HOME JOURNALS CONTACT

American Journal of Drug Discovery and Development

Year: 2017 | Volume: 7 | Issue: 1 | Page No.: 1-24
DOI: 10.3923/ajdd.2017.1.24
Undeniable Pharmacological Potentials of Quinazoline Motifs in Therapeutic Medicine
Olayinka Oyewale Ajani, Oluwatosin Yemisi Audu , Damilola Victoria Aderohunmu, Fisayo Elizabeth Owolabi and Ayodele Ojo Olomieja

Abstract: Despite the advances in technology and understanding of biological systems, drug discovery is still a lengthy, expensive, difficult and inefficient process owing to low rate of new therapeutic discovery. Among the numerous N-heterocyclic scaffolds, quinazoline core structural frameworks have received considerable attention because they form a privileged class of pharmacophores with diverse spectrum of therapeutic potential. Various conventional synthetic approaches methodologies for quinazoline synthesis by various synthetic chemists were explored in this review wherein their bases for structural validation were expatiated using analytical data and spectroscopic means such as FT-IR, UV-Visible, 1H- and 13C-NMR as well as mass spectra. Quinazoline derivatives are among the most useful heterocyclic compounds from both synthetic and medicinal chemistry aspects. They are considered as important precursors for the synthesis of various physiologically significant and pharmacologically utilized molecules. This present study unveils quinazoline core as a multifunctional nucleus which serves as a resourceful toolbox of information for synthetic modifications of old existing candidates in order to tackle drug resistance bottlenecks in therapeutic medicine. Based on diverse bioactivities and pharmacological potentials were explored, quinozaline motifs were concluded to be arsenals of warfare against infectious diseases in therapeutics. Hence, quinazolines might pave way to new drug discovery for fighting infectious diseases and increase researchers’ choice of quinazoline as excellent candidates for future drug design.

Fulltext PDF Fulltext HTML

How to cite this article
Olayinka Oyewale Ajani, Oluwatosin Yemisi Audu, Damilola Victoria Aderohunmu, Fisayo Elizabeth Owolabi and Ayodele Ojo Olomieja, 2017. Undeniable Pharmacological Potentials of Quinazoline Motifs in Therapeutic Medicine. American Journal of Drug Discovery and Development, 7: 1-24.

Keywords: HIV-1, antitumor activity, structure activity relationship, Quinazoline and cytotoxic study

INTRODUCTION

Heterocyclic compounds form the basis for pharmaceutical industrial operations, especially, in the synthesis of drugs. Also, most marketed drugs constitute of common structural units of heterocyclic compounds which could also be useful in the synthesis of biologically active agrochemicals1. Nitrogen-containing heterocyclic compounds are very crucial in drug design. In the recent years, there has been considerable attention on the preparation of useful N-heterocyclic compounds in organic synthesis2,3. Thus, the motivation for this present study was the known widespread application of benzo-fused N-heterocycles. For instance, in the pharmaceutical industry, among the top two hundred branded drugs, we have more than 75% of them which have heterocyclic fragments in their structures4. Quinazoline is a fused bicyclic heterocyclic framework which was formerly known as benzo-1,3-diazine or 1,3-diazanaphthalene5. Natural occurrence of quinazoline core has shown it to be isolated from the Chinese plant aseru6. Quinazolines are classes of fused heterocycles that are of significant interest because of the varied range of their biological properties7,8. They have also drawn a huge attention owing to their expanded applications in the field of pharmaceutical chemistry9.

Furthermore, quinazolines are considered an attractive target for medicinal chemists, because they are the scaffold of several potent antitumor drugs. Leading examples are the well-known erlotinib (trade name tarceva) and gefitinib (trade name iressa)10-12. Among the heterocyclic compounds, quinazoline is of great importance due to its important biological actions as well as synthetic applications in medicinal chemistry. Numerous compounds containing quinazoline moieties have been reported to exhibit diverse biological and pharmacological properties which include antimicrobial7, antitumor9, antimitotic10, anticancer11 and kinase inhibitory12 activities among others. The specific objectives are to: Highlight major synthetic pathways of valuable quinazoline derivatives, explore recent advances in pharmacological diversity of quinazoline for effective drug design and draw attention of researchers into the beneficial role of quinazoline in fighting diseases.

Classification of quinazolinones: Quinazoline can have different types of substituents attached to it, forming different derivatives of the parent quinazoline. Substituted quinazolines have properties which largely depend on: The nature of the substituent, position of substituent and type of conjugation in pyrimidine ring7. Quinazolines have many derivatives such as ketoquinazolines with a ketone substituent attached to it; it can otherwise be referred to as quinazolinones. Quinazolinones which are based on the substitution patterns of the ring system can be classified into the following five categories: 2-substituted-4(3H)-quinazolinones, 3-substituted-4(3H)-quinazolinones, 4-substituted-quinazolin ones, 2,3-disubstituted-4(3H)-quinazolinones and 2,4-disubstituted-4(3H)-quinazolinones7,13.

CHEMISTRY

Synthetic methods: In the past decade, a variety of synthetic methods have been employed for the preparation of functionalized quinazoline and quinazolinone motifs derivatives have been synthesized with the aim of obtaining more biologically active materials. Due to high diversity and level of interest in the current domain, many synthetic approaches have been utilized in preparing quinazoline and their polycyclic derivatives which have resulted in high output in highly relevant publication databases. 4-(Furan-2-yl)-4,4a,5,6,7,8-hexahydroquinazolin-2(3H)-one containing completely saturated pyrimidine ring system was synthesized by the reaction of α, β-unsaturated carbonyl otherwise known as chalcone with urea in the presence of silica sulfuric acid as the reusable heterogeneous catalyst14. Some specific synthesis of some selected quinazoline derivatives were as given below:

Synthesis via thermal cyclization of o-ureidobenzoic acid: Reaction of substituted anthranilic acid with potassium cyanate was reported to give o-ureidobenzoic acid which was subsequently treated with acid or alkali under high thermal condition for easy cyclization in order to produce substituted-1,2,3,4-tetrahydro-2,4-dioxoquinazolines, 1 as shown in the Scheme 1 below15.

Solid phase synthetic approach: Solid phase synthetic reaction of aminobenzophenone with benzylamine was successfully achieved using Ceryl Ammonium Nitrate (CAN) in acetonitrile to afford substituted-2,4-diphenylquinazoline, 2 in good yield16. It is worthy to note that 2.5 equivalent of benzylamine precursor and 0.1 equivalent of CAN-TBHP catalyst were used for efficient production of quinazoline derivative, 2 as show in Scheme 2.

Cascade reaction of (2-aminophenyl) methanols with aldehydes: According to a report, an efficient cascade reaction of (2-aminophenyl)methanols with aldehydes using the combination of cerium nitrate hexahydrate and ammonium chloride under the influence of catalytic amount of copper chloride (0.2 eq.) led to the formation of a wide range of substituted-2-phenylquinazolines, 3a in good yields17 as shown in Scheme 3.

Scheme 1:
Route for the synthesis of substituted-1,2,3,4-tetrahydro-2,4-dioxoquinazolines, 1

Scheme 2:
Route for the synthesis of substituted-2,4-diphenylquinazoline, 2

Scheme 3:
Route for the synthesis of substituted-2-phenylquinazolines, 3a

Scheme 4:
Route for the ruthenium-catalyzed synthesis of substituted-2-phenylquinazolines, 3b

The method was a convenient and practical strategy for synthesis of 2-substituted quinazoline derivatives that have only one phenyl substituent on heterocyclic ring of pyrimidine17.

Ruthenium-catalyzed dehydrogenative synthetic approach: A commercially available Ru3(CO2)12/Xantphos/t-BuOK catalyst system enables a straight forward ruthenium-catalyzed dehydrogenative synthesis of 2-arylquinazolines, 3b. It was recently adopted for the preparation of 2-arylquinazolines, 3b with substitution on its benzene ring by heating the reacting mixture under reflux for 16 h at 130°C as shown in Scheme 4. It was reported that this synthetic method offered operational simplicity, high atom efficiency and broad substrate scope18.

Microwave-assisted synthetic approach: A tandem experimental and efficient synthesis of 2,4-disubstituted-6,8-dibromoquinazolines, 4 through chemoselective sequential bis-SNAr/bis-Suzuki-Miyaura reactions under microwave irradiation technique has been recently reported19. This study was reported to allow rapid and efficient access to desired products in high yields when the procedure started with the bromination of 2-aminobenzonitrile as shown in Scheme 5.

β-Cyclodextrin-mediated MCR approach: β-cyclodextrin (β-CD) mediated multi-component reaction under microwave irradiation has been considered as green processes which have remarkable advantages over the reactions performed under conventional heating20.

Scheme 5:
Microwave-assisted synthesis of 2,4-disubstituted-6,8-dibromoquinazolines, 4

Scheme 6:
β-Cyclodextrin-mediated synthesis of dihydroisoindolo[2,1-a]quinazoline-5,11-dione, 5

Scheme 7:
Route for the aerobic synthesis of 2,6-disubstituted quinazoline, 6

Scheme 8:
Route for the synthesis of substituted 2-arylquinazolin-4(3H)-ones, 7

They have successfully used this approach in the synthesis of dihydroisoindolo[2,1-a]quinazoline-5,11-dione derivatives, 5 by reaction of isatoic anhydride, amine and 2-formylbenzoic acid as MCR using β-cyclodextrin as shown in Scheme 6.

Aerobic cyclizative synthetic approach: Thermal cyclization reaction of aldehydes with 2-aminobenzylamines after refluxing for 2 h in the presence of acetonitrile was reported to give an imine intermediate which subsequent underwent efficient aerobic oxidative cyclization in the presence of CuCl/DABCO/4-HO-TEMPO as the catalysts and oxygen as the terminal oxidant to afford 2,6-disubstituted quinazoline 6 as shown in Scheme 721.

Three-component one-pot synthetic approach: An operational simple palladium-catalyzed three-component reaction of readily available 2-aminobenzamides, aryl halides and tert-butyl isocyanide was efficiently constructed under one-pot to afford substituted 2-arylquinazolin-4(3H)-ones, 7 in good yields22 as shown in Scheme 8. This synthetic study was reported to proceed via a palladium-catalyzed isocyanide insertion/cyclization sequence at an elevated temperature of 145°C in toluene solvent22. Similar reaction condition was used to synthesized quinazolin-4(3H)-one derivative by isonitrile insertion in 1.5 molar equivalent of cesium carbonate (Cs2CO3) in up to 10 h reflux in toluene solvent23.

