Synthesis of Triazole-Substituted Quinazoline Hybrids for Anticancer Activity and a Lead Compound as the EGFR Blocker and ROS Inducer AgentClick to copy article linkArticle link copied!
- Biswadip Banerji*Biswadip Banerji*E-mail: [email protected], [email protected]. Phone: +91 33 24995709. Fax: +91 33 24735197, +91 33 24723967.Organic & Medicinal Chemistry Division and Academy of Scientific and Innovative Research (AcSIR), Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S. C. Mullick Road, Kolkata 700032, IndiaMore by Biswadip Banerji
- Kadaiahgari ChandrasekharKadaiahgari ChandrasekharOrganic & Medicinal Chemistry Division and Academy of Scientific and Innovative Research (AcSIR), Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S. C. Mullick Road, Kolkata 700032, IndiaMore by Kadaiahgari Chandrasekhar
- Kancham SreenathKancham SreenathNational Institute of Pharmaceutical Education and Research (NIPER)—Kolkata, 4, Raja S. C. Mullick Road, Kolkata 700032, IndiaMore by Kancham Sreenath
- Saheli RoySaheli RoyCancer Biology & Inflammatory Disorder, Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S. C. Mullick Road, Kolkata 700032, IndiaMore by Saheli Roy
- Sayoni NagSayoni NagCancer Biology & Inflammatory Disorder, Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S. C. Mullick Road, Kolkata 700032, IndiaMore by Sayoni Nag
- Krishna Das SahaKrishna Das SahaCancer Biology & Inflammatory Disorder, Indian Institute of Chemical Biology (CSIR-IICB), 4 Raja S. C. Mullick Road, Kolkata 700032, IndiaMore by Krishna Das Saha
Abstract
A series of triazole-substituted quinazoline hybrid compounds were designed and synthesized for anticancer activity targeting epidermal growth factor receptor (EGFR) tyrosine kinase. Most of the compounds showed moderate to good antiproliferative activity against four cancer cell lines (HepG2, HCT116, MCF-7, and PC-3). Compound 5b showed good antiproliferative activity (IC50 = 20.71 μM) against MCF-7 cell lines. Molecular docking results showed that compound 5b formed hydrogen bond with Met 769 and Lys 721 and π–sulfur interaction with Met 742 of EGFR tyrosine kinase (PDB ID: 1M17). Compound 5b decreases the expression of EGFR and p-EGFR. It also induces apoptosis through reactive oxygen species generation, followed by the change in mitochondrial membrane potential.
Introduction
Results and Discussion
Conclusions
Experimental Section
General Remarks
Synthetic Procedure
General Procedure To Synthesize Compound 2
General Procedure To Synthesize Compound 3
General Procedure To Synthesize Compound 4
General Procedure To Synthesize Compound 5
Characterization Data
6,7-Dimethoxy-N-(4-((4-phenyl-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5a)
2-Chloro-6,7-dimethoxy-N-(4-((4-phenyl-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5b)
N-(4-((4-Phenyl-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5c)
N-(4-((4-(4-Ethylphenyl)-1H-1,2,3-triazol-1-yl)methyl)phenyl)-6,7-dimethoxyquinazolin-4-amine (5d)
2-Chloro-N-(4-((4-(4-ethylphenyl)-1H-1,2,3-triazol-1-yl)methyl)phenyl)-6,7-dimethoxyquinazolin-4-amine (5e)
N-(4-((4-(4-Ethylphenyl)-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5f)
(1-(4-(6,7-Dimethoxyquinazolin-4-ylamino)benzyl)-1H-1,2,3-triazol-4-yl)methanol (5g)
(1-(4-(2-Chloro-6,7-dimethoxyquinazolin-4-ylamino)benzyl)-1H-1,2,3-triazol-4-yl)methanol (5h)
(1-(4-(Quinazolin-4-ylamino)benzyl)-1H-1,2,3-triazol-4-yl)methanol (5i)
6,7-Dimethoxy-N-(4-((4-(phenoxymethyl)-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5j)
2-Chloro-6,7-dimethoxy-N-(4-((4-(phenoxymethyl)-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5k)
N-(4-((4-(Phenoxymethyl)-1H-1,2,3-triazol-1-yl)methyl)phenyl)quinazolin-4-amine (5l)
(6,7-Dimethoxy-quinazolin-4-yl)-[4-(4-phenylaminomethyl-[1,2,3]triazol-1-ylmethyl)-phenyl]-amine (5m)
(2-Chloro-6,7-dimethoxy-quinazolin-4-yl)-[4-(4-phenylaminomethyl-[1,2,3]triazol-1-ylmethyl)-phenyl]-amine (5n)
