T. Lundback,
isoquinolin-1-yl-2-(cycloalk-2-enylidene) hydrazines and their antimicro- A. Jenmalm-Jensen, M. Larhed, Microwave heated flow synthesis of spiro-
bial properties, J. Chil. Chem. Soc. 54 (2009) 183e185. oxindole dihydroquinazolinone based IRAP inhibitors, Org. Process. Res.
[21] F.N. Khan, S.M. Roopan, V.R. Hathwar, S.W. Ng, 2-Chloro-3-hydroxymethyl- Dev. 18 (2014) 1582e1588.
6-methoxyquinoline, Acta Crystallogr. Sect. E.-Struct. Rep. Online 66 (2010) [48] W.G. Thomas, F.A.O. Mendelsohn, Angiotensin receptors: form and function
o201. and distribution, Int. J. Biochem. Cell. Biol. 35 (2003) 774e779.
[22] R. Subashini, S.M. Roopan, F.N. Khan, Synthesis and free radical scavenging [49] S.Y. Chai, R. Fernando, G. Peck, S.Y. Ye, F.A.O. Mendelsohn, T.A. Jenkins,
property of some quinoline derivatives, J. Chil. Chem. Soc. 55 (2010) A.L. Albiston, The angiotensin IV/AT4 receptor, Cell. Mol. Life Sci. 61 (2004)
317e319. 2728e2737.
[23] A. Bharathi, S.M. Roopan, A. Kajbafvala, R.D. Padmaja, M.S. Darsana, [50] P. Winstanley, Modern chemotherapeutic options for malaria, Lancet Infect.
G. Nandhini Kumari, Catalytic activity of TiO2 nanoparticles in the synthesis Dis. 1 (2001) 242e250.
of some 2,3-disubstituted dihydroquinazolin-4(1H)-ones, Chin. Chem. Lett. [51] P. Winstanley, S. Ward, Malaria chemotherapy, Adv. Parasitol. 61 (2006)
25 (2014) 324e326. 47e76.
[24] S.M. Roopan, F.R.N. Khan, Free radical scavenging activity of nitrogen [52] U. D'Alessandro, Existing antimalarial agents and malaria-treatment strate-
heterocyclics-quinazolinones and tetrahydrocarbazolones, Indian J. Hetero- gies, Exp. Opin. Pharmacother. 10 (2009) 1291e1306.
cycl. Chem. 18 (2008) 183e184. [53] F.K. Hansen, S.D.M. Sumanadasa, K. Stenzel, S. Meister, L. Marek,
[25] J. Palaniraja, S.M. Roopan, Iodine-mediated synthesis of indazolo- R. Schmetter, K. Kuna, A. Hamacher, B. Mordmuller, M.U. Kassack,
quinazolinones via a multi-component reaction, RSC Adv. 5 (2015) E.A. Winzeler, V.M. Avery, K.T. Andrews, S. Duffy, T. Kurz, Discovery of HDAC
8640e8646. inhibitors with potent activity against multiple malaria parasite life cycle
[26] D.J. Connolly, D. Cusack, T.P.O. Sullivan, P.J. Guiry, Synthesis of quinazoli- stages, Eur. J. Med. Chem. 82 (2014) 204e213.
nones and quinazolines, Tetrahedron 61 (2005) 10153e10202. [54] E.R. Derbyshire, R.K. Guy, J. Min, J. Clardy, W.A. Guiguemde, J.A. Clark,
[27] I. Khan, A. Ibrar, W. Ahmed, A. Saeed, Synthetic approaches, functionalization M.C. Connelly, A.D. Magalhaes, R.K. Guy, J. Clardy, Dihydroquinazolinone
and therapeutic potential of quinazoline and quinazolinone skeletons: the inhibitors of proliferation of blood and liver stage malaria parasites, Anti-
advances continue, Eur. J. Med. Chem. 90 (2015) 124e169. microb. Agents Ch 58 (2014) 1516e1522.
