Pharmacophore an International Research Journal
Pharmacophore
Submit Manuscript
Open Access | Published: 2022 - Issue 4

ANTICANCER BIOLOGICAL PROFILE OF SOME HETEROCYLIC MOIETIES-THIADIAZOLE, BENZIMIDAZOLE, QUINAZOLINE, AND PYRIMIDINE

Parin Salim Sidat1,2*, Tasneem Mohamadbin Kasim Jaber2, Shwetang Ramesh Vekariya2, Azmin Mahervan Mogal2, Aarifa Mustak Patel2, Malleshappa Noolvi2

 

  1. Pharmacy Branch, Gujarat Technological University, Ahmedabad, Gujarat, India.
  2. Department of Pharmaceutical Chemistry, Shree Dhanvantary Pharmacy College, Kim-394110, Gujarat, India.

ABSTRACT

Several five and six-membered aromatic systems with three heteroatoms such as S, O, and N have been intensively researched due to their intriguing pharmacological properties. Heterocyclic compounds are chemicals that allow life to exist. Aside from that, all heteroatoms in the ring interact better with amino acids, and these interactions aid in reducing transactivation, and enhancing lipophilicity, solubility, and absorption, all of which can be exploited to improve therapeutic action. Heterocyclic nucleus 1,3,4-thiadiazole, benzimidazole, quinazoline, and pyrimidine derivatives have shown considerable biological actions such as anticancer, antimicrobial, anti-inflammatory, antidepressant, antioxidant, antifungal, antimicrobial, Carbonic anhydrase inhibitors, anticonvulsant, antibacterial activity, etc. All ring system members play a significant role in the development of novel drugs. In this review, we have highlighted the FDA approved drugs of heterocyclic compound which will pave the way for development of new entities. The current analysis focuses on synthetic derivatives of thiadiazole, benzimidazole, quinazoline, and pyrimidine that have significant anticancer biological effects on several cancer cell lines.

Keywords: Thiadiaozle, Benzimidazole, Quinazoline, Pyrimidine, Anticancer activity


Introduction

Cancer is a disease characterized by genetic or epigenetic alterations in somatic cells that result in uncontrolled cell growth and spread to other regions of the body. They are one type of neoplasm. The uncontrolled proliferation of cells in a group known as neoplasm or tumor, forms a lump or mass and might be scattered diffusely [1, 2]. Over the last two decades, we have made tremendous progress in our understanding of cancer at the molecular level. This insight has revealed a plethora of interesting new targets for the creation of effective medicines, some of which are already in clinical use [3]. Resistance to existing medications is rapidly becoming a serious global issue. One of the most important topics of research today is the development of novel chemicals to combat resistance [4]. Because of their activity in a variety of diseases, heterocyclic compounds are regarded as one of the most important types of organic chemicals employed in a variety of biological domains. The heterocyclic ring is found in many biological compounds, including DNA and RNA, chlorophyll, hemoglobin, vitamins, and many others [5]. Heterocyclic compounds are cyclic compounds that contain oxygen, nitrogen, and sulfur in the ring surrounding the carbon atom. In this review, we discussed the role of various heterocyclic rings as anti-cancer drugs, such as thiadiazole, benzimidazole, quinazoline, and pyrimidine. Thiadiazole is a heterocyclic molecule with a five-membered aromatic ring with two nitrogen and one sulfur atom. Thiadiazole is a versatile moiety with diverse biological functions [6]. The thiadiazole moiety performs tasks such as the "two-electron donor system" and "hydrogen binding domain." It also functions as a limited pharmacophore [5]. Thiadiazole and its derivatives are known as 1, 3, and 4-thiadiazole (A five-membered ring that has two nitrogen and one additional heteroatom). Thiadiazole can serve as a bio-isosteric substitute for the thiazole molecule [7]. As a result, it works similarly to third and fourth-generation cephalosporins and can thus be utilized in antibiotic formulations. There are four isomeric forms of thiadiazole: 1,2,3-thiadiazole, 1,2,5-thiadiazole, 1,2,4-thiadiazole, and 1,3,4-thiadiazole. Because of their diverse biological activities, 1,3,4-thiadiazole are crucial [8]. Compounds with the nucleus 1,3,4-thiadiazole, in particular, are recognized to exhibit distinct antibacterial properties [9] and anti-inflammatory activities [10]. Other intriguing functions of other substituted thiadiazole moieties have been discovered, such as analgesic activity, anti-microbial, anti-tubercular, anti-depressant, anti-oxidant, anti-convulsant, and anti-fungal. Another heterocyclic ring such as benzimidazole, a benzene-fused heterocyclic molecule, has received considerable attention in the field of modern medicinal chemistry because of its biological application [11]. Benzimidazole, the benzo derivative of imidazole, is a bicyclic aromatic chemical molecule composed of a six-membered benzene ring fused to a five-membered imidazole ring at the 4- and 5-positions [12]. Extensive biological profile and synthetic applicability in medicinal chemistry, the benzimidazole heterocyclic nucleus is known as the "Master Key." Because of the fused nitrogen nuclei, benzimidazoles are structural isosteres of nucleobases that readily engage with biomolecular targets and elicit several biological actions such as anti-cancer activity, anti-inflammatory, anti-ulcer, anti-fungal, and anti-bacterial. Quinazoline nitrogen-bearing aromatic heterocycle compound made up of fused two six-membered rings- benzene and pyrimidine which is prepared by Gabriel in 1903. The quinazoline ring can occur in four isomeric forms depending on the position of the nitrogen atoms: quinazoline, quinoxaline, cinnolines, and pthalazines [13]. Quinazoline synthesis methods are divided into five categories based on whether they are traditional or innovative, including Aza-reaction, Microwave-assisted reaction, Metal-catalyzed reaction, Ultrasound-promoted reaction, and Phase-transfer catalysis [14]. Fused ring compounds have been the focus of medicinal chemists due to their attractive biological and pharmacological applications. Though quinazolines were first recognized for their anti-malarial activity [15], later investigators have found various activities of quinazoline derivatives, such as an anti-cancer, anti-inflammation, anti-microbial, anti-viral, anti-hypertensive, etc. Pyrimidine is a heterocyclic molecule with six members and two nitrogen atoms in positions 1 and 3. Many pyrimidines have been extracted from nucleic acid hydrolyses, including the nucleotides uracil, thiamine, cytosine, etc. [16]. pyrimidine is a colorless compound with a molecular formula of C4H4N2 and has a melting point of 22.5℃ and a boiling point of 124℃. Pyrimidines are synthesized through condensation processes between three carbon molecules and compounds having amidine structures in the presence of a catalyst such as sodium hydroxide or sodium ethoxide [17]. Pyrimidine and its derivatives are notable for their amazing biological activity due to the presence of pyrimidine bases in the DNA and RNA building blocks [18]. Many natural products and biologically active chemicals contain heterocyclic compounds containing nitrogen and/or Sulphur, including pyrimidines [19]. Pyrimidine possesses biological activities like anti-microbial, anti-inflammatory, anti-fungal, anti-viral, anti-cancer, anti-malarial, anti-tubercular, anti-HIV, etc. Based on these investigations of heterocyclic rings such as thiadiazole, benzimidazole, quinazoline and pyrimidines can provide a very good basis for the development of new hits. In Table 1 despite the numerous marketed dugs of thiadiazole, benzimidazole, quinazoline and pyrimidine with their particular targeted therapy in cancer. The review will be beneficial in upcoming research for synthesizing and developing more promising synthetic approaches.