Scheme 9:
Route for the synthesis of substituted 2-arylquinazolin-4(3H)-ones, 8

Scheme 10:
Route to substituted 2-methyl-3-((1-phenylethylidene)amino)quinazolin-4(3H)-one, 9

CuI-DMEDA mixed catalyst synthetic approach: An efficient reaction of substituted 2-bromo-benzonitriles with amidines allows an economical and practical synthesis of 4-aminoquinazoline and 2,4-diaminoquinazoline derivatives24. The reaction was carried out in the basified medium using CuI-DMEDA mixed catalyst reaction technique at a refluxing temperature of 80°C in DMF solvent as shown in Scheme 9.

Synthesis of Schiff base via acetic acid catalyzed approach: In quest for the quinazoline-based synthesis of some novel heterocyclic Schiff bases, a series of novel quinazolinone 9 has been designed and achieved recently25. This targeted compounds were prepared by condensation of 3-amino-2-methyl-3H-quinazolin-4-one with different substituted acetophenones in presence of acetic acid under classical procedure. The 3-amino-2-methyl-3H-quinazolin-4-one reactive intermediate used was earlier synthesized by the reaction of anthranilic acid with acetic anhydride under refluxing25 for 3 h. The pathway for this is as shown in Scheme 10.

BIOLOGICAL ACTIVITIES

Biological importance of quinazoline: Quinazoline and its quinazolinone skeletons are recurrently encountered heterocyclic com pounds in medicinal chemistry literature with applications including antibacterial, analgesic, anti-inflammatory, CNS depressant, anticonvulsant, antifungal, antimalarial, anticoccidial, anticancer activities among others. The wide reported biological activities of quinazoline derivative might have emanated from the fact that its corresponding monocyclic counterpart, pyrimidines are being prebiotic in nature to living cells in biodiversity which made them to be highly privileged motifs for the development of molecules of biological and pharmaceutical interest26.

Anticancer activity: Cancer is a disease characterized by a shift in the controlled mechanisms that govern cell proliferation and differentiation. Food and Drug Administration (FDA) has approved several quinazoline derivatives such as gefitinib, erlotinib, lapatinib and vandetanib as salient anticancer drugs. Compound 10 was found to be a particularly active growth inhibitor of the renal cancer (GI50 = 4.07 μM), CNS cancer (GI50 = 7.41 μM), ovarian cancer (GI50= 7.41 μM) and non-small cell lung cancer (GI50 = 7.94 μM)27. Investigation carried out on the anti-proliferative activity of quinazoline-based compounds against breast cancer cell line, revealed that 11 inhibited the cell growth at as low as IC50 = 5.52 μM28. Quinazolinedione-based redox modulators 12 had been discovered as therapy for pancreatic cancer since the oxidative stress induced by it, promoted activation of stress kinases (p38/JNK) resulting in cancer cell death29. Compound 13 has broad spectrum anti-cancer showing efficiency toward numerous cell lines that belong to different tumor subpanels (Fig. 1)30. In the evaluation of HepG2 xenograft model, 14 exhibited significant cancer growth inhibition with low host toxicity in vivo31. Quinazoline 15 was the most promising among the motifs screened against human32 cancer cell line A549.

EGFR inhibitory activity: The epidermal growth factor receptor (EGFR, erbB1, Her1) is a member of the erbB receptor protein33 which plays a critical role in many of the signal transduction pathways that regulate numerous cellular functions, such as proliferation, differentiation, migration, angiogenesis and apoptosis. The EGFR has been clinically validated as a rational target for cancer therapy and several small molecule inhibitors have been developed and released as quinazoline-based reversible inhibitor34, e.g., icotinib 16 and irreversible inhibitors namely; afatinib 1735, canertinib 18 and newly reported36 afatinib derivative 19 as shown in Fig. 2.

Fig. 1:
Some quinazoline derivative with reported anti cancer activity

Fig. 2:
Some reversible and irreversible EGFR inhibitors

According to results of the molecular docking study showed in Fig. 3, the comparison between the binding modes of the 6,7-dimethoxy-derivative 20 (LASSBio-1814; IC50 = 2.37 μM for EGFRwt and 1.02 μM for VEGFR-2) and the 6,7-methylenedioxy-derivative 21 (LASSBio-1815; IC50 = 34.6 μM for EGFRwt and 26.9 μM for VEGFR-2) demonstrated that the structural modification performed led to the loss of important hydrophobic interaction with Val726, Leu718 and Leu792 amino acid residues in EGFRwt binding site and with Val848, Leu840 and Phe918 amino acid residues in VEGFR-2 binding site37.

Antiproliferative activity: The successful synthesis of some 13-aryl-13H-benzo[g]benzothiazolo [2,3-b]quinazoline- 5,14-dione derivatives was reported and compound 22 (IC50 = 5.52 μM) was established as the most potent antiproliferative compound against two different human cancer cell lines38. Compound 23 displayed broad spectrum of antiproliferative activity on NCI 60-cell lines panel39 with mean GI50 of 1.53 μM. Antiproliferative activity investigation of 4-substituted-piperazine-1-carbodithioate derivatives of 2,4-diaminoquinazoline revealed 24, 25 and 26 as the most active members with IC50 values in the range of 1.58-2.27, 1.84-3.27 and 1.47-4.68 μM, respectively against five cancer cell lines examined (Fig. 4)40.

Antitubercular activity: There is an urgent demand for new antitubercular drugs possessing novel mechanisms of action with the goal of developing more effective combination treatments against drug resistant forms of Mycobacterium tuberculosis41. The discovery of 27 as potent apoptosis inducers with strong antitubercular activity was reported42. An unexpected intramolecular condensation with the formation of 3,4-dihydrobenzo[g]quinazoline heterocyclic system took place via click chemistry and the rifamycin mimetic obtained, 28 was reported to inhibit the growth of Mycobacterium tuberculosis appreciably43.

Fig. 3(a-d):
(a) Binding interactions of 20 (yellow) with EGFRwt, (b) Binding interactions of 21(gray) with EGFRwt, (c) Binding interactions of 20 (yellow) with VEGFR-2 and (d) Binding interactions of 21 (gray) with VEGFR-2. Docking studies were performed with the GOLD 5.1program. Apolar hydrogen atoms were omitted to improve clarity. The images were generated with PyMol software37

Compounds 29 exhibited significant anti-tubercular activity at MIC values 50 μM and the in vitro cytotoxicity data using THP-1 cells indicated that 29 was safe as its MIC value was much lower than the cytotoxic value as documented44 and shown in Fig. 5.

Anti-inflammatory activity: Quinazoline exhibits containment of inflammatory disorders such as osteoarthritis45 neurodegenerative impairments46 and inflammatory bowel syndrome47 and some of the anti-inflammatory drug bearing quinazolone nucleus48 are 30 and 31. In vivo experiment revealed that compound 32, could attenuate LPS-induced ALI in rats, via decreasing inflammatory cytokine production48, protein concentration in the BALF, pathological changes and macrophage infiltration. Schiff base 33 (IC50 = 52±0.81 μM) was reported to possess excellent anti-inflammatory due to its IC50 lower than that of aspirin standard drug49 (IC50 = 66±1.24 μM). Compound 34 (62.08% inhibition)50 showed in Fig. 6, competed favourably with indomethacin standard at 1.67 mL as the mean difference in paw volume after 4 h. Using Tumor Necrosis Factor-α (TNF-α) as the biomedical pathway, 35 was discovered to be a promising anti-inflammatory quinazoline derivative with 68% average inhibition on PDE4B enzyme at concentration51 of 10 μM (Fig. 6).

Fig. 4:
Quinazolinescaffolds with antipoliferative activity against various cell lines

Fig. 5:
Selected quinazoline with antitubercular activity

Fig. 6:
Reported quinazoline motifs with anti-inflammatory activity

Antileishmanial activity: In the recent review about antileishmanial drug discovery, quinazoline core was not left out52. Compound 36 (IC50 = 3.16 μM) exhibited higher anti-amastigote activity against L. donovani when compared with standard drug sodium stibogluconate53 (IC50 = 7.92 μM). A series of N2,N4-disubstituted quinazoline-2,4-diamines was synthesized and tested against Leishmania donovani and L. amazonensis intracellular amastigotes, wherein 37 emerged as the highly promising antileishmanial among the group54. Compounds 38 and 39 were reported to be effective against L. donovani at EC50 of 0.61±0.13 and 0.38±0.09 μM, respectively55. A series of quinazoline-2,4,6-triamine were synthesized and evaluated in vitro against Leishmania mexicana. Among them, 40 showed activity on promastigotes and intracellular amastigotes as well as low cytotoxicity in mammalian cells56. When administered in vivo in a hamster model, percentage inhibition of L. donovani parasite was found57 to be 73.15±12.69 μM for compound 41 (Fig. 7).

Antitumor activity: Most of the antitumor agents exert their effects by inhibiting both at the protein level and/or transcription level. Antitumor efficacy of quinazoline showed them to be biomimetic to the quinoxaline motif. This might be because they both belong to benzodiazine family58. Compound 42 possessed remarkable broad-spectrum antitumor activity which was almost sevenfold more active than the known drug 5-fluorouracil (5-FU) with GI50 values of 3.16 and 22.60 μM, respectively59. Among the series designed in another study, compounds 43 proved to be fifteen fold more active than the known antitumor 5-FU, with MG-MID GI50, TGI and LC50 values of 1.5, 46.8, 93.3 μM, respectively60. Caspase-3 activity and cell cycle regulation studies revealed that 44 exerted antitumor properties61 with IC50 of 3.4 μM. Bioassay results indicated that most of the prepared compounds demonstrated good activities against various cancer cells with 6-chloro-quinazoline derivatives 45 being the most active antitumor among the series62. Compared with the parental dasatinib, most of the new compounds synthesized, especially 2,4,6-trimethylaniline 46, demonstrated significant antitumor activities against six cell lines63 (Fig. 8).

Kinase inhibitory activity: The erythropoietin-producing hepatocellular (Eph) receptor tyrosine kinases have well established roles in the multitude of physiological and pathological processes. According to high throughput screening result, compound 47 displayed potent inhibitory activities64 toward EphA2 with IC50 of 0.17 μM. Out of the six quinazoline analogues synthesized via Biginelli approach, 48 was the most effective inhibitor of kinase65; although, being the most toxic too.