[4-(4-Phenylaminomethyl-[1,2,3]triazol-1-ylmethyl)-phenyl]-quinazolin-4-yl-amine (5o)
(2-Chloro-quinazolin-4-yl)-[4-(4-phenyl-[1,2,3]triazol-1-ylmethyl)-phenyl]-amine (5p)
(2-Chloro-quinazolin-4-yl)-{4-[4-(4-ethyl-phenyl)-[1,2,3]triazol-1-ylmethyl]-phenyl}-amine (5q)
{1-[4-(2-Chloro-quinazolin-4-ylamino)-benzyl]-1H-[1,2,3]triazol-4-yl}-methanol (5r)
(2-Chloro-quinazolin-4-yl)-[4-(4-phenoxymethyl-[1,2,3]triazol-1-ylmethyl)-phenyl]-amine (5s)
(2-Chloro-quinazolin-4-yl)-[4-(4-phenylaminomethyl-[1,2,3]triazol-1-ylmethyl)-phenyl]-amine (5t)
General Experimental Procedures for Biological Studies
Cytotoxicity Assay
Analysis of Cellular apoptosis
Measurement of ROS
Measurement of Mitochondrial Membrane Potential (JC1 Assay)
Western Blot
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsomega.8b01960.
1H NMR and 13C NMR spectra of all compounds and crystal data of compound 5c (PDF)
Crystallographic data of compound 5c (CIF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.
Acknowledgments
The authors want to thank CSIR for funding in this project and for a senior research fellowship to K.C. We also thank Dr. Arindam Talukdar, IICB for allowing us to utilize his molecular docking felicities using Discovery studio. The authors also want to thank Mr. Sunil Kumar Killi for his critical comments during the manuscript preparations. Thanks are due to Mr. Sandip Kundu, CSIR-IICB, for recording X-ray data, Mr. E.K. Padmanaban for recording the NMR spectra, Soumik Laha for recording the IR spectra, and Sandip Chowdhury for recording the EI HRMS spectra.
References
This article references 28 other publications.
- 1Fidler, M. M.; Bray, F.; Soerjomataram, I. The global cancer burden and human development: A review. Scand. J. Public Health 2018, 46, 27– 36, DOI: 10.1177/1403494817715400Google Scholar1https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cjjslentg%253D%253D&md5=94ce8f48e941130e946974bb5d60e747The global cancer burden and human development: A reviewFidler Miranda M; Bray Freddie; Soerjomataram IsabelleScandinavian journal of public health (2018), 46 (1), 27-36 ISSN:.AIMS: This review examines the links between human development and cancer overall and for specific types of cancer, as well as cancer-related risk-factors and outcomes, such as disability and life expectancy. METHODS: To assess human development, the Human Development Index was utilized continuously and according to four levels (low, medium, high, very high), where the low and very high categories include the least and most developed countries, respectively. All studies that assessed aspects of the global cancer burden using this measure were reviewed. RESULTS: Although the present cancer incidence burden is greater in higher Human Development Index countries, a greater proportion of the global mortality burden is observed in less developed countries, with a higher mean fatality rate in the latter countries. Further, the future cancer burden is expected to disproportionally affect less developed regions; in particular, it has been estimated that low and medium Human Development Index countries will experience a 100% and 81% increase in cancer incidence from 2008 to 2030, respectively. Disparities were also observed in risk factors and average health outcomes, such as a greater number of years of life lost prematurely and fewer cancer-related gains in life expectancy observed in lower versus higher Human Development Index settings. CONCLUSIONS: From a global perspective, there remain clear disparities in the cancer burden according to national Human Development Index scores. International efforts are needed to aid countries in social and economic transition in order to efficiently plan, implement and evaluate cancer control initiatives as a means to reduce the widening gap in cancer occurrence and survival worldwide.