 

Anti-Cancerous Biological Activity of Heterocyclic Moieties

  • Thiadiazole

Guan et al. discovered that a set of 1,3,4-thiadiazole-based hydroxamic acids were potent HDAC inhibitors. Some of them demonstrated strong growth suppression in some tumor cell lines as well as good inhibitory action in the HDAC enzyme assay. When compared to SAHA (IC50 = 0.15 lM), compound 1 (IC50 = 0.089 lM) had a stronger inhibitory effect [20].

Matysiak et al. investigated the antiproliferative effects of numerous N-substituted 2-amino-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazole derivatives. In the panel substitution, alkyl, aryl, and morpholinoalkyl derivatives were utilized. The elemental, IR, 1H, 13C, and MS spectra of compounds were used to determine their structures. Four human cell lines were tested for cytotoxicity in vitro: SW707 (rectal), HCV29T (bladder), A549 (lung), and T47D (breast). Phenyl derivatives had a substantially lower effect than alkyl and morpholinoalkyl derivatives. The chemical with the strongest antiproliferative action was 2-(2,4-dichlorophenylamino)-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazole, whose ID50 was two times lower (SW707, T47D) than that of the control compound, cisplatin, which was tested in comparison [21].

Kumar et al. discovered a class of 5-(3-indolyl)-2-substituted-1,3,4-thiadiazole derivatives and tested their cytotoxicity against six human cancer cell lines. The most effective substance for inhibiting the proliferation of cancer cells is indolyl-1,3,4-thiadiazole, among them compound 3 which also contains 5-bromo indolyl and 4-benzyloxy-3-methoxyphenyl substituents (IC50 1.5 mM, PaCa2) [22].

The 2,5-disubstituted 1,3,4-thiadiazole compounds were formed by Polkam et al. and tested for in vitro antimycobacterial activity against Mycobacterium smegmatis MC-155. By using the MTT colorimetric technique, these substances have also been examined for their cytotoxic activity against the cancer cell lines HT-29 and MDA-MB-231. By using spectral analysis, including 1HNMR, 13CNMR, FT-IR, mass, and HRMS, the compounds may be identified well. According to screening results, compounds 5g, 7a, and 9 showed potential cytotoxic activity against the tested cell lines and have good antitubercular activity with MIC values of 65.74 and 40.86, respectively. Among the studied series, compound 4 stands out as a powerful antimycobacterial and anticancer drug. Additionally, the above substances were examined in HEK293T human normal cells and are [23].

Luo et al. synthesized a variety of novel 1,3,4-thiadiazole-containing benzisoselenazolone derivatives by condensation of 2-chloroselenobenzoyl chloride and 2-amino-5-substituted-1,3,4-thiadiazole derivatives. In vitro antiproliferative activity in SSMC-7721, MCF-7, and A-549 cells was assessed. The findings indicate that compounds 5, 6, and 7 have effective antiproliferative activity in several tumor cells [24].

From 5-substituted-1,3,4-thiadiazol-2-amine, Noolvi et al. created some novel 2,6-disubstituted imidazo[2,1-b][1,3,4]thiadiazole compounds. The newly synthesized compounds were tested for a single dose in vitro primary cytotoxicity by the National Cancer Institute. The two tested compounds 8 (107166/760239) were automatically scheduled for testing against the full panel of 60 human tumor cell lines at a minimum of five concentrations at 10-fold dilutions after passing the requirements for activity in this assay. 3-(2-(4-methoxyphenyl)imidazo[2,1-b] [1,3,4]thiadiazol-6-yl) aniline (compound 8) significantly reduced the growth of the Non-Small Cell Lung Cancer HOP-92 cell line and the Renal Cancer CAKI-1 cell line in vitro (GI50: 0.114 mM) (GI50: 0.743 mM) [25].    

Some novel 2,6-dimethyl-N-substituted phenylmethylene-imidazo compounds were made by Terzioglu et al. [2,1-b] [1,3,4] From 2,6-dimethylimidazo-[2,1-b], thiadiazole-5-carbohydrazides derivatives are produced. [1,3,4] thiadiazole-5-carbohydrazide. The 3-cell line, one dose in vitro primary cytotoxicity assay used by the National Cancer Institute was used to assess the newly synthesized compounds. Cell stability or growth was determined using the Sulforhodamine B (SRB) protein assay. The best cytotoxicity was demonstrated by 2,6-Dimethyl-N-(2-hydroxyphenylmethylidene) imidazo[2,1-b] [1,3,4] thiadiazole-5-carbohydrazide (compound 9). In vitro screening of 60 human tumor cell lines by the National Cancer Institute revealed that this substance had the strongest impact on an ovarian cancer cell line (OVCAR log10 GI50 value: 5.51) [26].

Yang et al. developed and synthesized a variety of cinnamic acyl 1,3,4-thiadiazole amide derivatives and their biological activities were assessed as prospective tubulin polymerization and antiproliferation inhibitors. Compound 10 inhibited the growth of the MCF-7 and A549 cell lines, having the greatest in vitro activity of all the compounds, with IC50 values of 0.28 and 0.52 g/mL, respectively [27].

 

Figure 1. Derivatives of Thiadiazole

  • Benzimidazole

Wang et al. synthesized and assessed the antitumor efficacy of benzimidazole derivatives. With IC50 values of 3.95 M, compound 11 demonstrated the strongest anti-proliferative action against MFC cells [28].

Designed and produced as tubulin inhibitors with strong antiproliferative action by Ren et al. Compound 12 efficiently overcame paclitaxel resistance in vitro by showing virtually equivalent efficacy against a paclitaxel-resistant cancer cell line with an IC50 value of 9.7 nM and the matching parental cell line (IC50 = 6.2 nM) [29].

New series of benzimidazole-connected pyrazole compounds was designed by Akhtar et al. concerning lung cancer cell lines, compound 13 demonstrated the strongest activity (IC50 = 2.2 M) and EGFR binding (IC50 = 0.97 M) of the group. It also caused cell cycle arrest in G2 / M phase via inducing apoptosis [30].