Also, compound 49 carrying dioxygenated rings fused on the benzenoid portion and the biphenylamino substituent on pyrimidine portion of quinazoline, was identified as inhibitor of kinase66 at IC50 of 0.08±0.01 μM. Compound 50 provided a potent starting point (IC50 = 12 nM) for inhibition of Mps1 kinase with submicromolar cellular toxicity, reduced MW and TPSA, while 51 provided a further boost in biochemical potency67 with IC50 = 2 nM. Amino acid derived quinazoline 52 possessed sub-nanomolar inhibition of Rock and PKA, nanomolar potency in ppMLC cell based assays, low to fair cytochrome P-450 inhibition and good human microsomal stability67 (Fig. 9).

Anticonvulsant activity: Methaqualone 53, as a quinazoline analog, is an important landmark in the field of synthetic anticonvulsants68. The modification of 53 was carried out to obtain quinazolinone scaffold 54 which proved to be twofold more active than anticonvulsant drug sodium valproate69.

Fig. 7:
Some quinazoline motifs with anti-leishmanial activity

Fig. 8:
Some quinazoline motifs with antitumor activity

Fig. 9:
Some quinazoline motifs with kinase inhibitory activity

Fig. 10:
Some quinazoline motifs with anticonvulsant activity

According to the findings of another study, 8-substituted- 4(3H)-quinazolinone 55 demonstrated a better anticonvulsant activity and lower toxicity than the reference drugs70. Compound 56 showed a significant anticonvulsant activity at 200 mg kg–1 (dose level) and marked CNS depressant activities associated with sedation and hypnosis71 (Fig. 10).

Antimalarial activity: The strong effect of quinazoline derivative 57 as an antimalarial motif on human erythrocytes and on cell membrane molecular model was reported72, while quinazoline 58 was reported to inhibit the growth of Plasmodium falciparum73 at IC50of 43.4 nM. Among the series of 4(3H)-quinazolinones screened against P. berghei, 59 was the most active antimalarial compound74. Owing to the fact that malaria has become a devastating global health issue in tropical and subtropical regions, other research efforts yielded promising antimalarial quinazoline derivatives75-77 60 and 62 (Fig. 11).

HDAC inhibitory activity: Histone deacetylases (HDAC) play crucial roles in numerous biological processes78. The encouraged activity of compound 63 against HDAC conformed to the reported information that hydroxamic acid generally showed more potent HDAC inhibitory activity than carboxylic acids79 while quinazoline 64 with meta-substitution and unsaturation near hydroxamic showed even more potency78 with IC50 of 0.16±0.02 μM. Out of 26 different structural classes that showed anti-inflammatory effects in a pre-screen in HEK293T cells, quinazoline 65 was one of the best three classes which exhibited improved in vitro HDAC inhibition in a commercial fluorescence assay80 (Fig. 12).

Anti-HIV activity: Synthesis and anti-HIV1 activity of quinazoline-4(3H)-one derivatives was evaluated, where it was found that 66 exhibited wide range81 of anti-HIV-1. Some 2,3-disubstituted quinazolin-4(3H)-ones synthesized by condensation of benzo[1,3]oxazine-4-one and primaquine were screened in vitro against HIV-1 in MT-4 cell and 67 was reported to exhibit 15% maximum protection against replication of HIV-1 (IIIB) in acutely infected MT-4 cells82. A series of dihydrobenzo[h]quinazoline derivatives were synthesized for the purpose of anti-HIV examination83. They reported that 68 (IC50 = 2.11 μM) emerged as the best anti-HIV among the series with therapeutic index of 1.89×105 (Fig. 13).

Fig. 11:
Some quinazoline motifs with antimalarial activity

Fig. 12:
Some quinazoline motifs which served as HDAC inhibitors

Fig. 13:
Some quinazoline motifs with anti-HIV activity

Concerning the mechanism of action as anti-HIV, these compounds were reverse transcriptase inhibitors.

Phosphodiesterase (PDE) inhibitors: The PDE families contain many splice variants that are mostly unique in tissue-expression patterns, gene regulation, enzymatic regulation by phosphorylation and regulatory proteins, subcellular localization and interaction with association proteins84. The inhibitory potencies of quinazoline-4-thione 69 at submicromolar levels against the catalytic domain of PDE7 was reported85. Small-molecule phosphodiesterase probe investigated later, established 4-aminoquinazoline 70 and the 4-indanylquinazoline 71 as PDE1 inhibitors that readily cross the blood brain barrier86. Administration of 72 as optimized PDE7 inhibitor ameliorated brain damage and improved behavioral outcome in a permanent middle cerebral artery occlusion (pMCAO) stroke model87 (Fig. 14).

Antihypertensive activity: All members of the series of quinazoline synthesized were examine in vivo and showed moderate to good antihypertensive activity in albino rat88. Out of these templates that operated through α1-adrenergic blocking mechanism, 73 emerged as the most potent. In another study, 74 was reported to exhibit antihypertensive activity greater than the reference drug prazocine89. Quinazoline motif 75 was shown to have considerable impact on the heart rate and blood pressure based on it substitution pattern90 (Fig. 15). Inhibition of PARP by L-2286 influenced beneficially, the signaling pathways in hypertension induced cardiac remodeling91. The effect of L-2286 on Systolic Blood Pressure (SBP) was investigated in a study, where it was discovered that during the 32-week treatment period, systolic blood pressure of hypertensive (SHR-C, SHR-L) rats was significantly higher than that of normotensive (WKY) rats (at the age of 10 weeks SHR92: 180±5.6 mmHg, WKY: 130±5.4 mmHg, at the age of 42 weeks SHR-C: 230±6.3, SHR-L: 225±2.4 mmHg, WKY-C: 130±5.4, WKY-L: 135±5.5 mmHg, p < 0.05; SHR-C vs. WKY-C, SHR-C vs. WKY-L p<0.05; SHR-L vs. WKY-C, SHR-L vs. WKY-L p<0.05). Treatment with L-2286 did not alter systolic blood pressure of SHR animals during the 32-week treatment period (Fig. 16).

Melanin-concentrating hormone receptor 1 antagonists: Melanin-concentrating hormone receptor 1 (MCHR1) antagonising quinazoline derivatives are proved to possess remarkable and distinct anti-obesity activity93. Pharmacokinetic profile showed that oral administration of 30 mg kg–1 b.i.d. of 4-morpholinyl quinazoline 76 led to 12% weight reduction within fourteen days in DIOC57BL/6 J. mice. The 4-amino-2-cyclohexylamino quinazoline 7794 and 4-dimethylamino quinazoline 78 were reported to exhibit highly promising anti-obesity for suitable weight loss programme due to their good affinity for human MCHR1 (Fig. 17). Non-peptidic nature of these orally active melanin-concentrating hormone receptor 1 antagonists, play a key role in anxiolytic- and antidepressant-like profile95 of ATC0065 and ATC0175.

Fig. 14:
Selected quinazoline motifs with notable PDE inhibitory efficiency

Fig. 15:
Some quinazoline motifs with antihypertensive activity

Fig. 16:
SBP values of normotensive (WKY-C, WKY-L) and hypertensive (SHR-C, SHR-L) rats. Values are means ± SEM. *p < 0.05 (WKY groups vs. SHR groups)92

Fig. 17:
Selected quinazoline motifs as MCHR1 antagonists

Fig. 18:
Selected bischalcones with cathepsin inhibitory activity

Cathepsin inhibitory activity: Bischalcones containing quinazoline-2(1H)-ones and quinazoline-2(1H)-thiones analogs were synthesized and screened for their inhibitory potential against cathepsin B and cathepsin H96. It was found that 79 showed maximum inhibition at 0.1 nM while the thione analog 80 showed maximum inhibition at 0.05 nM which was two-fold more active76 than 79. Among quinazoline derivatives, 2,6-bis(4’-(dimethylamino)benzylidene)cyclohexanone 81 was found to exhibit 100% inhibition at 0.1 nM which was four times more active than the thione counterpart 82 with maximum inhibition of 0.025 nM (Fig. 18).

Antibacterial activity: The antibacterial properties of several quinazolines was evaluated against methicillin-sensitive and methicillin-resistant Staphylococcus aureus and 83 was reported to be the most potent among the quinazoline series screened97. Quinazoline metabolite, lapatin B 84 showed moderate inhibitory activity against aqua-pathogenic bacteria Vibrio harveyi98. The efficacy of 85 against biofilm formation in S. aureus represents a promising additional mode of action99. The primary in vitro bioassay at 100 mg mL–1 against tobacco and tomato bacterial wilt revealed 86 and 87 as strong antibacterial agents using turbidimeter test100. Also, ligand 88 was reported to exhibit large zones of inhibition against bacterial strains metal while its metal complex 89 competed favourable with gentamicin standard antibiotic (MIC = 10 μg mL–1) against E. coli growth inhibition101 (Fig. 19).

Antifungal activity: The activity of compound 90 was 8-fold stronger than that of fluconazole on Candida albican102. From the result of sensitivity testing, 91 inhibited the candida growth with zone of inhibition103, Z.O.I. being 14 mm. In another study, a series of benzo[h]thiazolo[2,3-b] quinazolinone was designed and successfully evaluated for their antifungal activity104. It was noticed that 92 was the most active compound of the series with Z.O.I. of 18 mm on C. albican, A. niger and A. parasiticus. According to SAR analysis of some tetrazolo[1,5-c]quinazoline-5-thione derivatives against Candida albicans, 93 emerged as the most potent at concentration105 of 100 μg (Fig. 20).

Antiviral activity: Systematic evaluation of a series of thioquinazoline against Tobacco Mosaic Virus (TMV) led to the discovery of 94 which possessed appreciable protective activities against TMV in vivo, with 50% effective concentration (EC50) values of 138.1 μg mL–1 which was superior to that of Ribavirin (436.0 μg mL–1) standard106. The result of screening showed 95 to be the most potent against TMV at concentration 500 μg mL–1 with the curing and protection rates of 55.55 and 52.33%, respectively107, while the curative effect108 of 96 was 52%. High indicator of the antiviral activity of 97 against influenza type A H3N2 was due to insertion of carcass amine109. A series of quinazoline prepared via Schiff bases formation were screened against herpes simplex virus-1 (KOS), virus-2 (G), vaccinia virus, vescular stomatitis virus, reovirus-1 wherein 98 emerged as the most promising antiviral motif against the entire tested virus110 while compound 99 was reported to possessed antiviral activity due to induction of up-regulation111 of PR-1a and PR-5 (Fig. 21).