- 2Ciardiello, F.; Tortora, G. Epidermal growth factor receptor (EGFR) as a target in cancer therapy: understanding the role of receptor expression and other molecular determinants that could influence the response to anti-EGFR drugs. Eur. J. Cancer 2003, 39, 1348– 1354, DOI: 10.1016/s0959-8049(03)00235-1Google ScholarThere is no corresponding record for this reference.
- 3Cohen, M. H.; Johnson, J. R.; Chen, Y. F.; Sridhara, R.; Pazdur, R. FDA Drug Approval Summary: Erlotinib (Tarceva(R)) Tablets. Oncologist 2005, 10, 461– 466, DOI: 10.1634/theoncologist.10-7-461Google ScholarThere is no corresponding record for this reference.
- 4Cohen, M. H.; Williams, G. A.; Sridhara, R.; Chen, G.; Pazdur, R. FDA Drug Approval Summary: Gefitinib (ZD1839) (Iressa(R)) Tablets. Oncologist 2003, 8, 303– 306, DOI: 10.1634/theoncologist.8-4-303Google ScholarThere is no corresponding record for this reference.
- 5Tan, F.; Shi, Y.; Wang, Y.; Ding, L.; Yuan, X.; Sun, Y. Icotinib, a selective EGF receptor tyrosine kinase inhibitor, for the treatment of non-small-cell lung cancer. Future Oncol. 2015, 11, 385– 397, DOI: 10.2217/fon.14.249Google Scholar5https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXisF2ls7s%253D&md5=a19fbbc42fd06b8971bc123155d180c0Icotinib, a selective EGF receptor tyrosine kinase inhibitor, for the treatment of non-small-cell lung cancerTan, Fenlai; Shi, Yuankai; Wang, Yinxiang; Ding, Lieming; Yuan, Xiaobin; Sun, YanFuture Oncology (2015), 11 (3), 385-397CODEN: FOUNBN; ISSN:1479-6694. (Future Medicine Ltd.)A review. Advanced non-small-cell lung cancer (NSCLC) is the main cause for cancer-related mortality. Treatments for advanced NSCLC are largely palliative and a benefit plateau appears to have reached with the platinum-based chemotherapy regimens. EGF receptor (EGFR) tyrosine kinase inhibitors gefitinib, erlotinib and afatinib came up with prolonged progression-free survival and improved quality of life, esp. in EGFR-mutated patients. Icotinib is an oral selective EGFR tyrosine kinase, which was approved by China Food and Drug administration in June 2011 for treating advanced NSCLC. Its approval was based on the registered Phase III trial (ICOGEN), which showed icotinib is noninferior to gefitinib. This review will discuss the role of icotinib in NSCLC, and its potential application and ongoing investigations.
- 6Wu, P.; Nielsen, T. E.; Clausen, M. H. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol. Sci. 2015, 36, 422– 439, DOI: 10.1016/j.tips.2015.04.005Google Scholar6https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXns1KisLg%253D&md5=a6aa3dbb11e6fe3001fa5fa03a4c950aFDA-approved small-molecule kinase inhibitorsWu, Peng; Nielsen, Thomas E.; Clausen, Mads H.Trends in Pharmacological Sciences (2015), 36 (7), 422-439CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)A review. Kinases have emerged as one of the most intensively pursued targets in current pharmacol. research, esp. for cancer, due to their crit. roles in cellular signaling. To date, the US FDA has approved 28 small-mol. kinase inhibitors, half of which were approved in the past 3 years. While the clin. data of these approved mols. are widely presented and structure-activity relationship (SAR) has been reported for individual mols., an updated review that analyzes all approved mols. and summarizes current achievements and trends in the field has yet to be found. Here we present all approved small-mol. kinase inhibitors with an emphasis on binding mechanism and structural features, summarize current challenges, and discuss future directions in this field.