Huang et al. synthesized a benzimidazole molecule as an anticancer drug, and they tested it for cytotoxicity against the human carcinoma cell lines A-549, BFTC-905, RD, MES-SA, and HELA with an IC50 value of 2.8 M, Compound 14 is more effective against human lung (A-549) and HeLa cell lines than UK-1 [31].

Yadav et al. designed and evaluated benzimidazole derivatives concerning in vitro anticancer activity. Among them, compound 15, the most effective benzimidazole derivative was created. Isocitrate lyase, pantothenate synthetase, and chorismite mutase were all inhibited to varying degrees (67.56%, 53.45%, 753, and 47.56%) [32].

Marri et al. found in vitro anticancer efficacy of benzimdazolyl 2-amino-1,3,4-oxadiazole derivatives against HeLa, MCF7, A549, and HEK293 cell lines with IC50 values of 6.07, 0.028, 5.30, 0.09, 7.16, 0.061 and 7.56, 0.073, 7.20, 0.048, and 11.30, 0.018 M against the HeLa, MCF-7, and A549 cell lines, respectively, compounds 16a and 16b have also demonstrated good anticancer activity. It also causes less harm to HEK-293 cells. The molecular docking results of the synthesized compounds with the EGFR protein target [33].

Woo et al. designed and evaluate some derivatives of benzimdazolyl curcumin imitators. Compound 17 has a potent inhibitory effect on the development of MCF-7 cancer cells in this study, with an IC50 of 1.9 M [34].

Ulviye et al. synthesized new benzimidazole-triazolothiadiazine compounds to operate as aromatase inhibitors with anticancer action. This compound 18, with an IC50 of 0.032 to 0.042 M compared to letrozole's IC50 of 0.024 to 0.001 M, had slightly less effective aromatase inhibitory action [35].

New series of benzimidazole compounds were synthesized and evaluated by Saglik et al. for the suppression of aromatase inhibitors. The 4-benzylpiperidine derivatives compound 19, with IC50 values of 0.024, 0.001 M compared to the reference medication cisplatin (IC50 = 0.021, 0.001 M), were the most effective compounds in this series for the MCF-7 cell line [36].

Ulviye et al. formulate and assess novel hydrazone-modified benzimidazole compounds as anticancer drugs. In this compound 20, the IC50 for MCF-7 inhibition was 0.0316 M. Due to its stronger anticancer properties and the effect of substituents on cytotoxic activity [37].

Figure 2. Derivatives of Benzimidazole

  • Quinazoline

New quinazoline compounds were created by Abuelizz et al. as anticancer medications. By reacting 2- amino-5-methyl benzoic acid with butyl isothiocyanate, two new 2-thioxoquinazolin-4-one molecules were synthesized. These substances were tested in vitro against the HeLa and MDA-MB231 cancer cell lines. compound 24, 25 and 26 may be potential anticancer agents, with IC50 values of compounds 24 (IC50 = 1.85 µM), 25 (IC50 = 2.5 µM) and 26 (IC50 = 2.6 µM) the HeLa and MDA-MB231 cell line [38].

Faraj et al. synthesized and tested quinazoline Schiff bases 1 and 2 against the MCF7 human breast cancer cell line for anticancer activity. After 72 hours of treatment, compounds 27 and 28 displayed substantial antiproliferative activity, with IC50 values of 6.246 106 mol/L and 5.910 106 mol/L, respectively [39].

Syed et al. developed and analysed structurally modified aryl quinazoline-isoxazole derivatives. The MTT assay was used to investigate these compounds for anticancer uses against four human cancer cell lines MCF-7 (breast cancer). Among them, compounds 29a, 29b, 29c, 29d and 29j exhibited more potent activities, with IC50 values of compound 29a (IC50 = 1.92 ± 0.85 µM), 29b (IC50 = 1.47 ± 0.51 µM), 29c (IC50 = 0.01 ± 0.008 µM), 29d (IC50 = 2.08 ± 0.77 µM) 29j (IC50 = 0.083 ± 0.001 µM) [40].

Dhunmati et al. synthesized and tested the in-vitro anticancer activity of quinazoline derivatives on MCF 7 (Breast cancer) cell lines via the MTT assay at various concentration levels. In the investigations of the study, nearly 11 compounds were synthesized out of which two compounds such as 30a and 30b showed anti-cancer activity. The compound 30a has shown an inhibitory action on breast cancer cell lines in the range of 51.9% at the concentration of 62.5 (µg/ml). compounds 30b has shown an inhibitory action on breast cancer cell line in the range of 50% at the concentration of 62.5 (µg/ml) [41].

Yong et al. designed isoxazole-moiety-containing quinazoline derivatives for preliminary anticancer efficacy against MCF-7 cell lines utilizing the MTT technique. Among them, most compounds showed good to excellent anticancer activity, especially 31a, 31b, 31c, and 31d exhibited the more potent anticancer activity against MCF-7 cell lines. IC50 values of compound 31a (IC50 = 42.82±0.1324µM), 31b (IC50 = 0.11±0.0381 µM), 31c (IC50 = 1.99×10-4±0.0189 µM), 31d (IC50 = 5.74±0.00861 µM) [42].

Madhavi et al. created and synthesized a new class of chalcone-incorporated quinazoline derivatives. All of the chemicals created were tested for anticancer activity against four human cancer cell lines HT-29. Among them, four compounds, 32a, 32b, 32c and 32d showed more potent anticancer activity than the control drug, Combretastatin – A4, with IC50 values of compound 32a (IC50 = 0.18 µM), 32b (IC50 = 0.13 µM), 32c (IC50 = 1.56 µM), 32d (IC50 = 2.89 µM) [43].

Akgun et al. Some 6,7-disubstituted-3-2-[4-(substituted) piperazin-1-yl] derivatives were synthesized and tested. 2-oxoethyl quinazoline in vitro activity of 2,4(1H,3H)-dione derivatives against MCF-7 human cancer cell lines. The cytotoxicity screening findings show that 3-2-[4-(4-chlorobenzyl)piperazin-1-yl] 33 -2-oxoethyl quinazoline-2,4(1H,3H)-dione The maximum activity was seen against the MCF-7 cell line, with an IC50 value of 6.8 M [44].

Fröhlich et al. developed and synthesized five new quinazoline-artemisinin hybrids for in vitro anticancer activities against leukemia cells (CCRF-CEM and CEM/ADR5000). The antileukemia action of Hybrid 34 was similar to that of artesunic acid, with EC50 values in the low micromolar range, and it was 45 times more active against the multidrug-resistant CEM/ADR5000 cells (EC50 = 0.5 M) than the conventional medication doxorubicin [45].