[28] I. Khan, A. Ibrar, N. Abbas, A. Saeed, Recent advances in the structural library [55] V. Tzin, G. Galili, New insights into the Shikimate and aromatic amino acids
of functionalized quinazoline and quinazolinone scaffolds: synthetic ap- biosynthesis pathways in plants, Mol. Plant 3 (2010) 956e972.
proaches and multifarious applications, Eur. J. Med. Chem. 76 (2014) [56] M. Okvist, P. Kast, R. Dey, S. Sasso, E. Grahn, U. Krengel, 1.6 Å crystal structure
K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630 629

of the secreted chorismate mutase from mycobacterium tuberculosis: novel [79] L. Boyman, G.S.B. Williams, D. Khananshvili, I. Sekler, W.J. Lederer, NCLX: the
fold topology revealed, J. Mol. Biol. 357 (2006) 1483e1499. mitochondrial sodium calcium exchanger, J. Mol. Cell. Cardiol. 59 (2013)
[57] D. Rambabu, S.K. Kumar, B.Y. Sreenivas, S. Sandra, A. Kandale, M.V.B. Rao, 205e213.
M. Pal, P. Misra, Ultrasound-based approach to spiro-2, 3-dihydroquinazolin- [80] T. Kalogeris, C.P. Baines, M. Krenz, R.J. Korthuis, Cell biology of ischemia/
4(1H)-ones: their in vitro evaluation against chorismate mutase, Tetrahe- reperfusion injury, Int. Rev. Cell Mol. Biol. 298 (2012) 229e317.
dron Lett. 54 (2013) 495e501. [81] H. Hasegawa, M. Muraoka, K. Matsui, A. Kojima, A novel class of sodium/
[58] R.N. Dubois, S.B. Abramson, R.A. Gupta, L.S. Simon, L. Crofford, L.B.A.V. Putte, calcium exchanger inhibitors: design, synthesis, and structure-activity re-
P.E. Lipsky, Cyclooxygenase in biology and disease, FASEB J. 12 (1998) lationships of 4-phenyl-3-(piperidin-4-yl)-3, 4-dihydro-2(1H)-quinazoli-
1063e1073. none derivatives, Bioorg. Med. Chem. Lett. 16 (2006) 727e730.
[59] E. Manivannan, S.C. Chaturvedi, Analogue-based design, synthesis and [82] H. Hasegawa, M. Muraoka, K. Matsui, A. Kojima, Discovery of a novel potent
docking of non-steroidal anti-inflammatory agents. Part 2: methyl sulfanyl/ Naþ/Ca2þ exchanger inhibitor: design, synthesis and structure-activity re-
methyl sulfonyl substituted 2, 3-diaryl-2, 3-dihydro-1H-quinazolin-4-ones, lationships of 3, 4-dihydro-2(1H)-quinazolinone derivatives, Bioorg. Med.
Eur. J. Med. Chem. 20 (2012) 7119e7127. Chem. Lett. 13 (2003) 3471e3475.
[60] R.S. Hoonur, B.R. Patil, D.S. Badiger, R.S. Vadavi, K.B. Gudasi, P.R. Dandawate, [83] H. Hasegawa, K. Matsui, M. Muraoka, M. Ohmori, A. Kojima, A novel class of
M.M. Ghaisas, S.B. Padhye, M. Nethaji, Transition metal complexes of 3-aryl- sodium/calcium exchanger inhibitor: design, synthesis, and structure-
2-substituted 1, 2-dihydroquinazolin-4(3H)-one derivatives: new class of activity relationships of 3, 4-dihydro-2(1H)-quinazolinone derivatives, Bio-
analgesic and anti-inflammatory agents, Eur. J. Med. Chem. 45 (2010) organ. Med. Chem. 13 (2005) 3721e3735.
2277e2282. [84] Y. Endo, K. Kawai, T. Asano, S. Amano, Y. Asanuma, K. Sawada, K. Ogura,
[61] S. Arora, X.I. Wang, S.M. Keenan, C. Andaya, Q. Zhang, Y. Peng, W.J. Welsh, N. Nagata, N. Ueo, N. Takahashi, Y. Sonoda, N. Kamei, Discovery and SAR
Novel microtubule polymerization inhibitor with potent anti-proliferative study of 2-(4-pyridylamino)thieno[3, 2-d] pyrimidin-4(3H)-ones as soluble
and anti-tumor activity, Cancer Res. 69 (2009) 1910e1915. and highly potent PDE7 inhibitors, Bioorg. Med. Chem. Lett. 25 (2015)
[62] A. Kamal, E.V. Bharathi, J.S. Reddy, M.J. Ramaiah, M.K. Reddy, A. Viswanath, 649e653.