- 7Dungo, R. T.; Keating, G. M. Afatinib: first global approval. Drugs 2013, 73, 1503– 1515, DOI: 10.1007/s40265-013-0111-6Google Scholar7https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVWmurnM&md5=21851cbfac56e2fffcba6c731f123cffAfatinib: First Global ApprovalDungo, Rosselle T.; Keating, Gillian M.Drugs (2013), 73 (13), 1503-1515CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. Afatinib, an irreversible inhibitor of the ErbB family of tyrosine kinases, is under development with Boehringer Ingelheim for the once-daily, oral treatment of cancer. Afatinib downregulates ErbB signalling by covalently binding to epidermal growth factor receptor (EGFR), human epidermal growth factor receptor (HER) 2 and HER4, irreversibly inhibiting tyrosine kinase autophosphorylation. It also inhibits transphosphorylation of HER3. Oral afatinib (Gilotrif®) has been approved in the US for the first-line treatment of patients with metastatic non-small-cell lung cancer (NSCLC) who have tumors with EGFR exon 19 deletions or exon 21 (L858R) substitution mutations as detected by a US FDA-approved test. Afatinib has also been approved in Taiwan for the first-line treatment of patients with EGFR mutation-pos. NSCLC. In addn., the European Medicines Agency's Committee for Medicinal Products for Human Use has recommended the approval of afatinib (Giotrif) for the treatment of patients with locally advanced or metastatic NSCLC with activating EGFR mutations who are EGFR tyrosine kinase inhibitor naive. Afatinib is also under regulatory review in Canada, Japan and other Asian countries. This article summarizes the milestones in the development of afatinib, leading to this first approval in patients with metastatic NSCLC.
- 8Leung, E. L.-H.; Fan, X.-X.; Wong, M. P.; Jiang, Z.-H.; Liu, Z.-Q.; Yao, X.-J.; Lu, L.-L.; Zhou, Y.-L.; Yau, L.-F.; Tin, V. P.-C.; Liu, L. Targeting tyrosine kinase inhibitor-resistant non-small cell lung cancer by inducing epidermal growth factor receptor degradation via methionine 790 oxidation. Antioxid. Redox Signaling 2016, 24, 263– 279, DOI: 10.1089/ars.2015.6420Google Scholar8https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvV2ktrs%253D&md5=3eefc008e2ebac6a82122f3d87e849c2Targeting Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer by Inducing Epidermal Growth Factor Receptor Degradation via Methionine 790 OxidationLeung, Elaine Lai-Han; Fan, Xing-Xing; Wong, Maria Pik; Jiang, Zhi-Hong; Liu, Zhong-Qiu; Yao, Xiao-Jun; Lu, Lin-Lin; Zhou, Yan-Ling; Yau, Li-Fong; Tin, Vicky Pui-Chi; Liu, LiangAntioxidants & Redox Signaling (2016), 24 (5), 263-279CODEN: ARSIF2; ISSN:1523-0864. (Mary Ann Liebert, Inc.)Aims: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been developed to treat non-small cell lung cancer (NSCLC) patients with EGFR mutation, but TKI resistance is common. Almost half of the acquired resistance patients are due to addnl. T790M mutation on EGFR (EGFRT790M), thus overcoming TKI resistance is important. In this study, we aim to investigate the role of reactive oxygen species (ROS) in TKI resistance as well as the mol. and biol. effects of EGFRT790M after redox manipulation. Results: The basal ROS levels in EGFRT790M-contg. TKI-resistant NSCLC cell lines were substantially high. Sixty-three human lung tumors showed higher NADPH oxidase isoform 2 (NOX2) expression than normal lung tissues, which may contribute to high basal ROS in cancer and poor survival. Interestingly, only NOX3 was upregulated by sanguinarine, a pharmacol. agent to elevate ROS, and resulted in EGFR overoxidn., degrdn., and apoptosis. By contrast, such responses were lacking in EGFRWT cells. Selective EGFRT790M degrdn. was manipulated by redox imbalance between NOX3 and methionine reductase A (MsrA). Furthermore, the in vivo tumor suppression effect of sanguinarine, NOX3 upregulation, and EGFR degrdn. were confirmed. Innovation: We have found a new treatment strategy to overcome TKI resistance by selectively inducing EGFRT790M degrdn. via specific stimulation of methionine 790 (M790) oxidn. It can be achieved via manipulating redox imbalance between NOX3 and MsrA. Conclusion: Targeting EGFR by elevating ROS and redox imbalance is a potential new strategy to develop a new EGFR inhibitor for TKI-resistant patients with a wide therapeutic window between EGFRT790M and EGFRWT. Antioxid. Redox Signal. 24, 263-279.