Sharma et al., Novel quinazolinone compounds were synthesized by reacting N-benzoyl substituted piperazine-1-carbothioamide with 2-chloromethyl quinazolinone derivatives and investigated for anticancer efficacy on MCF 7 (Breast cancer cell) using the MTT technique. Among them, compound 35 (IC50 = 0.16 ± 0.16 µM) was found to be the most active compared to standard methotrexate (IC50 = 2.20 ± 0.18 µM) and 5-fluorouracil (IC50 = 2.30 ± 0.49 µM) [46].                                    

 

Figure 3. Derivatives of Quinazoline

  • Pyrimidine

Emami et al. created and synthesized two new anticancer quinazolinone-pyrimidine and benzyl-pyrimidine hybrids. The molecule's cytotoxic properties were also tested against three malignant cell lines (HT-29, SW1116, and A549). When compared to a lung cancer cell line, almost all of the drugs showed greater antiproliferative action on colon cancer cell lines (HT-29 and SW1116). (A549). In which compounds 36 and 37 showed excellent inhibitory activities against the HT-29 cell line with an IC50 of 10.67±0.3 μM and 27.9±6.5 μM [47].

Masoud et al. synthesized 5-(2hydroxyphenylide) barbituric acid (L1), 5-( phenyl azo) thiobarbituric acid (L2), and 5-(phenyl azo) barbituric acid (L3) and its complexes with Os(VIII), Ru(III), Zr(IV) and V(III) ions as anticancer agents. In which compound 5-( phenyl azo) thiobarbituric acid (L2) 38 was good inhibitory activity against the MCF-7 cancer cell line with an IC50 of 22 ± 0.9 (µg/ml) [19].

Gabera et al. developed a new series of 1H-pyrazolo[3,4-d]pyrimidine derivatives for cancer treatment and investigated their inhibitory capabilities against the epidermal growth factor receptor (EGFR). compound 39 was further tested for its antiproliferative activities against three cancer cell lines bearing EGFRWT (MCF-7, HepG2, A549) with an IC50 of 0.50 μM, 0.01 μM, 0.62 μM, respectively, and two cancer cell lines bearing EGFRT790M (H1975 and HCC827) with an IC50 of 0.04 μM and 0.12 μM [48].

Kumar et al. designed and synthesized of novel series of pyrimidine bridged derivatives that were examined against breast cancer (MCF-7) and lung cancer (A549) cell lines using MTT assays. From this series, compounds 40 and 41 were found most potent in the series with IC50 values of 4.67 μM & 3.38 μM and 4.63 μM & 3.71 μM against MCF7 and A549 cancer cell lines, respectively [49].

Reddy et al. designed and synthesized biscoumarin-pyrimidine conjugates that were tested for anticancer efficacy in vitro. All of the compounds, particularly compound 42, demonstrated good selectivity profiles by exhibiting harmless behavior against healthy HEK293 cells and strong binding affinities with a drug carrier protein, HAS. Compound 42 inhibits HEK293 cells effectively, having an IC50 of 4.85 M [50].

El-Metwally et al. created a novel series of thieno[2,3-d] pyrimidine derivatives with IC50 values ranging from 4 to 10 M against malignant HepG2 and MCF7 cell lines. Only compound 43 increased p53 expression by 3-4 folds and decreased Topo II expression by 60% in their investigation. Furthermore, 43 demonstrated selective cytotoxicity, cell cycle arrest, and the induction of apoptosis [51].

Cherukupalli et al. designed and synthesized 4,6-disubstituted pyrazolo [3,4-6 d] pyrimidines as CDK2 inhibitors. SAR revealed that compounds containing thiopentane/thiophenethyl groups at C-6 and heteroatom-containing bicyclic moieties (benzofuran) at C-4 have higher CDK2 inhibitory action. Furthermore, compound 44, the most powerful molecule in this series, demonstrated antiproliferative activity against the cell lines K-562 (chronic myelogenous leukemia) and MCF-7 (breast adenocarcinoma) with IC50 values of 19.8 M and 18.9 M, respectively [52].

Diao et al. designed and synthesized pyrimidine-based benzothiazole derivatives as anticancer agents. Compound 46 exhibited outstanding CDK2 inhibitory activity with an IC50 value of 15.4 nM, which was nearly three times as potent as AZD5438 Compound 45 inhibited cell cycle progression and caused apoptosis in a concentration-dependent manner [53].

Ye et al. developed and produced a new anticancer 2,4-bismorpholinyl-thieno [3, 2-d] pyrimidine Compound 46 was the most effective against HCT116, PC-3, MCF-7, A549, and MDA-MB-231 cell lines, with IC50 values of 3.24 M, 14.37 M, 7.39 M, 7.10 M, and 16.85 M, respectively. This compound also inhibited the proliferation of A549 cell lines and decreased mitochondrial membrane potential. Compound 46 as the potent compound was selected for further in vitro anti-PI3Kα and anti-PI3Kβ which demonstrated 92.4% and 62.29% inhibitory activity at 1 μM [54].

M. M. Ghorab et al. novel pyrazolo pyrimidine compounds synthesized and investigated for anticancer efficacy in vitro against Ehrlich Ascite Carcinoma cell line 5-Benzyl-1-phenyl-1, 5-dihydropyrazolo (3, 4-d) pyrimidin-4-one shown intermediate compound 47 antitumor activity when compared to doxorubicin as a positive control, with IC50 values of 90g/ml [55].

Nadia S. El-S. et al. reported a new class of sulfonamide derivatives of [1,3,4]thiadiazolo[3,2-a]pyrimidines was produced and tested for anticancer activity. The synthesized compounds were examined for their anticancer actions in vitro and in vivo. Preliminary biological tests suggested that some compounds had the highest affinity to DNA, while others had modest activity. In addition, certain substances outperformed 5-fluorouracil in terms of percentage increase in the lifetime of mice implanted with Ehrlich ascites cells (positive control) [56].

 

Figure 4. Derivatives of Pyrimidine

Table 1. FDA approved drugs of Thiadiazole, Benzimidazole, Quinazoline, and Pyrimidine

Sr. No.

Marketed drug structure with Name

Manufacture Company name

Cancer types

Target Molecule

FDA approval year

Ref.

Thiadiazole

1

Acetazolamide

EMCURE PHARMA

Renal cell carcinomas

Carbonic anhydrous inhibitor

2005

[57]

2

Methazolamine

REMEDYREPACK INC.

-

-

-

[58]

3

Megazol

AKUMENTIS HEALTHCARE LTD

-

-

-

[59]

Benzimidazole

4

Bendamustine

TEVA PHARMACEUTICAL INDUSTRIES LTD.