T.L. Reddy, T.B. Shaik, D. Dastagiri, N.C.V.L. Pushpavalli, M.P. Bhadra, Syn- [85] J. Guo, A. Watson, J. Kempson, M. Carlsen, J. Barbosa, K. Stebbins, D. Lee,
thesis and biological evaluation of 3, 5-diaryl isoxazoline/isoxazole linked 2, J. Dodd, S.G. Nadler, M. McKinnon, J. Barrish, W.J. Pitts, Identification of
3-dihydroquinazolinone hybrids as anticancer agents, Eur. J. Med. Chem. 46 potent pyrimidine inhibitors of phosphodiesterase 7 (PDE7) and their ability
(2011) 691e703. to inhibit T cell proliferation, Bioorg. Med. Chem. Lett. 19 (2009) 1935e1938.
[63] S.M. Roopan, J.S. Jin, F.N. Khan, R.S. Kumar, An efficient one pot-three [86] E. Lorthiois, A.-K. Mafroud, M. Idrissi, P. Bernardelli, A. Tertre, E. Proust,
component cyclocondensation in the synthesis of 2-(2-chloroquinolin-3- A. Descours, B. Bertin, F. Vergne, P. Soulard, L. Heuze, M. Coupe, C. Oliveira,
yl)-2, 3-dihydroquinazolin-4(1H)-ones: potential antitumor agents, Res. F. Moreau, R. Wrigglesworth, P. Berna, Spiroquinazolinones as novel, potent,
Chem. Intermed. 37 (2011) 919e927. and selective PDE7 inhibitors. Part 1, Bioorg. Med. Chem. Lett. 14 (2004)
[64] F.L. Faraj, M. Zahedifard, M.J. Paydar, C.Y. Looi, N.A. Majid, H.M. Ali, N. Ahmad, 4623e4626.
N.S. Gwaram, M.A. Abdulla, Synthesis, characterization, and anticancer ac- [87] P. Bernardelli, A.-K. Mafroud, F. Moreau, E. Chevalier, E. Lorthiois, F. Vergne,
tivity of new quinazoline derivatives against MCF-7 cells, Sci. World J. 2014 E. Proust, M. Idrissi, A. Descours, A. Tertre, N. Pham, C. Oliveira, P. Ducrot,
(2014) 1e15. B. Bertin, F. Berlioz-Seux, M. Coupe, P. Berna, M. Li, Spiroquinazolinones as
[65] M. Mahdavi, K. Pedrood, M. Safavi, M. Saeedi, M. Pordeli, S.K. Ardestani, novel, potent, and selective PDE7 inhibitors. Part 2: optimization of 5, 8-
S. Emami, A. Foroumadi, A. Shafiee, M. Adib, Synthesis and anticancer activity disubstituted derivatives, Bioorg. Med. Chem. Lett. 14 (2004) 4627e4631.
of N-substituted 2-arylquinazolinones bearing trans-stilbene scaffold, Eur. J. [88] P.P. Roux, J. Blenis, ERK and p38 MAPK-activated protein kinases: a family of
Med. Chem. 95 (2015) 492e499. protein kinases with diverse biological functions, Microbiol. Mol. Biol. Rev.
[66] B.J. Sargent, A.J. Henderson, Targeting 5-HT receptors for the treatment of 68 (2004) 320e344.
obesity, Curr. Opin. Pharmacol. 11 (2011) 52e58. [89] A. Cuenda, S. Rousseau, p38 MAP-Kinases pathway regulation, function and
[67] A.C. Dutton, N.M. Barnes, Anti-obesity pharmacotherapy: future perspectives role in human diseases, Biochim. Biophys. Acta 1773 (2007) 1358e1375.