- 9Schacher-Kaufmann, S.; Pless, M. Acute fatal liver toxicity under erlotinib. Case Rep. Oncol. 2010, 3, 182– 188, DOI: 10.1159/000315366Google Scholar9https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sbos1yqsQ%253D%253D&md5=3180e9e98456bd0eacd6dc721c7eb932Acute Fatal Liver Toxicity under ErlotinibSchacher-Kaufmann Sabina; Pless MiklosCase reports in oncology (2010), 3 (2), 182-188 ISSN:1662-6575.We describe the case of a never-smoker who received second-line erlotinib as a treatment for his non-small cell lung cancer. Within one month, acute hepatic failure developed as well as a thrombotic-thrombocytopenic microangiopathy, with fatal outcome. In patients with non-small cell lung cancer, hepatic toxicity of erlotinib is a rare but severe complication; so far three fatal cases have been reported. Patients' liver function should be assessed before starting erlotinib and special care is recommended if pretreatment bilirubin is elevated.
- 10Stathopoulos, G.; Trafalis, D.; Athanasiou, A.; Bardi, G.; Chandrinou, H. Serious hematologic complications following erlotinib treatment. Anticancer Res. 2010, 30, 973– 976Google Scholar10https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlslelu7s%253D&md5=bf3eb9980434cddda198562f4a226716Serious hematologic complications following erlotinib treatmentStathopoulos, G. P.; Trafalis, Dim; Athanasiou, A.; Bardi, G.; Chandrinou, H.Anticancer Research (2010), 30 (3), 973-976CODEN: ANTRD4; ISSN:0250-7005. (International Institute of Anticancer Research)Background: Erlotinib is an oral, small-mol. targeting therapy which inhibits epidermal growth factor tyrosine kinase receptors. Erlotinib has been administered for the treatment of advanced pancreatic cancer and non-small cell lung cancer. Patients and Methods: In the present report, unusual hematol. complications were detected after erlotinib was administered as second-line monotherapy in pretreated patients with advanced non-small cell lung cancer. Four patients pre-treated with cisplatin or its analog-based combinations, were evaluated. Erlotinib was given at a dose of 150 mg daily. In cases of intolerable adverse reactions, the dose was either reduced to 100 mg daily or treatment was interrupted for a max. of two weeks. Results: Serious hematol. toxicity (or complications) developed in these 4 patients after 4-8.5 mo of treatment. Two patients developed leukemias (AML, CML) and two, myelodysplastic syndrome. Conclusion: Whether or not these hematol. complications were related to erlotinib treatment is comprehensively discussed.
- 11Rajput, R.; Mishra, A. P. A review on biological activity of quinazolinones. Int. J. Pharm. Pharm. Sci. 2012, 4, 66– 70Google Scholar11https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xms1Cmtrg%253D&md5=253d611f1b2c3e887bf4d403d261026cA review on biological activity of quinazolinonesRajput, Rakhi; Mishra, Abhinav PrasoonInternational Journal of Pharmacy and Pharmaceutical Sciences (2012), 4 (2), 66-70CODEN: IJPPKB; ISSN:0975-1491. (International Journal of Pharmacy and Pharmaceutical Sciences)A review. Quinazolinones is considered as an important chem. synthesis of various physiol. significance and pharmacol. utility. Quinazolinones are a large class of active chem. compds. exhibiting a broad spectrum of biol. activities in animals as well as in humans. Literature studies on quinazolinones have shown that these derivs. possess a wide variety of biol. activities such as anti HIV, anticancer, antifungal, antibacterial, antimutagenic, anticoccidial, anticonvulsant, anti-inflammatory, CNS depressant, antimalarial, antioxidant, antileukemic activity, antileishmanial activity. This review focused on the various biol. activities of quinazolinones.
- 12(a) Cheng, C.-M.; Lee, Y.-J.; Wang, W.-T.; Hsu, C.-T.; Tsai, J.-S.; Wu, C.-M.; Ou, K.-L.; Yang, T.-S. Determining the binding mode and binding affinity constant of tyrosine kinase inhibitor PD153035 to DNA using optical tweezers. Biochem. Biophys. Res. Commun. 2011, 404, 297– 301, DOI: 10.1016/j.bbrc.2010.11.110Google ScholarThere is no corresponding record for this reference.(b) Selvam, T. P.; Kumar, P. V. Quinazoline marketed drugs. Res. Pharm. 2011, 1 ().Google ScholarThere is no corresponding record for this reference.