GERMAN PRODUCT

Chronic lymphocytic leukemia

(Cancer of WBC)

β- cell

2015

[60]

5

Mebendazole

MEDIX LABORATORIES

Thyroid cancer

Tubulin protein

2016

[61]

6

Albendazole

CIPLA FROM INDIA.

ENALTEC LABS INDIA.

BEIJING INFOARK CO LTD FROM CHINA.

KA MALLE PHARMACEUTICALS FROM INDIA

Head and neck squamous cell cancer.

CDK4/6

1996

[62]

7

Abemaciclib

ELI LILLY AND COMPANY

Breast cancer

HER2

2015

[63]

8

Glasdegib

PFIZER ONCOLOGY

Acute myeloid

leukemia

hedgehog receptor inhibitor

2018

[64]

9

Pracinostat

MEI PHARMA, INC.

Tumors

Histon deacetylase inhibitor

2013

[65]

10

Crenolanib

AROG PHARMACEUTICALS,

Acute myeloid Leukemia,

Gastrointestinal Stromal Tumor

Type 1 kinase inhibitor

2017

[66]

11

Flubendazole

JANSSEN PHARMACEUTICA N.V.

Breast cancer,

prostate cancer,

colorectal cancer,

lung cancer.

P53

1952

[67]

12

Dovotinib

ALLERITY THERAPEUTICS

 

Renal cell carcinoma,

Multiple myeloma

TKI-258

2021

[68]

Quinazoline

13

ASTRAZENECA

Medullary thyroid cancer (MTC)

EGFR, VEGFR

2011

[69]

14

BOEHRINGER INGELHEIM

Non-small cell lung cancer (NSCLC)

EGFR

2013

[70]

15

GILEAD

Blood cancers

PI3K

2014

[71]

16

PFIZER

Non-small cell lung cancer (NSCLC) with EGFR mutation

EGFR, ERBB

2018

[72]

17

SEATTLE GENETICS

Advanced unresectable or metastatic HER-2 positive breast cancer

EGFR, HER2

2020

[73]

18

NOVARTIS AND GSK

Advanced or metastatic breast cancer

EGFR, ERBB

2007

[74]

19

OSI PHARMS

Non-small cell lung cancer (NSCLC), pancreatic cancer

EGFR

2004

[75]

20

ASTRAZENECA

Non-small cell lung cancer (NSCLC)

EGFR

2003

[76]

21

ASTRAZENECA

Malignant neoplasm of colon and rectum

TS

1998

[77]

22

BETTA PHARMA

Non-small cell lung cancer (NSCLC)

EGFR-TKI

2014

(SFDA)

[78]

Pyrimidine

23

Fluorouracil

BIOCHEM PHARMACEUTICAL INDUSTRIES LTD,

OM BIOTEC (ONCOMED PHARMACEUTICALS)

Colorectal Cancer, Esophageal Cancer, Pancreatic Cancer,

Breast Cancer,

Cervical Cancer

HDAC

1962

[79]

24

Cytarabine

EUTICALS FROM ITALY,

SHANDONG OCTAGON CHEMICALS LIMITED FROM CHINA,

ZHEJIANG HISUN PHARMA FROM CHINA.

Acute myeloid leukemia,

Acute lymphocytic leukemia (ALL),

non-Hodgkin’s lymphoma primary central nervous system (CNS) lymphoma

Ara-C

1969

[80]

25

Clofarabine

ABON PHARMS LLC,

ACCORD HLTHCARE,

MYLAN LABS LTD,

GLAND PHARMA LTD,

AMNEAL.

Acute lymphoblastic leukemia

T47D

2004

[80]

26

Pralatrexate

ALLOS THERAPEUTICS LTD

Treatment of relapsed

or

refractory

peripheral T-cell lymphoma (TCL)

dihydrofolate reductase (DHFR)

2009

[81]

27

Fedratinib

IMPACT BIOMEDICINES

Myelofibrosis

BRD4 and

JAK2

2019

[82]

28

Floxuridine

LGM PHARMA,

ZHEJIANG HISUN PHARMA

Cancer of gastrointestinal (GI) tract (cancer of the stomach or intestines) that has spread to the liver.

-

1970

[83]

29

Orotic acid

SHREYA PHARMACHEM PRIVATE LIMITED

Powerful tumor promoter, particularly in the liver.

Hep G2

2015

[16]

 

Conclusion

 

In medicinal chemistry, heterocyclic compounds are one of the most important forms of organic molecules, and they are applicable to treat a variety of ailments. The primary goal of research promised in this review specifies a wide range of pharmacological activities revealed in heterocyclic compounds such as thiadiazole, quinazoline, pyrimidine, and benzimidazole. The potential applications of heterocycles as anticancer, antitubercular, anti-inflammatory, antifungal, antimicrobial, antihypertensive, anti-HIV, antiviral, antidiabetic agents, etc. A heterocycle presence in a therapeutic molecule has been found as a possible candidate for continuing drug research, and our main motive behind the work presented in this paper is to help the creation and designing of new potential medicinal agents with better activities by medicinal researchers.

Acknowledgments: Authors are thankful to the Principal of Shree Dhanvantary Pharmacy College for Guidance.

Conflict of interest: None

Financial support: None

Ethics statement: None

References

1.        Saini A, Kumar M, Bhatt S, Saini V. Introduction: Cancer: Cancer is a disorder. Int J Pharm Sci Res. 2020;11:3121-34.

2.        Ames BN, Gold LS, Willett WC. The causes and prevention of cancer. Proc Natl Acad Sci U S A. 1995;92(12):5258-65.

3.        Bhatia A, Thakur A. Choice of Diversification Strategies in an Emerging Market: Evidence from Indian Manufacturing Industries. J Asia-Pacific Bus. 2017;18(1):21-45.

4.        Kempegowda SK, Kumar DP, TT M. Thiadiazoles: Progress report on biological activities. Der Pharma Chem. 2011;3(2):330-41.

5.        Almulla AF, Pharma D, Al-Mulla A. A review: biological importance of heterocyclic compounds. Der Pharma Chemica. 2017;9(13):141-7.

6.        Mishra G, Singh AK, Jyoti K. Thiadiazole derivaties and it’s Pharmacological activities. Int J Chem Tech Res. 2011;3(3):1380-93.

7.        Harayama T. Synthetic studies on aromatic heterocyclic compounds. Yakugaku Zasshi. 2006;126(8):543-64.

8.        Li Y, Geng J, Liu Y, Yu S, Zhao G. Thiadiazole-a Promising Structure in Medicinal Chemistry. Chem Med Chem. 2013;8(1):27-41.

9.        Wu Z, Shi J, Chen J, Hu D, Song B. Design, Synthesis, Antibacterial Activity, and Mechanisms of Novel 1,3,4-Thiadiazole Derivatives Containing an Amide Moiety. J Agric Food Chem. 2021;69(31):8660-70.