Antioxidant activity: Radical scavenging activities screening showed 100 to exhibit 19.2% DPPH inhibition which is about 50% of that of standard butylated hydrotoluene BHT112, while 101 was twice more active than the same standard113 at 25 g mL–1. Using reducing power assay114,115, 102 and 103 competed favourable with standard antioxidant. Other quinazoline scaffolds with highly promising antioxidant potentials116-118 are 104, 105 and 106 as shown in Fig. 22.

Antidiabetic activity: GPR119 agonist has emerged as a promising target for the treatment of type 2 diabetes. The analogues bearing azabicyclic amine substituents 107 exhibited better EC50 values than that of OEA though they appeared to be partial agonists119. Quinazolinone 108 was tested using sublethal dose level (10 mg kg–1 b.wt. day–1 for 3 weeks) and was found to have potent anti-hyperglycemic and antidiabetic properties120.

Fig. 19:
Selected quinazoline motifs with antibacterial activity

Fig. 20:
Selected quinazoline motifs with antifungal activity

Fig. 21:
Selected quinazoline motifs with antiviral activity

Fig. 22:
Selected quinazoline motifs with antioxidant activity

Fig. 23:
Selected quinazoline motifs with antidiabetic activity

The one-pot synthesis of a series of quinazolin-4-one in the presence of as green and reusable catalyst was reported and the attempted investigation of antidiabetic activity showed 109 to be the most active in this series121 (Fig. 23).

Heat shock protein (Hsp90) inhibitor: Heat shock protein (Hsp90) is a molecular chaperone that plays a role in the conformational stability, maturation and function of several proteins122. The FAXS NMR titration measurement of 110 was performed and it was considered as a suitable Hsp90 inhibitor based on its binding activity constant, Ki of 32.2 μM which corresponded to a ligand efficiency123 of 0.37. Quinazoline derivatives 111 was a promising lead with a 30,000 fold gain in activity, relative124 to 110. Other prominent Hsp90 inhibitors are 112, 113 and 114 (Fig. 24). The structural explanation of the binding study of the structurally related compounds 113 and 114 in the active site124 of Hsp90 is as shown in Fig. 25.

Antimimotic activity: A new series of pyrrolo[3,4-h]quinazolines was conveniently prepared with a broad substitution pattern. Most of these compounds showed antimitotic activity and a reduction of tubulin polymerization in a concentration-dependent manner. Compound 115 was active10 at 10 μM whereas, another study investigation led to the discovery of 5,8-disubstituted quinazolines 116 which blocked mitosis and induced apoptosis of tumor cells by inhibiting microtubule polymerization125.

Fig. 24:
Selected quinazoline motifs which serve as Hsp90 inhibitors

Fig. 25(a-b):
(a) View of compound 113 bound in the active site of Hsp90. Compound 113 (yellow carbons atoms) and key residues of the active site (green carbon atoms) are shown as sticks, ordered water molecules are shown as red spheres and hydrogen bonds as black dashed lines and (b) View of compound 114 bound in the active site of Hsp90 114 (yellow carbons atoms) and key residues of the active site (green carbon atoms) are shown as sticks, ordered water molecules are shown as red spheres and hydrogen bonds as black dashed lines124

Fig. 26:
Selected quinazoline motifs with antimimotic activity

Compound 117 was identified as potent and selective PLK1 inhibitor which was in turn reported as valuable antimitotic agent126 (Fig. 26). This is owing to PLK1 being a key component of the cell cycle control machinery with important roles in the mitotic entry, centrosome duplication, bipolar mitotic spindle formation, transition from metaphase to anaphase, cytokinesis and maintenance of genomic stability127.

Anticoccidial activity: Synthetic modification of the ring of febrifugine resulted in quinazoline 118 with noticeable anticoccidial activity against Eimeria tenella in the chicken at a dose128 of 9 mg kg–1. In another study, a series of 4-(2-methoxyphenyl)-2-oxobutylquinazoline derivatives were synthesized via facile synthetic approach and 119 emerged as the motif with highest anticoccidial activity among this series129 as shown in Fig. 27.

Fig. 27:
Selected quinazoline motifs with anticoccidial activity

Fig. 28:
Selected quinazoline motifs with antidepressant activity

Fig. 29:
Elinogrel as selected quinazoline-based antithrombotic marketed drug

Thus, 119 may serve as an open door of opportunity in future veterinary drug design due to its reported strong anticoccidial properties in the preliminary screening.

Antidepressant activity: According to the finding of a study on a series of quinazoline motif, 120 showed high antidepressant properties similar to that of the standard drug and without any neurotoxicity130. From the result of the antidepressant drug screening via forced swim pool method, quinazoline without any substituent on the phenyl ring 121, emerged as the most active while the counterpart with p-methoxy 122 was projected as a promising antidepressant agent for future drug development131. Also compound 123 was unveiled as a candidate for further study due to its improved CNS antidepressant nature132 (Fig. 28).

Antithrombosis activity: Activated factor X (FXa) is an important player in the coagulation cascade responsible for thrombin generation, which is activated during atrial fibrillation. Activated factor X FXa and tachyarrhythmia act synergistically to increase expression of protease-activated receptors and inflammatory mediators133. Some quinazoline derivative exerted inhibition of inflammation via this route. Elinogrel 118, the structure shown in Fig. 29 is a marketed available antithrombosis drug containing quinazolinone moiety134.

Structure Activity Relationship (SAR) study: Structure Activity Relationship (SAR) of the reported compounds revealed that the choice of a suitable substitution pattern including electron-donating, electron-withdrawing groups as well as some heterocyclic moieties, on the basic skeleton plays a key role in regulating the biological potential of the synthesized compounds. From the painstaking examination of the SAR, it was collectively noted that the positions 2, 4 and 6 are major points of biological relevance for improved activities for possible drug design135. Hence, 2-, 4- and 6-positions substituted quinazoline analogs remain majority among the products of this family of heterocycle and various 2,4, 6-trisubstituted quinazoline derivatives have been evaluated for bioactivity136. In addition, the substitution on the pyrimidine ring shows more of an anti-malarial activity variation of a quinazoline derivative137. Triazolo-fused quinazoline 119 with morpholinyl in 2-position and chloro on 6-position was the most potent antimicrobial scaffold among the series synthesized in a recent research endeavor138. Presence of electron donating diamine at 4-position increased the anti-inflammatory efficacy80 of 120. Introduction of polar group such as hydroxamate on the 4-position of the quinazoline core was reported to likely provide a potent HDACi/HER2i hybrid as seen in 121 which was the most potent HDAC inhibitor among the quinazolines designed and synthesized in a recent study78. The isopropyl group on 2-position of 69 was reported to enhance its PDE-7 inhibitory potential being the most active in the series screened recently85. Some structural representations of quinazoline templates for SAR study are as shown in Fig. 30.

Fig. 30:
Representative quinazoline motifs for SAR study

CONCLUSION

In conclusion, it is reported that in recent times lots of pathogens and causative agent of diseases have grown resistant abilities to the old existing and commercially available drugs, which have made the cure of these diseases harder and almost impossible. This present article provides the researchers with a pool of diagnostic information in quinazoline templates and thorough understanding of their structure activity relationship study in order to help mankind against the adverse effect of drug-resistance pathogens and newly occuring infectious diseases. This further helps in designing large number of quinazoline and quinazolinone compounds with a strong impact in curing many fatal disorders and infectious diseases. Quinazoline derivatives are thereby presented as resourceful tools in the designing of expeditious pharmacophores which may serve as great opening to new drug discovery and development in the therapeutic medicine.

SIGNIFICANT STATEMENT

Recent advances on quinazoline motifs were reviewed herein
Relevant binding and SAR studies were expatiated for information on druggability
Their diverse bioactivities and pharmacological potentials were explored
They were unveiled as arsenals of warfare against infectious diseases in therapeutics
Quinazoline cores are undeniable toolbox in public health and human total wellbeing

REFERENCES

  • Martins, P., J. Jesus, S. Santos, L.R. Raposo, C. Roma-Rodrigues, P.V. Baptista and A.R. Fernandes, 2015. Heterocyclic anticancer compounds: Recent advances and the paradigm shift towards the use of nanomedicine's tool box. Molecules, 20: 16852-16891.
    CrossRef    Direct Link    


  • Ajani, O.O., C.A. Obafemi, O.C. Nwinyi and D.A. Akinpelu, 2010. Microwave assisted synthesis and antimicrobial activity of 2-quinoxalinone-3-hydrazone derivatives. Bioorg. Med. Chem., 18: 214-221.
    CrossRef    PubMed    Direct Link    


  • Ajani, O.O. and O.C. Nwinyi, 2009. Synthesis and evaluation of antimicrobial activity of phenyl and furan-2-yl[1,2,4] triazolo[4,3-a]quinoxalin-4(5H)-one and their hydrazone precursors. Can. J. Pure Applied Sci., 3: 983-992.
    Direct Link    


  • Joule, J.A. and K. Mills, 2007. Heterocyclic Chemistry at a Glance. Blackwell Publishing, Oxford, UK., ISBN-13: 978-1405139182, Pages: 160


  • Al-Ebaisat, H.S., 2015. A review on synthesis and spectral properties of quinazolines and pyrimidines. Am. Chem. Sci. J., 6: 213-223.
    CrossRef    Direct Link    


  • Selvam, T.P. and P.V. Kumar, 2011. Quinazoline marketed drugs-a review. Res. Pharm., 1: 1-21.
    Direct Link    


  • Rajput, R. and A.P. Mishra, 2012. A review on biological activity of quinazolinones. Int. J. Pharm. Pharmaceut. Sci., 4: 66-70.
    Direct Link    


  • Ramesh, K., K. Karnakar, G. Satish, K.H.V. Reddy and Y.V.D. Nageswar, 2012. Tandem supramolecular synthesis of substituted 2-aryl-2,3-dihydroquinazolin-4(1H)-ones in the presence of β-cyclodextrin in water. Tetrahedron Lett., 53: 6095-6099.
    CrossRef    Direct Link    