utilising 5-HT2C receptor agonists, Drug Discov. Today Ther. Strateg. 3 (2006) [90] J.E. Stelmach, G. Scapin, J.R. Strauss, E.A. O'Neill, L. Liu, S. Singh,
577e583. P.M. Cameron, D.M. Schmatz, D.M. Zaller, S.B. Patel, J.V. Pivnichny,
[68] S. Ahmad, K. Ngu, K.J. Miller, G. Wu, C. Hung, S. Malmstrom, G. Zhang, C.E.C.A. Hop, E.A. Nichols, C.D. Schwartz, J.B. Doherty, Z. Wang, S.J. O'Keefe,
E. O'Tanyi, W.J. Keim, M.J. Cullen, K.W. Rohrbach, M. Thomas, T. Ung, Q. Qu, C.M. Thompson, Design and synthesis of potent, orally bioavailable dihy-
J. Gan, R. Narayanan, M.A. Pelleymounter, J.A. Robl, Tricyclic dihy- droquinazolinone inhibitors of p38 MAP kinase, Bioorg. Med. Chem. Lett. 13
droquinazolinones as novel 5- HT2C selective and orally efficacious anti- (2003) 277e280.
obesity agents, Bioorg. Med. Chem. Lett. 20 (2010) 1128e1133. [91] J.A. Hunt, S.X. McCormick, Z. Wang, F. Kallashi, J.E. Thompson, R.D. Ruzek,
[69] J.M. Gillbro, M.J. Olsson, The melanogenesis and mechanisms of skin- J.V. Pivnichny, S.J. O'Keefe, A. Woods, P.J. Sinclair, C.E.C.A. Hop, E.A. O'Neill,
lightening agents-Existing and new approaches, Int. J. Cosmet. Sci. 33 D.M. Zaller, I. Ita, S. Kumar, G. Porter, J.B. Doherty, p38 Inhibitors: piperidine-
(2011) 210e221. and 4-aminopiperidine-substituted naphthyridinones, quinolinones, and
[70] H. Kim, S. Lee, Induction of ATP synthase b by H2O2 induces melanogenesis dihydroquinazolinones, Bioorg. Med. Chem. Lett. 13 (2003) 467e470.
by activating PAH and cAMP/CREB/MITF signaling in melanoma cells, Int. J. [92] A. Schlapbach, R. Heng, F. Di Padova, A novel Pd-catalyzed cyclization reac-
Biochem. Cell B 45 (2013) 1217e1222. tion of ureas for the synthesis of dihydroquinazolinone p38 kinase inhibitors,
[71] P. Thanigaimalai, K. Lee, S. Bang, J. Lee, C. Yun, E. Roh, B. Hwang, Y. Kim, Bioorg. Med. Chem. Lett. 14 (2004) 357e360.
S. Jung, Evaluation of 3, 4-dihydroquinazoline-2(1H)-thiones as inhibitors of [93] A. Arif, Extraneuronal activities and regulatory mechanisms of the atypical
a-MSH-induced melanin production in melanoma B16 cells, Bioorgan. Med. cyclin-dependent kinase Cdk5, Biochem. Pharmacol. 84 (2012) 985e993.
Chem. 18 (2010) 1555e1562. [94] J. Zhu, W. Li, Z. Mao, Cdk5: mediator of neuronal development, death and the
[72] D.J. Triggle, Calcium channel antagonists: clinical uses-Past, present and response to DNA damage, Mech. Ageing Dev. 132 (2011) 389e394.
future, Biochem. Pharmacol. 74 (2007) 1e9. [95] R.M. Rzasa, T.D. Osslund, H. Wang, M.R. Kaller, D. Powers, X. Xiong, G. Liu,
[73] R.L. Kraus, Y. Li, A. Jovanovska, J.J. Renger, Trazodone inhibits T-type calcium V.J. Santora, W. Zhong, E. Magal, T.T. Nguyen, V.N. Viswanadhan,
channels, Neuropharmacology 53 (2007) 308e317. M.H. Norman, Structure-activity relationships of 3, 4-dihydro-1H-quinazo-
[74] J.C. Barrow, P. Bondiskey, C. Tang, S.M. Doran, K.E. Rittle, G.B. McGaughey, lin-2-one derivatives as potential CDK5 inhibitors, Bioorgan. Med. Chem. 15
J. Ballard, M.J. Marino, S.V. Fox, T.S. Reger, Y. Kuo, M.G. Bock, Z. Yang, (2007) 6574e6595.
G.D. Hartman, T. Prueksaritanont, S.L. Garson, V.K. Graufelds, R.L. Kraus, [96] D.B. Sattelle, D. Cordova, T.R. Cheek, Insect ryanodine receptors: molecular
V.N. Uebele, C.E. Nuss, Y. Li, J.J. Renger, Discovery of 4, 4-disubstituted qui- targets for novel pest control chemicals, Invert. Neurosci. 8 (2008) 107e119.