10.     Kadi AA, Al-Abdullah ES, Shehata IA, Habib EE, Ibrahim TM, El-Emam AA. Synthesis, antimicrobial and anti-inflammatory activities of novel 5-(1-adamantyl)-1,3,4-thiadiazole derivatives. Eur J Med Chem. 2010;45(11):5006-11. doi:10.1016/j.ejmech.2010.08.007

11.     Hashem HE, El Bakri Y. An overview on novel synthetic approaches and medicinal applications of benzimidazole compounds: An overview on novel synthetic approaches and medicinal applications. Arab J Chem. 2021;14(11):103418. doi:10.1016/j.arabjc.2021.103418

12.     Goud NS, Kumar P, Bharath RD. Recent Developments of Target-Based Benzimidazole Derivatives as Potential Anticancer Agents. Heterocycles-Synth Biol Act. 2020:1-18.

13.     Dhuguru J, Ghoneim OA. Quinazoline Based HDAC Dual Inhibitors as Potential Anti-Cancer Agents. Molecules. 2022;27(7):1-29.

14.     Wang D, Gao F. Quinazoline derivatives: Synthesis and bioactivities. Chem Cent J. 2013;7(1):1-15.

15.     Sen D, Banerjee A, Ghosh A, Chatterjee T. Synthesis and antimalarial evaluation of some 4-quinazolinone derivatives based on febrifugine. J Adv Pharm Technol Res. 2010;1(4):401-5.

16.     Crossing B, Nagar S, Potential T. A review on the synthesis and therapeutic potential of pyrimidine. Int J Pharm Sci Res. 2010;1(5):34-49.

17.     Merugu R, Garimella S, Balla D, Sambaru K. Synthesis and biological activities of pyrimidines: A review. Synthesis. 2015;8(6):88-93.

18.     Yerragunta V, Patil P, Anusha V, Kumaraswamy T, Suman D, Samhitha T. Pyrimidine and its biological activity: a review. PharmaTutor. 2013;1(2):39-44.

19.     Masoud MS, Sweyllam AM, Ahmed MM. Synthesis, characterization, coordination chemistry and biological activity of some pyrimidine complexes. J Mol Struct. 2020;1219:128612. doi:10.1016/j.molstruc.2020.128612

20.     Guan P, Sun F, Hou X, Wang F, Yi F, Xu W, et al. Design, synthesis and preliminary bioactivity studies of 1,3,4-thiadiazole hydroxamic acid derivatives as novel histone deacetylase inhibitors. Bioorg Med Chem. 2012;20(12):3865-72. doi:10.1016/j.bmc.2012.04.032

21.     Matysiak J, Opolski A. Synthesis and antiproliferative activity of N-substituted 2-amino-5-(2,4-dihydroxyphenyl)-1,3,4-thiadiazoles. Bioorg Med Chem. 2006;14(13):4483-9.

22.     Kumar D, Maruthi Kumar N, Chang KH, Shah K. Synthesis and anticancer activity of 5-(3-indolyl)-1,3,4-thiadiazoles. Eur J Med Chem. 2010;45(10):4664-8. doi:10.1016/j.ejmech.2010.07.023

23.     Polkam N, Rayam P, Anireddy JS, Yennam S, Anantaraju HS, Dharmarajan S, et al. Synthesis, in vitro anticancer and antimycobacterial evaluation of new 5-(2,5-dimethoxyphenyl)-1,3,4-thiadiazole-2-amino derivatives. Bioorg Med Chem Lett. 2015;25(7):1398-402. doi:10.1016/j.bmcl.2015.02.052

24.     Luo Z, Chen B, He S, Shi Y, Liu Y, Li C. Synthesis and antitumor-evaluation of 1,3,4-thiadiazole-containing benzisoselenazolone derivatives. Bioorg Med Chem Lett. 2012;22(9):3191-3. doi:10.1016/j.bmcl.2012.03.043

25.     Noolvi MN, Patel HM, Kamboj S, Kaur A, Mann V. 2,6-Disubstituted imidazo[2,1-b][1,3,4]thiadiazoles: Search for anticancer agents. Eur J Med Chem. 2012;56:56-69. doi:10.1016/j.ejmech.2012.08.012

26.     Terzioglu N, Gürsoy A. Synthesis and anticancer evaluation of some new hydrazone derivatives of 2,6-dimethylimidazo[2,1-b][1,3,4]thiadiazole-5-carbohydrazide. Eur J Med Chem. 2003;38(7-8):781-6.

27.     Yang XH, Wen Q, Zhao TT, Sun J, Li X, Xing M, et al. Synthesis, biological evaluation, and molecular docking studies of cinnamic acyl 1,3,4-thiadiazole amide derivatives as novel antitubulin agents. Bioorg Med Chem. 2012;20(3):1181-7.

28.     Wang Z, Deng X, Xiong S, Xiong R, Liu J, Zou L, et al. Design, synthesis and biological evaluation of chrysin benzimidazole derivatives as potential anticancer agents. Nat Prod Res. 2018;32(24):2900-9. doi:10.1080/14786419.2017.1389940

29.     Ren Y, Wang Y, Li G, Zhang Z, Ma L, Cheng B, et al. Discovery of Novel Benzimidazole and Indazole Analogues as Tubulin Polymerization Inhibitors with Potent Anticancer Activities. J Med Chem. 2021;64(8):4498-515.

30.     Akhtar MJ, Khan AA, Ali Z, Dewangan RP, Rafi M, Hassan MQ, et al. Synthesis of stable benzimidazole derivatives bearing pyrazole as anticancer and EGFR receptor inhibitors. Bioorg Chem. 2018;78:158-69. doi:10.1016/j.bioorg.2018.03.002

31.     Huang ST, Hsei IJ, Chen C. Synthesis and anticancer evaluation of bis(benzimidazoles), bis(benzoxazoles), and benzothiazoles. Bioorg Med Chem. 2006;14(17):6106-19.

32.     Yadav S, Narasimhan B, Lim SM, Ramasamy K, Vasudevan M, Shah SAA, et al. Synthesis and evaluation of antimicrobial, antitubercular and anticancer activities of benzimidazole derivatives. Egypt J Basic Appl Sci. 2018;5(1):100-9. doi:10.1016/j.ejbas.2017.11.001

33.     Katikireddy R, Marri S, Kakkerla R, Murali Krishna MPS, Gandamalla D, Reddy YN. Synthesis, Anticancer Activity and Molecular Docking Studies of Hybrid Benzimidazole-1,3,4-Oxadiazol-2-N-Alkyl/Aryl Amines. Polycycl Aromat Compd. 2021:1-15. doi:10.1080/10406638.2021.1959352

34.     Woo HB, Eom YW, Park KS, Ham J, Ahn CM, Lee S. Synthesis of substituted benzimidazolyl curcumin mimics and their anticancer activity. Bioorg Med Chem Lett. 2012;22(2):933-6. doi:10.1016/j.bmcl.2011.12.074

35.     Osmaniye D. New Benzimidazole- Triazolothiadiazine Derivatives. 2020;25:1-15. doi:10.3390/molecules25071642

36.     Sağlık BN, Şen AM, Evren AE, Çevik UA, Osmaniye D, Çavuşoğlu BK, et al. Synthesis, investigation of biological effects and in silico studies of new benzimidazole derivatives as aromatase inhibitors. Z Naturforsch. 2020;75(9-10):353-62.