  • Khan, I., A. Ibrar, N. Abbas and A. Saeed, 2014. Recent advances in the structural library of functionalized quinazoline and quinazolinone scaffolds: Synthetic approaches and multifarious applications. Eur. J. Med. Chem., 76: 193-244.
    CrossRef    Direct Link    


  • Spano, V., A. Montalbano, A. Carbone, B. Parrino and P. Diana et al., 2014. Synthesis of a new class of pyrrolo[3,4-h]quinazolines with antimitotic activity. Eur. J. Med. Chem., 74: 340-357.
    CrossRef    Direct Link    


  • Sordella, R., D.W. Bell, D.A. Haber and J. Settleman, 2004. Gefitinib-sensitizing EGFR mutations in lung cancer activate anti-apoptotic pathways. Science, 305: 1163-1167.
    CrossRef    Direct Link    


  • Raymond, E., S. Faivre and J.P. Armand, 2000. Epidermal growth factor receptor tyrosine kinase as a target for anticancer therapy. Drugs, 60: 15-23.
    CrossRef    Direct Link    


  • Mhaske, S.B. and N.P. Argade, 2006. The chemistry of recently isolated naturally occurring quinazolinone alkaloids. Tetrahedron, 62: 9787-9826.
    CrossRef    Direct Link    


  • Ajani, O.O., R.I. Ituen and A. Falomo, 2011. Facile synthesis and characterization of substituted pyrimidin-2(1H)-ones and their chalcone precursors. Pak. J. Sci. Ind. Res., 54: 59-67.
    Direct Link    


  • Asif, M., 2014. Chemical characteristics, synthetic methods and biological potential of quinazoline and quinazolinone derivatives. Int. J. Med. Chem.
    CrossRef    


  • Karnakar, K., J. Shankar, S.N. Murthy, K. Ramesh and Y.V.D. Nageswar, 2011. An efficient protocol for the synthesis of 2-phenylquinazolines catalyzed by Ceric Ammonium Nitrate (CAN). Synlett, 8: 1089-1096.
    CrossRef    Direct Link    


  • Chen, Z., J. Chen, M. Liu, J. Ding, W. Gao, X. Huang and H. Wu, 2013. Unexpected copper-catalyzed cascade synthesis of quinazoline derivatives. J. Org. Chem., 78: 11342-11348.
    CrossRef    Direct Link    


  • Chen, M., M. Zhang, B. Xiong, Z. Tan, W. Lv and H. Jiang, 2014. A novel ruthenium-catalyzed dehydrogenative synthesis of 2-arylquinazolines from 2-aminoaryl methanols and benzonitriles. Org. Lett., 16: 6028-6031.
    CrossRef    Direct Link    


  • Kabri, Y., M.D. Crozet, S. Redon and P. Vanelle, 2014. One-pot chemoselective synthesis of 2,4,6,8-tetrasubstituted quinazolines via microwave-assisted consecutive Bis-SNAr/Bis-Suzuk-Miyaura cross-coupling reactions. Synthesis, 46: 1613-1620.
    CrossRef    Direct Link    


  • Reddy, G.R., T.R. Reddy, R.G. Chary, S.C. Joseph, S. Mukherjee and M. Pal, 2013. β-Cyclodextrin mediated MCR in water: synthesis of dihydroisoindolo[2,1-a]quinazoline-5,11-dione derivatives under microwave irradiation. Tetrahedron Lett., 54: 6744-6746.
    CrossRef    Direct Link    


  • Han, B., X.L. Yang, C. Wang, Y.W. Bai, T.C. Pan, X. Chen and W. Yu, 2012. CuCl/DABCO/4-HO-TEMPO-catalyzed aerobic oxidative synthesis of 2-substituted quinazolines and 4H-3,1-benzoxazines. J. Org. Chem., 77: 1136-1142.
    CrossRef    Direct Link    


  • Jiang, X., T. Tang, J.M. Wang, Z. Chen, Y.M. Zhu and S.J. Ji, 2014. Palladium-catalyzed one-pot synthesis of quinazolinones via tert-butyl isocyanide insertion. J. Org. Chem., 79: 5082-5087.
    CrossRef    Direct Link    


  • Wang, Y., H. Wang, J. Peng and Q. Zhu, 2011. Palladium-catalyzed intramolecular C(sp2)-H amidination by isonitrile insertion provides direct access to 4-aminoquinazolines from N-arylamidines. Org. Lett., 13: 4604-4607.
    CrossRef    Direct Link    


  • Yang, X., H. Liu, H. Fu, R. Qiao, Y. Jiang and Y. Zhao, 2010. Efficient copper-catalyzed synthesis of 4-aminoquinazoline and 2,4-diaminoquinazoline derivatives. Synlett, 1: 101-106.
    CrossRef    Direct Link    


  • Navale, V., R. Shinde, S. Patil, A. Vibhute and S. Zangade, 2016. Quinazoline based synthesis of some novel heterocyclic Schiff bases. J. Adv. Chem. Sci., 2: 201-203.
    Direct Link    


  • Ajani, O.O., J.T. Isaac, T.F. Owoeye and A.A. Akinsiku, 2015. Exploration of the chemistry and biological properties of pyrimidine as a privilege pharmacophore in therapeutics. Int. J. Biol. Chem., 9: 148-177.
    CrossRef    Direct Link    


  • Alanazi, A.M., A.A.M. Abdel-Aziz, I.A. Al-Suwaidan, S.G. Abdel-Hamide, T.Z. Shawer and A.S. El-Azab, 2014. Design, synthesis and biological evaluation of some novel substituted quinazolines as antitumor agents. Eur. J. Med. Chem., 79: 446-454.
    CrossRef    Direct Link    


  • Yassen, A.S.A., H.E.A.E.A. Elshihawy, M.M.A. Said and K.A.M. Abouzid, 2014. Molecular modelling and synthesis of quinazoline-based compounds as potential antiproliferative agents. Chem. Pharmaceut. Bull., 62: 454-466.
    CrossRef    Direct Link    


  • Pathania, D., M. Sechi, M. Palomba, V. Sanna and F. Berrettini et al., 2014. Design and discovery of novel quinazolinedione-based redox modulators as therapies for pancreatic cancer. Biochimica Biophysica Acta (BBA)-Gen. Subj., 1840: 332-343.
    CrossRef    Direct Link    


  • Gawad, N.M.A., H.H. Georgey, R.M. Youssef and N.A. El-Sayed, 2010. Synthesis and antitumor activity of some 2,3-disubstituted quinazolin-4(3H)-ones and 4,6-disubstituted-1,2,3,4-tetrahydroquinazolin-2H-ones. Eur. J. Med. Chem., 45: 6058-6067.
    CrossRef    Direct Link    


  • Li, S., X. Wang, Y. He, M. Zhao and Y. Chen et al., 2013. Design and synthesis of novel quinazoline nitrogen mustard derivatives as potential therapeutic agents for cancer. Eur. J. Med. Chem., 67: 293-301.
    CrossRef    Direct Link    


  • Choi, D.L., S.J. Jang, S. Cho, H.E. Choi, H.K. Rim, K.T. Lee and J.Y. Lee, 2014. Inhibition of cellular proliferation and induction of apoptosis in human lung adenocarcinoma A549 cells by T-type calcium channel antagonist. Bioorg. Med. Chem. Lett., 24: 1565-1570.
    CrossRef    Direct Link    


  • Hynes, N.E. and H.A. Lane, 2005. ERBB receptors and cancer: The complexity of targeted inhibitors. Nat. Rev. Cancer, 5: 341-354.
    CrossRef    Direct Link    


  • Tan, F., X. Shen, D. Wang, G. Xie and X. Zhang et al., 2012. Icotinib (BPI-2009H), a novel EGFR tyrosine kinase inhibitor, displays potent efficacy in preclinical studies. Lung Cancer, 76: 177-182.
    CrossRef    Direct Link    


  • Minkovsky, N. and A. Berezov, 2008. BIBW-2992, a dual receptor tyrosine kinase inhibitor for the treatment of solid tumors. Curr. Opin. Invest. Drug, 9: 1336-1346.
    PubMed    


  • Zhang, X., T. Peng, X. Ji, J. Li and L. Tong et al., 2013. Design, synthesis and biological evaluation of novel 4-anilinoquinazolines with C-6 urea-linked side chains as inhibitors of the epidermal growth factor receptor. Bioorg. Med. Chem., 21: 7988-7998.
    CrossRef    Direct Link    


  • De Castro Barbosa, M.L., L.M. Lima, R. Tesch, C.M.R. Sant'Anna and F. Totzke et al., 2014. Novel 2-chloro-4-anilino-quinazoline derivatives as EGFR and VEGFR-2 dual inhibitors. Eur. J. Med. Chem., 71: 1-14.
    CrossRef    Direct Link    


  • Wu, L., C. Zhang and W. Li, 2014. Synthesis and antiproliferative evaluation of 13-aryl-13H-benzo[g]benzothiazolo [2,3-b]quinazoline-5,14-diones. Bioorg. Med. Chem. Lett., 24: 1462-1465.
    CrossRef    Direct Link    


  • Mowafy, S., N.A. Farag and K.A.M. Abouzid, 2013. Design, synthesis and in vitro anti-proliferative activity of 4,6-quinazolinediamines as potent EGFR-TK inhibitors. Eur. J. Med. Chem., 61: 132-145.
    CrossRef    Direct Link    


  • Cao, S.L., Y. Han, C.Z. Yuan, Y. Wang and Z.K. Xiahou et al., 2013. Synthesis and antiproliferative activity of 4-substituted-piperazine-1-carbodithioate derivatives of 2,4-diaminoquinazoline. Eur. J. Med. Chem., 64: 401-409.
    CrossRef    Direct Link    


  • Mathew, B., L. Ross and R.C. Reynolds, 2013. A novel quinoline derivative that inhibits mycobacterial FtsZ. Tuberculosis, 93: 398-400.
    CrossRef    Direct Link    


  • Sirisoma, N., A. Pervin, H. Zhang, S. Jiang and J.A. Willardsen et al., 2010. Discovery of N-methyl-4-(4-methoxyanilino)quinazolines as potent apoptosis inducers: Structure-activity relationship of the quinazoline ring. Bioorganic Med. Chem. Lett., 20: 2330-2334.
    CrossRef    Direct Link    