nazolin-2-ones as T-type calcium channel antagonists, ACS Med. Chem. Lett. [97] S. Fleischer, Personal recollections on the discovery of the ryanodine re-
1 (2010) 75e79. ceptors of muscle, Biochem. Biophys. Res. Commun. 369 (2008) 195e207.
[75] K.S. Schlegel, Z. Yang, T.S. Reger, Y. Shu, K.E. Rittle, P. Bondiskey, M.G. Bock, [98] Y. Zhou, Q. Feng, F. Di, Q. Liu, D. Wang, Y. Chen, L. Xiong, H. Song, Y. Li, Z. Li,
G.D. Hartman, R. Cube, C. Tang, Y. Kuo, T. Prueksaritanont, C.E. Nuss, Synthesis and insecticidal activities of 2, 3-dihydroquinazolin-4(1H)-one
S.M. Doran, S.L. Garson, R.L. Kraus, Y. Li, V.N. Uebele, J. Ballard, S.V. Fox, derivatives targeting calcium channel, Bioorgan. Med. Chem. 21 (2013)
J.J. Renger, J.C. Barrow, Discovery and expanded SAR of 4, 4-disubstituted 4968e4975.
quinazolin-2-ones as potent T-type calcium channel antagonists, Bioorg. [99] A. Engelman, P. Cherepanov, The structural biology of HIV-1: mechanistic
Med. Chem. Lett. 20 (2010) 5147e5152. and therapeutic insights, Nat. Rev. Microbiol. 10 (2012) 279e290.
[76] L. Hedstrom, IMP dehydrogenase: structure, mechanism and inhibition, [100] D.A. Babkov, V.T. Valuev-Elliston, M.P. Paramonova, A.A. Ozerov, A.V. Ivanov,
Chem. Rev. 109 (2009) 2903e2928. A.O. Chizhov, A.L. Khandazhinskaya, S.N. Kochetkov, J. Balzarini,
[77] R. Petrelli, P. Vita, I. Torquati, K. Felczak, D.J. Wilson, P. Franchetti, D. Daelemans, C. Pannecouque, K.L. Seley-Radtke, M.S. Novikov, Scaffold
L. Cappellacci, Novel inhibitors of inosine monophosphate dehydrogenase in hopping: exploration of acetanilide-containing uracil analogues as potential
patent literature of the last decade, Recent Pat. Anticancer Drug Discov. 8 NNRTIs, Bioorgan. Med. Chem. 23 (2015) 1069e1081.
(2013) 103e125. [101] J.W. Corbett, S.S. Ko, J.D. Rodgers, L.A. Gearhart, N.A. Magnus, L.T. Bacheler,
[78] H.L. Birch, G.M. Buckley, N. Davies, H.J. Dyke, E.J. Frost, P.J. Gilbert, S. Diamond, S. Jeffrey, R.M. Klabe, B.C. Cordova, S. Garber, K. Logue,
D.R. Hannah, A.F. Haughan, M.J. Madigan, T. Morgan, W.R. Pitt, A.J. Ratcliff e, G.L. Trainor, P.S. Anderson, S.K. Erickson-Viitanen, Inhibition of clinically
N.C. Ray, M.D. Richard, A. Sharpe, A.J. Taylor, J.M. Whitworth, S.C. Williams, relevant mutant variants of HIV-1 by quinazolinone non-nucleoside reverse
Novel 7-methoxy-6-oxazol-5-yl-2, 3-dihydro-1H-quinazolin-4-ones as transcriptase inhibitors, J. Med. Chem. 43 (2000) 2019e2030.