37.     Çevik UA, Sağlık BN, Ardıç CM, Özkay Y, Atlı Ö. Synthesis and evaluation of new benzimidazole derivatives with hydrazone moiety as anticancer agents. Turk J Biochem. 2018;43(2):151-8.

38.     Abuelizz HA, Marzouk M, Ghabbour H, Al-Salahi R. Synthesis and anticancer activity of new quinazoline derivatives. Saudi Pharm J. 2017;25(7):1047-54. doi:10.1016/j.jsps.2017.04.022

39.     Faraj FL, Zahedifard M, Paydar M, Looi CY, Majid NA, Ali HM, et al. Synthesis, characterization, and anticancer activity of new quinazoline derivatives against MCF-7 cells. Sci World J. 2014;2014.

40.     Syed T, Asiri YI, Shaheen S, Gangarapu K. Design, synthesis and anticancer evaluation of structurally modified substituted aryl-quinazoline derivatives as anticancer agents. Synth Commun. 2021;51(18):2782-95. doi:10.1080/00397911.2021.1941113

41.     Wasfy AAF, Mohmed NA, Salman AA. Synthesis and anti-cancer properties of novel quinazoline derivatives. Int J Res Pharm Chem. 2015;5(1):34-40.

42.     Yong JP, Lu CZ, Wu X. Potential Anticancer Agents. I. Synthesis of Isoxazole Moiety Containing Quinazoline Derivatives and Preliminarily in vitro Anticancer Activity. Anticancer Agents Med Chem. 2014;15(1):131-6.

43.     Madhavi S, Sreenivasulu R, Yazala JP, Raju RR. Synthesis of chalcone incorporated quinazoline derivatives as anticancer agents. Saudi Pharm J. 2017;25(2):275-9. doi:10.1016/j.jsps.2016.06.005

44.     Akgun H, Us Yilmaz D, Cetin Atalay R, Gozen D. A Series of 2,4(1H,3H)-Quinazolinedione Derivatives: Synthesis and Biological Evaluation as Potential Anticancer Agents. Lett Drug Des Discov. 2015;13(1):64-76.

45.     Fröhlich T, Reiter C, Ibrahim MM, Beutel J, Hutterer C, Zeitträger I, et al. Synthesis of Novel Hybrids of Quinazoline and Artemisinin with High Activities against Plasmodium falciparum, Human Cytomegalovirus, and Leukemia Cells. ACS Omega. 2017;2(6):2422-31.

46.     Sharma RN, Ravani R. Synthesis and screening of 2-(2-(4-substituted piperazine-1-yl)-5- phenylthiazol-4-yl)-3-aryl quinazolinone derivatives as anticancer agents. Med Chem Res. 2013;22(6):2788-94.

47.     Emami L, Faghih Z, Sakhteman A, Rezaei Z, Faghih Z, Salehi F, et al. Design, synthesis, molecular simulation, and biological activities of novel quinazolinone-pyrimidine hybrid derivatives as dipeptidyl peptidase-4 inhibitors and anticancer agents. New J Chem. 2020;44(45):19515-31.

48.     Gaber AA, Bayoumi AH, El-morsy AM, Sherbiny FF, Mehany ABM, Eissa IH. Design, synthesis and anticancer evaluation of 1H-pyrazolo[3,4-d]pyrimidine derivatives as potent EGFR WT and EGFR T790M inhibitors and apoptosis inducers. Bioorg Chem. 2018;80:375-95. doi:10.1016/j.bioorg.2018.06.017

49.     Kumar B, Sharma P, Gupta VP, Khullar M, Singh S, Dogra N, et al. Synthesis and biological evaluation of pyrimidine bridged combretastatin derivatives as potential anticancer agents and mechanistic studies. Bioorg Chem. 2018;78:130-40. doi:10.1016/j.bioorg.2018.02.027

50.     Reddy DS, Kongot M, Singh V, Siddiquee MA, Patel R, Singhal NK, et al. Biscoumarin–pyrimidine conjugates as potent anticancer agents and binding mechanism of hit candidate with human serum albumin. Arch Pharm (Weinheim). 2021;354(1):1-15.

51.     El-Metwally SA, Khalil AK, El-Sayed WM. Design, molecular modeling and anticancer evaluation of thieno[2,3-d]pyrimidine derivatives as inhibitors of topoisomerase II. Bioorg Chem. 2020;94:103492. doi:10.1016/j.bioorg.2019.103492

52.     Cherukupalli S, Chandrasekaran B, Aleti RR, Sayyad N, Hampannavar GA, Merugu SR, et al. Synthesis of 4,6-disubstituted pyrazolo[3,4-d]pyrimidine analogues: Cyclin-dependent kinase 2 (CDK2) inhibition, molecular docking and anticancer evaluation. J Mol Struct. 2019;1176:538-51. doi:10.1016/j.molstruc.2018.08.104

53.     Diao PC, Lin WY, Jian XE, Li YH, You WW, Zhao PL. Discovery of novel pyrimidine-based benzothiazole derivatives as potent cyclin-dependent kinase 2 inhibitors with anticancer activity. Eur J Med Chem. 2019;179:196-207. doi:10.1016/j.ejmech.2019.06.055

54.     Ye T, Han Y, Wang R, Yan P, Chen S, Hou Y, et al. Design, synthesis and biological evaluation of novel 2,4-bismorpholinothieno[3,2-d]pyrimidine and 2-morpholinothieno[3,2-d]pyrimidinone derivatives as potent antitumor agents. Bioorg Chem. 2020;99:103796. doi:10.1016/j.bioorg.2020.103796

55.     Ghorab MM, Ragab FA, Alqasoumi SI, Alafeefy AM, Aboulmagd SA. Synthesis of some new pyrazolo[3,4-d]pyrimidine derivatives of expected anticancer and radioprotective activity. Eur J Med Chem. 2010;45(1):171-8. doi:10.1016/j.ejmech.2009.09.039

56.     El-Sayed NS, El-Bendary ER, El-Ashry SM, El-Kerdawy MM. Synthesis and antitumor activity of new sulfonamide derivatives of thiadiazolo[3,2-a]pyrimidines. Eur J Med Chem. 2011;46(9):3714-20. doi:10.1016/j.ejmech.2011.05.037

57.     Cazzamalli S, Figueras E, Petho L, Borbély A, Steinkühler C, Neri D, et al. In Vivo Antitumor Activity of a Novel Acetazolamide-Cryptophycin Conjugate for the Treatment of Renal Cell Carcinomas. ACS Omega. 2018;3(11):14726-31.