  • Pyta, K., K. Klich, J. Domagalska and P. Przybylski, 2014. Structure and evaluation of antibacterial and antitubercular properties of new basic and heterocyclic 3-formylrifamycin SV derivatives obtained via 'click chemistry' approach. Eur. J. Med. Chem., 84: 651-676.
    CrossRef    Direct Link    


  • Maurya, H.K., R. Verma, S. Alam, S. Pandey and V. Pathak 2013. Studies on substituted benzo[h]quinazolines, benzo[g]indazoles, pyrazoles, 2,6-diarylpyridines as anti-tubercular agents. Bioorg. Med. Chem. Lett., 23: 5844-5849.
    CrossRef    Direct Link    


  • Rudolph, J., W.P. Esler, S. O'Connor, P.D.G. Coish and P.L. Wickens et al., 2007. Quinazolinone derivatives as orally available ghrelin receptor antagonists for the treatment of diabetes and obesity. J. Med. Chem., 50: 5202-5216.
    CrossRef    Direct Link    


  • Iwashita, A., S. Yamazaki, K. Mihara, K. Hattori and H. Yamamoto et al., 2004. Neuroprotective effects of a novel poly(ADP-ribose) polymerase-1 inhibitor, 2-{3-[4-(4-chlorophenyl)-1-piperazinyl]propyl}-4(3H)-quinazolinone (FR255595), in an in vitro model of cell death and in mouse 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine model of Parkinson's disease. J. Pharmacol. Exp. Therapeut., 309: 1067-1078.
    CrossRef    Direct Link    


  • Tamaoki, S., Y. Yamauchi, Y. Nakano, S. Sakano, A. Asagarasu and M. Sato, 2007. Pharmacological properties of 3-amino-5,6,7,8-tetrahydro-2-{4-[4-(quinolin-2-yl) piperazin-1-yl] butyl} quinazolin-4 (3H)-one (TZB-30878), a novel therapeutic agent for diarrhea-predominant irritable bowel syndrome (IBS) and its effects on an experimental IBS model. J. Pharmacol. Exp. Ther., 322: 1315-1323.
    CrossRef    Direct Link    


  • Hu, J., Y. Zhang, L. Dong, Z. Wang and L. Chen et al., 2015. Design, synthesis and biological evaluation of novel quinazoline derivatives as anti-inflammatory agents against lipopolysaccharide-induced acute lung injury in rats. Chem. Biol. Drug Des., 85: 672-684.
    CrossRef    Direct Link    


  • Rakesh, K.P., H.M. Manukumar and D.C. Gowda, 2015. Schiff's bases of quinazolinone derivatives: Synthesis and SAR studies of a novel series of potential anti-inflammatory and antioxidants. Bioorg. Med. Chem. Lett., 25: 1072-1077.
    CrossRef    Direct Link    


  • Nallasivan, P.K., B. Jayakar and N. Gopal, 2015. Anti-inflammatory activity of novel pyrazoline containing quinazolinone derivatives. Asian J. Pharm. Anal. Med. Chem., 3: 31-38.
    Direct Link    


  • Serya, R.A.T., A.H. Abbas, N.S.M. Ismail, A. Esmat and D.A.A. El Ella, 2015. Design, synthesis and biological evaluation of novel quinazoline-based anti-inflammatory agents acting as PDE4B inhibitors. Chem. Pharm. Bull., 63: 102-116.
    CrossRef    Direct Link    


  • Sangshetti, J.N., F.A.K. Khan, A.A. Kulkarni, R. Arote and R.H. Patil, 2015. Antileishmanial drug discovery: Comprehensive review of the last 10 years. RSC. Adv., 5: 32376-32415.
    CrossRef    Direct Link    


  • Kumar, S., N. Shakya, S. Gupta, J. Sarkar and D.P. Sahu, 2009. Synthesis and biological evaluation of novel 4-(hetero) aryl-2-piperazino quinazolines as anti-leishmanial and anti-proliferative agents. Bioorg. Med. Chem. Lett., 19: 2542-2545.
    CrossRef    Direct Link    


  • Van Horn, K.S., X. Zhu, T. Pandharkar, S. Yang and B. Vesely et al., 2014. Antileishmanial activity of a series of N2, N4-disubstituted quinazoline-2,4-diamines. J. Med. Chem., 57: 5141-5156.
    CrossRef    Direct Link    


  • Zhu, X., K.S. Van Horn, M.M. Barber, S. Yang, M.Z. Wang, R. Manetsch and K.A. Werbovetz, 2015. SAR refinement of antileishmanial N2,N4-disubstituted quinazoline-2,4-diamines. Bioorg. Med. Chem., 23: 5182-5189.
    CrossRef    Direct Link    


  • Mendoza-Martinez, C., N. Galindo-Sevilla, J. Correa-Basurto, V.M. Ugalde-Saldivar and R.G. Rodriguez-Delgado et al., 2015. Antileishmanial activity of quinazoline derivatives: Synthesis, docking screens, molecular dynamic simulations and electrochemical studies. Eur. J. Med. Chem., 92: 314-331.
    CrossRef    Direct Link    


  • Sharma, M., K. Chauhan, R. Shivahare, P. Vishwakarma and M.K. Suthar et al., 2013. Discovery of a new class of natural product-inspired quinazolinone hybrid as potent antileishmanial agents. J. Med. Chem., 56: 4374-4392.
    CrossRef    Direct Link    


  • Ajani, O.O., 2014. Present status of quinoxaline motifs: Excellent pathfinders in therapeutic medicine. Eur. J. Med. Chem., 85: 688-715.
    CrossRef    Direct Link    


  • Al-Suwaidan, I.A., A.M. Alanazi, A.A.M. Abdel-Aziz, M.A. Mohamed and A.S. El-Azab, 2013. Design, synthesis and biological evaluation of 2-mercapto-3-phenethylquinazoline bearing anilide fragments as potential antitumor agents: Molecular docking study. Bioorg. Med. Chem. Lett., 23: 3935-3941.
    CrossRef    Direct Link    


  • El-Subbagh, H.I., G.S. Hassan, S.M. El-Messery, S.T. Al-Rashood, F.A.M. Al-Omary, Y.S. Abulfadl and M.I. Shabayek, 2014. Nonclassical antifolates, part 5. Benzodiazepine analogs as a new class of DHFR inhibitors: Synthesis, antitumor testing and molecular modeling study. Eur. J. Med. Chem., 74: 234-245.
    CrossRef    Direct Link    


  • Palop, J.A., D. Plano, E. Moreno and C. Sanmartin, 2014. Novel quinazoline and pyrido [2,3-d]pyrimidine derivatives and their hydroselenite salts as antitumoral agents. ARKIVOC, 2: 187-206.
    Direct Link    


  • Luo, H., S. Yang, Y. Cai, Z. Peng and T. Liu, 2014. Synthesis and biological evaluation of novel 6-chloro-quinazolin derivatives as potential antitumor agents. Eur. J. Med. Chem., 84: 746-752.
    CrossRef    Direct Link    


  • Cai, J., M. Sun, X. Wu, J. Chen, P. Wang, X. Zong and M. Ji, 2013. Design and synthesis of novel 4-benzothiazole amino quinazolines Dasatinib derivatives as potential anti-tumor agents. Eur. J. Med. Chem., 63: 702-712.
    CrossRef    Direct Link    


  • Lim, C.J., K.S. Oh, J.D. Ha, J.H. Lee and H.W. Seo et al., 2014. 4-Substituted quinazoline derivatives as novel EphA2 receptor tyrosine kinase inhibitors. Bioorg. Med. Chem. Lett., 24: 4080-4083.
    CrossRef    Direct Link    


  • Abnous, K., B. Barati, S. Mehri, M.R.M. Farimani and M. Alibolandi et al., 2013. Synthesis and molecular modeling of six novel monastrol analogues: Evaluation of cytotoxicity and kinesin inhibitory activity against HeLa cell line. DARU J. Pharm. Sci., Vol. 21.
    CrossRef    


  • Conconi, M.T., G. Marzaro, L. Urbani, I. Zanusso and R. Di Liddo et al., 2013. Quinazoline-based multi-tyrosine kinase inhibitors: Synthesis, modeling, antitumor and antiangiogenic properties. Eur. J. Med. Chem., 67: 373-383.
    CrossRef    Direct Link    


  • Bursavich, M.G., D. Dastrup, M. Shenderovich, K.M. Yager, D.M. Cimbora, B. Williams and D.V. Kumar, 2013. Novel Mps1 kinase inhibitors: From purine to pyrrolopyrimidine and quinazoline leads. Bioorg. Med. Chem. Lett., 23: 6829-6833.
    CrossRef    Direct Link    


  • Kashaw, S.K., V. Kashaw, P. Mishra, N.K. Jain and J.P. Stables, 2009. Synthesis, anticonvulsant and CNS depressant activity of some new bioactive 1-(4-substituted-phenyl)-3-(4-oxo-2-phenyl/ethyl-4H-quinazolin-3-yl)-urea. Eur. J. Med. Chem., 44: 4335-4343.
    CrossRef    Direct Link    


  • El-Azab, A.S., S.G. Abdel-Hamide, M.M. Sayed-Ahmed, G.S. Hassan and T.M. El-Hadiyah et al., 2013. Novel 4(3H)-quinazolinone analogs: Synthesis and anticonvulsant activity. Med. Chem. Res., 22: 2815-2827.
    CrossRef    Direct Link    


  • El-Azab, A.S. and K.E.H. ElTahir, 2012. Design, synthesis and anticonvulsant evaluation of novel 8-substituted-4(3H)-quinazolines. Med. Chem. Res., 21: 3785-3796.
    CrossRef    Direct Link    


  • Kadi, A.A., A.S. El-Azab, A.M. Alafeefy and S.G. Abdel-Hamide, 2009. Synthesis and biological screening of some new substituted 2-Mercapto-4-(3H)-quinazolinone analogs as anticonvulsant agents. Arizona J. Pharm. Sci., 34: 147-167.
    Direct Link    


  • Rojas-Aguirre, Y., F. Hernandez-Luis, C. Mendoza-Martinez, C.P. Sotomayor and L.F. Aguilar et al., 2012. Effects of an antimalarial quinazoline derivative on human erythrocytes and on cell membrane molecular models. Biochimica Biophysica Acta (BBA)-Biomembr., 1818: 738-746.
    CrossRef    Direct Link    