IMPDH inhibitors, Bioorg. Med. Chem. Lett. 15 (2005) 5335e5339. [102] Q. Hu, J. Kunde, N. Hanke, R.W. Hartmann, Identification of 4-(4-nitro-2-
630 K. Hemalatha, G. Madhumitha / European Journal of Medicinal Chemistry 123 (2016) 596e630

phenethoxyphenyl)pyridine as a promising new lead for discovering in- DMPK: drug metabolism and pharmacokinetics
hibitors of both human and rat 11b-hydroxylase, Eur. J. Med. Chem. 96 DPEPhos: (Oxydi-2, 1-phenylene)bis(diphenylphosphine)
(2015) 139e150. DPPA: diphenylphosphoryl azide
[103] C.M. Grombein, Q. Hu, R. Heim, S. Rau, C. Zimmer, R.W. Hartmann, 1- EDC: 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide
Phenylsulfinyl-3-(pyridin-3-yl)naphthalen-2-ols: a new class of potent and EEG: electroencephalogram
selective aldosterone synthase inhibitors, Eur. J. Med. Chem. 89 (2015) FLIPR: fluorometric imaging plate reader
597e605. GDP: guanosine diphosphate
[104] C.M. Grombein, Q. Hu, S. Rau, C. Zimmer, R.W. Hartmann, Heteroatom GMP: guanosine monophosphate
insertion into 3, 4-dihydro-1H-quinolin-2-ones leads to potent and selective GTP: guanosine triphosphate
inhibitors of human and rat aldosterone synthase, Eur. J. Med. Chem. 90 HCl: hydrochloric acid
(2015) 788e796. HIV-1: human immunodeficiency virus type 1
[105] Z. Guan, K. Hillrichs, C. Unlu, K. Rissanen, M. Nieger, A. Schmidt, Synthesis of HOBt: hydroxybenzotriazole
2-anilinobenzimidates, anthranilamides, and 2, 3dihydroquinazolin-4(1H)- HPLC: high performance liquid chromatography
ones from N-heterocyclic carbenes of indazole, Tetrahedron 71 (2015) HTRF: homogenous time resolved fluorescence
276e282. IL-1: interleukin 1
[106] J. Zhang, D. Ren, Y. Ma, W. Wang, H. Wu, CuO nanoparticles catalyzed simple IMP: inosine 50 -monophosphate
and efficient synthesis of 2, 3-dihydroquinazolin-4(1H)-ones and quinazolin- IMPDH: inosine-50 -monophosphate dehydrogenase
4(3H)-ones under ultrasound irradiation in aqueous ethanol under ultra- IRAP: insulin-regulated aminopeptidase
sound irradiation in aqueous ethanol, Tetrahedron 70 (2014) 5274e5282. KOH: potassium hydroxide
[107] Y. Shang, L. Fan, X. Li, M. Liu, Y(OTf)3-catalyzed heterocyclic formation via LiAlH4: lithium aluminium hydride
aerobic oxygenation: an approach to dihydro quinazolinones and quinazo- LR: lawesson's reagent
linones, Chin. Chem. Lett. 26 (2015) 1355e1358. mAchR: muscarinic acetylcholine receptors
[108] M.J. Mphahlele, M.M. Maluleka, T.A. Khoza, 2-Aryl-6, 8-dibromo-2, 3- MAPK: mitogen-activated protein kinase
dihydroquinazolin-4(1H)-ones as substrates for the synthesis of 2, 6, 8- MAPKK: MAPK kinase
triarylquinazolin-4-ones, Bull. Chem. Soc. Ethiop. 28 (2014) 81e90. MAPKKK: MAPKK kinase
[109] K. Hemalatha, G. Madhumitha, C.S. Vasavi, P. Munusami, 2, 3- MC1R: melanocortin receptor 1
Dihydroquinazolin-4(1H)-ones: visible light mediated synthesis, sol- MC4R: melanocortin receptor 4
vatochromism and biological activity, J. Photoch. Photobio. B 143 (2015) METH: methamphetamine
139e147. MITF: microphthalmia-associated transcription factor
[110] K. Hemalatha, G. Madhumitha, Inhibition of poly(adenosine diphosphate- MPO: myeloperoxidase
ribose) polymerase using quinazolinone nucleus, Appl. Microbiol. Bio- MTT: 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide
technol. (2016), http://dx.doi.org/10.1007/s00253-016-7731-1. MW: microwave
[111] K. Hemalatha, G. Madhumitha, N.A. Al-Dhabi, M.V. Arasu, Importance of NaBH4: sodium borohydride
fluorine in 2, 3-dihydroquinazolinone and its interaction study with lyso- NAD: nicotinamide adenine dinucleotide
zyme, J. Photoch. Photobio. B 162 (2016) 176e188. NaH: sodium hydride
[112] K. Hemalatha, G. Madhumitha, Study of binding interaction between N-boc: tert-butyloxycarbonyl
anthelmintic 2, 3-dihydroquinazolin-4-ones with bovine serum albumin by NBS: N-bromosuccinimide
spectroscopic methods, J. Lumin 178 (2016) 163e171. NCS: N-chlorosuccinimide
[113] K. Hemalatha, G. Madhumitha, L. Ravi, V.G. Khanna, N.A. Al-Dhabi, NCX: sodium calcium exchanger
M.V. Arasu, Binding mode of dihydroquinazolinones with lysozyme and its NMR: nuclear magnetic resonance
antifungal activity against Aspergillus species, J. Photoch. Photobio. B 161 NNRTIs: nonnucleoside reverse transcriptase inhibitors
(2016) 71e79. NO: nitric oxide
[114] K. Hemalatha, G. Madhumitha, Eco-friendly synthesis of palladium nano- NRTIs: nucleoside reverse transcriptase inhibitors
particles, environmental toxicity assessment and its catalytic application in NSAIDs: non-steroidal anti-inflammatory drugs
suzuki miyaura coupling, Res. J. Pharm. Tech 8 (2015) 1691e1700. PARP: poly(adenosine diphosphate-ribose)polymerase
PBMC: peripheral blood mononuclear cell
PCC: pyridinium chlorochromate
Abbreviations PCy3: tricyclohexylphosphine
Pd(PPh3)4: tetrakis(triphenylphosphine)palladium(0)
5-HT: 5-hydroxytryptamine Pd2dba3: tris(dibenzylideneacetone)dipalladium(0)
ADP: adenosine diphosphate PdCl2(dppf): bis(diphenylphosphino)ferrocene]palladium(II) dichloride
AMP: 50 -adenosine monophosphate PdCl2(PPh3)2: bis(triphenylphosphine)palladium(II) dichloride
AT4: angiotensin IV PDE7: phosphodiesterase 7
ATP: adenosine triphosphate PMB: p-methoxy benzyl
BCCP: biotin-carboxyl-carrier protein POMC: proopiomelanocortin
Boc2O: di-tert-butyl-dicarbonate PPA: polyphosphoric acid
cAMP: 30 ,50 -cyclic adenosine monophosphate Psi: per square inch
CDI: 1, 1-carbonyl diimidazole PTSA: p-toluenesulphonic acid
CDK1: cyclin-dependent kinase 1 PXR: pregnane X receptor
CDK5: cyclin-dependent kinase 5 REM: rapid eye movement
CF: continuous flow RT: reverse transcriptase
cGMP: 30 ,50 -cyclic guanosine monophosphate RyR: ryanodine receptor
CGRP: calcitonin gene-related peptide SAR: structural-activity relationship
CM: chorismate mutase SiC: silicon carbide
CNS: central nervous system TBAF: tetra-n-butylammonium fluoride
COX: cyclooxygenase TFA: trifluoroacetic acid
CREB: cAMP-responsive element-binding protein THF: tetrahydrofuran
CuO: copper (II) oxide TMSNCO: trimethylsilylisocyanate
CYP: cytochromes P450 TNFa: tumor necrosis factor alpha
DAST: diethylaminosulfur trifluoride UV: ultraviolet
DBU: 1, 8-diazabicycloundec-7-ene WBC: white blood cells
DCM: dichloromethane XMP: xanthosine 50 - monophosphate
DEAD: diethyl azodicarboxylate Xphos: 2-dicyclohexylphosphino-20 , 40 , 60 -triisopropylbiphenyl
DMAP: dimethylaminopyridine Y(OTf)3: ytterbium(III) triflate
DME: dimethoxyethane a-MSH: alpha-melanocyte stimulating hormone

You might also like