58.     Assaly SA El, Bakary NS El, Abdel Aal MT, El-Sayed WA, Nassar IF. Synthesis and Anticancer Activity Towards Hepg-2 and Mcf-7 of New 2-Amino-1,3,4-Thiadiazole and Their Sugar Derivatives. Univ J Pharm Res. 2022:1-8.

59.     Gupta V. Pharmacological potential of M. recutita - A review. Int J Pharm Sci Drug Res. 2010;2(1):12-6.

60.     Dennie TW, Kolesar JM. Bendamustine for the treatment of chronic lymphocytic leukemia and rituximab-refractory, indolent B-cell non-hodgkin lymphoma. Clin Ther. 2009;31:2290-311. doi:10.1016/j.clinthera.2009.11.031

61.     Al-Karmalawy AA, Khattab M. Molecular modelling of mebendazole polymorphs as a potential colchicine binding site inhibitor. New J Chem. 2020;44(33):13990-6.

62.     Zhu L, Yang Q, Hu R, Li Y, Peng Y, Liu H, et al. Novel therapeutic strategy for melanoma based on albendazole and the CDK4/6 inhibitor palbociclib. Sci Rep. 2022;12(1):1-10. doi:10.1038/s41598-022-09592-0

63.     Corona SP, Generali D. Abemaciclib: A CDK4/6 inhibitor for the treatment of HR+/HEeR2- advanced breast cancer. Drug Des Devel Ther. 2018;12:321-30.

64.     Norsworthy KJ, By K, Subramaniam S, Zhuang L, Del Valle PL, Przepiorka D, et al. FDA approval summary: Glasdegib for newly diagnosed acute myeloid leukemia. Clin Cancer Res. 2019;25(20):6021-5.

65.     Ganai SA. Histone deacetylase inhibitor pracinostat in doublet therapy: a unique strategy to improve therapeutic efficacy and to tackle herculean cancer chemoresistance. Pharm Biol. 2016;54(9):1926-35.

66.     Weisberg E, Meng C, Case AE, Sattler M, Tiv HL, Gokhale PC, et al. Comparison of effects of midostaurin, crenolanib, quizartinib, gilteritinib, sorafenib and BLU-285 on oncogenic mutants of KIT, CBL and FLT3 in haematological malignancies. Br J Haematol. 2019;187(4):488-501.

67.     Zhou X, Zou L, Chen W, Yang T, Luo J, Wu K, et al. Flubendazole, FDA-approved anthelmintic, elicits valid antitumor effects by targeting P53 and promoting ferroptosis in castration-resistant prostate cancer. Pharmacol Res. 2021;164:105305. doi:10.1016/j.phrs.2020.105305

68.     Yadav SS, Li J, Stockert JA, Herzog B, O’Connor J, Garzon-Manco L, et al. Induction of Neuroendocrine Differentiation in Prostate Cancer Cells by Dovitinib (TKI-258) and its Therapeutic Implications. Transl Oncol. 2017;10(3):357-66. doi:10.1016/j.tranon.2017.01.011

69.     Tsang VHM, Robinson BG, Learoyd DL. The safety of vandetanib for the treatment of thyroid cancer. Expert Opin Drug Saf. 2016;15(8):1107-13.

70.     Yang JCH, Wu YL, Schuler M, Sebastian M, Popat S, Yamamoto N, et al. Afatinib versus cisplatin-based chemotherapy for EGFR mutation-positive lung adenocarcinoma (LUX-Lung 3 and LUX-Lung 6): Analysis of overall survival data from two randomised, phase 3 trials. Lancet Oncol. 2015;16(2):141-51. doi:10.1016/S1470-2045(14)71173-8

71.     Schlenk RF, Kayser S. Small Molecules in Hematology. Springer. 2018;212:199-214. Available from: http://link.springer.com/10.1007/978-3-319-91439-8

72.     Shirley M. Dacomitinib: First Global Approval. Drugs. 2018;78(18):1947-53. doi:10.1007/s40265-018-1028-x

73.     Lee A. Tucatinib: First Approval. Drugs. 2020;80(10):1033-8. doi:10.1007/s40265-020-01340-w

74.     Higa GM, Abraham J. Lapatinib in the treatment of breast cancer. Expert Rev Anticancer Ther. 2007;7(9):1183-92.

75.     Cohen MH, Johnson JR, Chen YF, Sridhara R, Pazdur R. FDA drug approval summary: erlotinib (Tarceva) tablets. Oncologist. 2005;10(7):461-6.

76.     Rawluk J, Waller CF. Gefitinib. Small Mol Oncol. 2018:235-46.

77.     Xue S, Chen YX, Qin SK, Yang AZ, Wang L, Xu HJ, et al. Raltitrexed induces mitochondrial-mediated apoptosis in SGC7901 human gastric cancer cells. Mol Med Rep. 2014;10(4):1927-34.

78.     Ismail RSM, Ismail NSM, Abuserii S, Abou El Ella DA. Recent advances in 4-aminoquinazoline based scaffold derivatives targeting EGFR kinases as anticancer agents. Futur J Pharm Sci. 2016;2(1):9-19. doi:10.1016/j.fjps.2016.02.001

79.     Cavaliere A, Probst KC, Westwell AD, Slusarczyk M. Fluorinated nucleosides as an important class of anticancer and antiviral agents. Futur Med Chem. 2017;9(15):1809-33.

80.     Parker WB. Enzymology of purine and pyrimidine antimetabolites used in the treatment of cancer. Chem Rev. 2009;109(7):2880-93.

81.     Raimondi MV, Randazzo O, Franca M La, Barone G, Vignoni E, Rossi D, et al. DHFR inhibitors: Reading the past for discovering novel anticancer agents. Molecules. 2019;24(6):1-19.

82.     Sochacka-ćwikła A, Mączyński M, Regiec A. Fda-approved drugs for hematological malignancies—the last decade review. Cancers (Basel). 2022;14(1):87.

83.     Ma Y, Liu H, Mou Q, Yan D, Zhu X, Zhang C. Floxuridine-containing nucleic acid nanogels for anticancer drug delivery. Nanoscale. 2018;10(18):8367-71.

QR code:

Short Link:
Views: 118

Downloads: 36
Quick Access

Pharmacophore
ISSN: 2229-5402

Pharmacophore
© 2024 All rights reserved
Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.