  • Sundriyal, S., N.A. Malmquist, J. Caron, S. Blundell and F. Liu et al., 2014. Development of diaminoquinazoline histone lysine methyltransferase inhibitors as potent blood-stage antimalarial compounds. ChemMedChem, 9: 2360-2373.
    CrossRef    Direct Link    


  • Bule, M.H., A. Haymete and B. Kefale, 2015. Synthesis and in-vivo pharmacological evaluation of some novel 4(3H)-quinazolinone derivatives as potential anti-malarial agents. Drug Des., Vol. 4.
    CrossRef    


  • Mishra, M., V.K. Mishra, P. Senger, A.K. Pathak and S.K. Kashaw, 2014. Exploring QSAR studies on 4-substituted quinazoline derivatives as antimalarial compounds for the development of predictive models. Med. Chem. Res., 23: 1397-1405.
    CrossRef    Direct Link    


  • Jantschi, L. and S. Bolboaca, 2006. Molecular descriptors family on structure activity relationships 5. Antimalarial activity of 2,4-diamino-6-quinazoline sulfonamide derivates. Leonardo J. Sci., 8: 77-88.
    Direct Link    


  • Guan, J., Q. Zhang, M. O'Neil, N. Obaldia III, A. Ager, L. Gerena and A.J. Lin, 2005. Antimalarial activities of new pyrrolo[3,2-f]quinazoline-1,3-diamine derivatives. Antimicrob. Agents Chemother., 49: 4928-4933.
    CrossRef    Direct Link    


  • Zhang, X., M. Su, Y. Chen, J. Li and W. Lu, 2013. The design and synthesis of a new class of RTK/HDAC dual-targeted inhibitors. Molecules, 18: 6491-6503.
    CrossRef    Direct Link    


  • Kramer, O.H., 2009. HDAC2: A critical factor in health and disease. Trends Pharmacol. Sci., 30: 647-655.
    CrossRef    Direct Link    


  • Lin, Z., P.M. Murray, Y. Ding, W.A. Denny and L.R. Ferguson, 2010. Quinazolines as novel anti-inflammatory histone deacetylase inhibitors. Mutat. Res./Fundam. Mol. Mech. Mutagen., 690: 81-88.
    CrossRef    Direct Link    


  • Vijayakumar, K., A.J. Ahamed and G. Thiruneelakandan, 2013. Synthesis, antimicrobial and anti-HIV1 activity of quinazoline-4 (3H)-one derivatives. J. Applied Chem.
    CrossRef    


  • Selvam, P., D. Sridharan, N. Murugesh, M. Chandramohan and M. Witurouw, 2008. Synthesis and anti-HIV activity of some novel 2,3-disubstituted quinazolin-4(3H)-ones. Int. J. Chem. Sci., 6: 1196-1201.
    Direct Link    


  • Mohamed, Y.A., A.E.G.E. Amr, S.F. Mohamed, M.M. Abdalla, M.A. Al-Omar and S.H. Shfik, 2012. Cytotoxicity and anti-HIV evaluations of some new synthesized quinazoline and thioxopyrimidine derivatives using 4-(thiophen-2-yl)-3,4,5,6-tetrahydrobenzo[h]quinazoline-2(1H)-thione as synthon. J. Chem. Sci., 124: 693-702.
    CrossRef    Direct Link    


  • Omori, K. and J. Kotera, 2007. Overview of PDEs and their regulation. Circ. Res., 100: 309-327.
    CrossRef    Direct Link    


  • Sanchez, A.I., V. Martinez-Barrasa, C. Burgos, J.J. Vaquero, J. Alvarez-Builla, E. Terricabras and V. Segarra, 2013. Synthesis and evaluation of quinazoline derivatives as phosphodiesterase 7 inhibitors. Bioorg. Med. Chem., 21: 2370-2378.
    CrossRef    Direct Link    


  • Humphrey, J.M., E. Yang, C.W. am Ende, E.P. Arnold and J.L. Head et al., 2014. Small-molecule phosphodiesterase probes: Discovery of potent and selective CNS-penetrable quinazoline inhibitors of PDE1. MedChemComm, 5: 1290-1296.
    CrossRef    Direct Link    


  • Redondo, M., J.G. Zarruk, P. Ceballos, D.I. Perez and C. Perez et al., 2012. Neuroprotective efficacy of quinazoline type phosphodiesterase 7 inhibitors in cellular cultures and experimental stroke model. Eur. J. Med. Chem., 47: 175-185.
    CrossRef    Direct Link    


  • Rahman, M.U., A. Rathore, A.A. Siddiqui, G. Parveen and M. Shahar Yar, 2014. Synthesis and antihypertensive screening of new derivatives of quinazolines linked with isoxazole. BioMed Res. Int.
    CrossRef    


  • Alagarsamy, V. and U.S. Pathak, 2007. Synthesis and antihypertensive activity of novel 3-benzyl-2-substituted-3H-[1,2,4]triazolo[5,1-b]quinazolin-9-ones. Bioorg. Med. Chem., 15: 3457-3462.
    CrossRef    Direct Link    


  • Al-Salahi, R., K.E. Tahir, I. Alswaidan, N. Lolak, M. Hamidaddin and M. Marzouk, 2014. Biological effects of a new set 1,2,4-triazolo[1,5-a]quinazolines on heart rate and blood pressure. Chem. Cent. J., Vol. 8.
    CrossRef    


  • Bartha, E., I. Solti, L. Kereskai, J. Lantos and E. Plozer et al., 2009. PARP inhibition delays transition of hypertensive cardiopathy to heart failure in spontaneously hypertensive rats. Cardiovascular Res., 83: 501-510.
    CrossRef    Direct Link    


  • Magyar, K., L. Deres, K. Eros, K. Bruszt and L. Seress et al., 2014. A quinazoline-derivative compound with PARP inhibitory effect suppresses hypertension-induced vascular alterations in spontaneously hypertensive rats. Biochimica Biophysica Acta (BBA)-Mol. Basis Dis., 1842: 935-944.
    CrossRef    Direct Link    


  • Sasmal, S., G. Balaji, H.R.K. Reddy, D. Balasubrahmanyam and G. Srinivas et al., 2012. Design and optimization of quinazoline derivatives as Melanin Concentrating Hormone Receptor 1 (MCHR1) antagonists. Bioorg. Med. Chem. Lett., 22: 3157-3162.
    CrossRef    Direct Link    


  • Kanuma, K., K. Omodera, M. Nishiguchi, T. Funakoshi and S. Chaki et al., 2005. Lead optimization of 4-(dimethylamino)quinazolines, potent and selective antagonists for the melanin-concentrating hormone receptor 1. Bioorg. Med. Chem. Lett., 15: 3853-3856.
    CrossRef    Direct Link    


  • Chaki, S., T. Funakoshi, S. Hirota-Okuno, M. Nishiguchi and T. Shimazaki et al., 2005. Anxiolytic- and antidepressant-like profile of ATC0065 and ATC0175: Nonpeptidic and orally active melanin-concentrating hormone receptor 1 antagonists. J. Pharmacol. Exp. Therapeut., 313: 831-839.
    CrossRef    Direct Link    


  • Raghav, N. and M. Singh, 2014. Design, synthesis and docking studies of bischalcones based quinazoline-2(1H)-ones and quinazoline-2(1H)-thiones derivatives as novel inhibitors of cathepsin B and cathepsin H. Eur. J. Pharmaceut. Sci., 54: 28-39.
    CrossRef    Direct Link    


  • Parhi, A.K., Y. Zhang, K.W. Saionz, P. Pradhan and M. Kaul et al., 2013. Antibacterial activity of quinoxalines, quinazolines and 1,5-naphthyridines. Bioorg. Med. Chem. Lett., 23: 4968-4974.
    CrossRef    Direct Link    


  • Liu, Q., Y. Zhao, H. Fu and C. Cheng, 2013. Copper-catalyzed sequential N-arylation and aerobic oxidation: Synthesis of quinazoline derivatives. Synlett, 24: 2089-2094.
    CrossRef    Direct Link    


  • Yestrepsky, B.D., Y. Xu, M.E. Breen, X. Li and W.G. Rajeswaran et al., 2013. Novel inhibitors of bacterial virulence: Development of 5,6-dihydrobenzo[h]quinazolin-4(3H)-ones for the inhibition of group A streptococcal streptokinase expression. Bioorg. Med. Chem., 21: 1880-1897.
    CrossRef    Direct Link    


  • Wang, X., P. Li, Z. Li, J. Yin and M. He et al., 2013. Synthesis and bioactivity evaluation of novel arylimines containing a 3-aminoethyl-2-[(p-trifluoromethoxy)anilino]-4(3H)-quinazolinone moiety. J Agric. Food Chem., 61: 9575-9582.
    CrossRef    Direct Link    


  • Siddappa, K., S.B. Mane and D. Manikprabhu, 2014. Spectral characterization and 3D molecular modeling studies of metal complexes involving the O, N-donor environment of quinazoline-4(3H)-one Schiff base and their biological studies. Scient. World J.
    CrossRef    


  • Ji, Q., D. Yang, X. Wang, C. Chen and Q. Deng et al., 2014. Design, synthesis and evaluation of novel quinazoline-2,4-dione derivatives as chitin synthase inhibitors and antifungal agents. Bioorg. Med. Chem., 22: 3405-3413.
    CrossRef    Direct Link    


  • El-Gohary, N.S. and M.I. Shaaban, 2013. Synthesis, antimicrobial, antiquorum-sensing, antitumor and cytotoxic activities of new series of fused [1,3,4]thiadiazoles. Eur. J. Med. Chem., 63: 185-195.
    CrossRef    Direct Link    


  • Gali, R., J. Banothu, M. Porika, R. Velpula and S. Hnamte et al., 2014. Indolylmethylene benzo[h]thiazolo[2,3-b]quinazolinones: Synthesis, characterization and evaluation of anticancer and antimicrobial activities. Bioorg. Med. Chem. Lett., 24: 4239-4242.
    CrossRef    Direct Link    


  • Antypenko, L.M., S.I. Kovalenko, O.M. Antypenko, A.M. Katsev and O.M. Achkasova, 2013. Design and evaluation of novel antimicrobial and anticancer agents among tetrazolo [1,5-c]quinazoline-5-thione S-derivatives. Sci. Pharm., 81: 15-52.
    CrossRef    Direct Link    


  • Wan, Z., D. Hu, P. Li, D. Xie and X. Gan, 2015. Synthesis, antiviral bioactivity of novel 4-thioquinazoline derivatives containing chalcone moiety. Molecules, 20: 11861-11874.
    CrossRef    Direct Link    


  • Xiao, H., P. Li, D. Hu and B.A. Song, 2014. Synthesis and anti-TMV activity of novel β-amino acid ester derivatives containing quinazoline and benzothiazole moieties. Bioorg. Med. Chem. Lett., 24: 3452-3454.
    CrossRef    Direct Link    


  • Luo, H., D. Hu, J. Wu, M. He, J. Jin, S. Yang and B. Song, 2012. Rapid synthesis and antiviral activity of (quinazolin-4-ylamino)methyl-phosphonates through microwave irradiation. Int. J. Mol. Sci., 13: 6730-6746.
    CrossRef    Direct Link    


  • Nosulenko, I.S., O.Y. Voskoboynik, G.G. Berest, S.L. Safronyuk and S.I. Kovalenko et al., 2014. Synthesis and antiviral activity of [(9-R1-10-R2-3-R-2-оxo-2H-[1,2,4]triazino[2,3-с]quinazolin-6-yl)thio]acetamides derivatives with the fragments of carcass amines. J. Org. Pharm. Chem., 12: 17-27.
    Direct Link    


  • Kumar, K.S., S. Ganguly, R. Veerasamy and E. de Clercq, 2010. Synthesis, antiviral activity and cytotoxicity evaluation of Schiff bases of some 2-phenyl quinazoline-4(3)H-ones. Eur. J. Med. Chem., 45: 5474-5479.
    CrossRef    Direct Link    


  • Gao, X., X. Cai, K. Yan, B. Song, L. Gao and Z. Chen, 2007. Synthesis and antiviral bioactivities of 2-aryl- or 2-methyl-3-(substituted- benzalamino)-4(3H)-quinazolinone derivatives. Molecules, 12: 2621-2642.
    CrossRef    Direct Link    


  • Tetere, Z., D. Zicane, I. Ravina, I. Mierina and I. Rijkure, 2014. Synthesis and antioxidant properties of novel quinazoline derivatives. Mater. Sci. Applied Chem., 30: 51-54.
    CrossRef    Direct Link    


  • Zaranappa, H.M. Vagdevi, M.R. Lokesh and B.C. Gowdarshivannanavar, 2012. Synthesis and antioxidant activity of 3-substituted Schiff bases of quinazoline-2,4-diones. Int. J. ChemTech Res., 4: 1527-1533.
    Direct Link    


  • Vijayakumar, B., C.P. Kumar, G. Sivaprasad, K. Nirosha, S. Rokkam and E. Mohanambal, 2011. Evaluation of in vitro antioxidant activity of 1,2,3,4-tetrahydroquinazoline derivatives derivatives. Int. J. Novel Trends Pharmaceut. Sci., 1: 1-5.
    Direct Link    


  • Priya, M.G.R., K. Girija and N. Ravichandran, 2011. In vitro study of anti-inflammatory and antioxidant activity of 4-(3H)-quinazolinone derivatives. RASAYAN J. Chem., 4: 418-424.
    Direct Link    


  • Al-Omar, M.A., A.S. El-Azab, H.A. El-Obeid and S.G.A. Hamide, 2006. Synthesis of some new 4-(3H)-quinazoline analogs as potential antioxidant agents. J. Saudi Chem. Soc., 10: 113-128.
    Direct Link    


  • Saravanan, G., V. Alagarsamy and C.R. Prakash, 2010. Synthesis and evaluation of antioxidant activities of novel quinazolinone derivatives. Int. J. Pharm. Pharmaceut. Sci., 2: 83-86.
    Direct Link    


  • Rajasekaran, S., G. Rao, P.P.N. Sanjay and G.S. Sodhi, 2010. Synthesis, antibacterial and in vitro antioxidant activity of 2,3-substituted quinazolin-4 (3H)-ones. J. Chem. Pharmaceut. Res., 2: 482-488.
    Direct Link    


  • Pham, T.A.N., Z. Yang, Y. Fang, J. Luo, J. Lee and H. Park, 2013. Synthesis and biological evaluation of novel 2,4-disubstituted quinazoline analogues as GPR119 agonists. Bioorg. Med. Chem., 21: 1349-1356.
    CrossRef    Direct Link    


  • Ahmed, O.M., A.M. Hussein and R.R. Ahmed, 2012. Antidiabetic and antioxidant effects of newly synthesized pyrimido[1,6-a]pyrimidine derivatives in neonatal streptozotocin-induced diabetic rats. Med. Chem., 2: 20-28.
    CrossRef    


  • Dongamanti, A., R. Gadiparthi, R. Redamala, J. Anireddy, N. Burri and J. Vantikommu, 2012. Synthesis, anti-bacterial, anti-asthmatic and anti-diabetic activities of novel 3-substituted quinazolin-4-ones using 1-Butyl-3-methyl-imidazoliumtetrafluoro borate [BMIM+][BF4-] as a green, efficient and reusable catalyst under solvent free conditions. J. Chem. Pharmaceut. Res., 4: 3991-4000.
    Direct Link    


  • Neckers, L. and P. Workman, 2012. Hsp90 molecular chaperone inhibitors: Are we there yet? Clin. Cancer Res., 8: 64-76.
    CrossRef    Direct Link    


  • Hopkins, A.L., G.M. Keseru, P.D. Leeson, D.C. Rees and C.H. Reynolds, 2014. The role of ligand efficiency metrics in drug discovery. Nat. Rev. Drug Discovery, 13: 105-121.
    CrossRef    Direct Link    


  • Casale, E., N. Amboldi, M.G. Brasca, D. Caronni and N. Colombo et al., 2014. Fragment-based hit discovery and structure-based optimization of aminotriazoloquinazolines as novel Hsp90 inhibitors. Bioorg. Med. Chem., 22: 4135-4150.
    CrossRef    Direct Link    


  • Tian, W., L. Qin, Q. Song, L. He and M. Ai et al., 2010. A novel synthetic analog of 5, 8-disubstituted quinazolines blocks mitosis and induces apoptosis of tumor cells by inhibiting microtubule polymerization. PLoS One, Vol. 5.
    CrossRef    


  • Beria, I., R.T. Bossi, M.G. Brasca, M. Caruso and W. Ceccarelli et al., 2011. NMS-P937, a 4,5-dihydro-1H-pyrazolo[4,3-h]quinazoline derivative as potent and selective Polo-like kinase 1 inhibitor. Bioorg. Med. Chem. Lett., 21: 2969-2974.
    CrossRef    Direct Link    


  • Garland, L.L., C. Taylor, D.L. Pilkington, J.L. Cohen and D.D. von Hoff, 2006. A phase I pharmacokinetic study of HMN-214, a novel oral stilbene derivative with polo-like kinase-1-interacting properties, in patients with advanced solid tumors. Clin. Cancer Res., 12: 5182-5189.
    CrossRef    PubMed    Direct Link    


  • Ye, C., J. You, X.F. Li, R. You, Y. Weng, J. Li and Y. Wang, 2010. Design, synthesis and anticoccidial activity of a series of 3-(2-(2-methoxyphenyl)-2-oxoethyl) quinazolinone derivatives. Pestic. Biochem. Physiol., 97: 194-198.
    CrossRef    Direct Link    


  • You, J., C. Ye, Y. Weng, X. Mo and Y. Wang, 2008. Synthesis and anticoccidial activity of 4-(2-methoxyphenyl)-2-oxobutylquinazolinone derivatives. Arkivoc, 17: 1-11.
    Direct Link    


  • Jatav, V., P. Mishra, S. Kashaw and J.P. Stables, 2008. CNS depressant and anticonvulsant activities of some novel 3-[5-substituted 1,3,4-thiadiazole-2-yl]-2-styryl quinazoline-4(3H)-ones. Eur. J. Med. Chem., 43: 1945-1954.
    CrossRef    Direct Link    


  • Jatav, V., P. Mishra, S. Kashaw and J.P. Stables, 2008. Synthesis and CNS depressant activity of some novel 3-[5-substituted 1,3,4-thiadiazole-2-yl]-2-styryl quinazoline-4(3H)-ones. Eur. J. Med. Chem., 43: 135-141.
    CrossRef    Direct Link    


  • Chioua, R., F. Benabdelouahab, M. Chioua, R. Martinez-Alvarez and A.H. Fernandez, 2002. Synthesis of novel quinazoline derivatives via pyrimidine ortho-quinodimethane. Molecules, 7: 507-510.
    CrossRef    Direct Link    


  • Bukowska, A., I. Zacharias, S. Weinert, K. Skopp, C. Hartmann, C. Huth and A. Goette, 2013. Coagulation factor Xa induces an inflammatory signalling by activation of protease-activated receptors in human atrial tissue. Eur. J. Pharmacol., 718: 114-123.
    CrossRef    Direct Link    


  • Abida, P.N. and M. Arpanarana, 2011. An updated review: Newer quinazoline derivatives under clinical trial. Int. J. Pharmaceut. Biol. Arch., 2: 1651-1657.


  • Wang, D. and F. Gao, 2013. Quinazoline derivatives: Synthesis and bioactivities. Chem. Central J., Vol. 7.
    CrossRef    


  • Chandrika, P.M., T. Yakaiah, B. Narsaiah, V. Sridhar and G. Venugopal et al., 2009. Synthesis leading to novel 2,4,6-trisubstituted quinazoline derivatives, their antibacterial and cytotoxic activity against THP-1, HL-60 and A375 cell lines. Indian J. Chem., 48B: 840-847.
    Direct Link    


  • Khan, I., A. Ibrar, W. Ahmed and A. Saeed, 2015. Synthetic approaches, functionalization and therapeutic potential of quinazoline and quinazolinone skeletons: The advances continue. Eur. J. Med. Chem., 90: 124-169.
    CrossRef    Direct Link    


  • Jantova, S., R. Ovadekova, S. Letasiova, K. Spirkova and S. Stankovsky, 2005. Antimicrobial activity of some substituted triazoloquinazolines. Folia Microbiologica, 50: 90-94.
    CrossRef    Direct Link    

  • © Science Alert. All Rights Reserved