- 13Medapi, B.; Suryadevara, P.; Renuka, J.; Sridevi, J. P.; Yogeeswari, P.; Sriram, D. 4-Aminoquinoline derivatives as novel Mycobacterium tuberculosis GyrB inhibitors: structural optimization, synthesis and biological evaluation. Eur. J. Med. Chem. 2015, 103, 1– 16, DOI: 10.1016/j.ejmech.2015.06.032Google ScholarThere is no corresponding record for this reference.
- 14Christopher, E.; Bedir, E.; Dunbar, C.; Khan, I. A.; Okunji, C. O.; Schuster, B. M.; Iwu, M. M. Indoloquinazoline Alkaloids fromAraliopsis tabouensis. Helv. Chim. Acta 2003, 86, 2914– 2918, DOI: 10.1002/hlca.200390239Google ScholarThere is no corresponding record for this reference.
- 15Mendoza-Martínez, C.; Galindo-Sevilla, N.; Correa-Basurto, J.; Ugalde-Saldivar, V. M.; Rodríguez-Delgado, R. G.; Hernández-Pineda, J.; Padierna-Mota, C.; Flores-Alamo, M.; Hernández-Luis, F. Antileishmanial activity of quinazoline derivatives: Synthesis, docking screens, molecular dynamic simulations and electrochemical studies. Eur. J. Med. Chem. 2015, 92, 314– 331, DOI: 10.1016/j.ejmech.2014.12.051Google ScholarThere is no corresponding record for this reference.
- 16Antipenko, L. N.; Karpenko, A. V.; Kovalenko, S. I.; Katsev, A. M.; Komarovska-Porokhnyavets, E. Z.; Novikov, V. P. Synthesis, Cytotoxicity by Bioluminescence Inhibition, Antibacterial and Antifungal Activity of ([1,2,4]Triazolo[1,5-c]quinazolin-2-ylthio)carboxylic Acid Amides. Arch. Pharm. 2009, 342, 651– 662, DOI: 10.1002/ardp.200900077Google Scholar16https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlGgsLnM&md5=64f2172cb9615f7a320ad9fb559987cbSynthesis, Cytotoxicity by Bioluminescence Inhibition, Antibacterial and Antifungal Activity of ([1,2,4]Triazolo[1,5-c]quinazolin-2-ylthio)carboxylic Acid AmidesAntipenko, Lyudmila N.; Karpenko, Alexander V.; Kovalenko, Sergey I.; Katsev, Andrew M.; Komarovska-Porokhnyavets, Elena Z.; Novikov, Vladimir P.Archiv der Pharmazie (Weinheim, Germany) (2009), 342 (11), 651-662CODEN: ARPMAS; ISSN:0365-6233. (Wiley-VCH Verlag GmbH & Co. KGaA)We report in this work the synthesis, cytotoxicity, and antimicrobial activity of ([1,2,4]triazolo[1,5-c]quinazolin-2-ylthio)carboxylic acid amides 4-7 in connection with our previous research in the prepn. of triazoloquinazoline derivs. Due to simplicity, general availability of starting materials, and high yields, the most reliable method of synthesis appeared to be the one with N,N-carbonyldiimidazole activation stage. The chem. structures of all obtained substances were deduced from FT-IR, 1H-NMR, EI-MS, and LC-MS spectral data. The results of cytotoxicity evaluated by bioluminescence inhibition of bacterium Photobacterium leiognathi, strain Sh1 showed that compds.4.1, 4.6, and 6.1 were the most cytotoxic. Investigation of the antimicrobial and antifungal activity of amides 4-7 (concn. 5 mg/mL) was carried out by the stiff-plate agar-diffusion method. We found that the compds. possessed low (4.1, 4.7) antifungal activity against Candida tenuis and strong (4.21, 5.1, 5.9) or inefficient (4.7, 4.12, 4.16) activity against Aspergillus niger. Substances 5.1 and 5.9 slightly affected Mycobacterium luteum. Staphylococcus aureus was resistant to all obtained substances, and only the n-butyramide derivs. 7.1 and 7.5 inhibited the growth of Escherichia coli. Hence, there was no strong correlation between bioluminescence inhibition and antimicrobial activity of the investigated substances.
- 17Horne, W. S.; Yadav, M. K.; Stout, C. D.; Ghadiri, M. R. Heterocyclic Peptide Backbone Modifications in an α-Helical Coiled Coil. J. Am. Chem. Soc. 2004, 126, 15366– 15367, DOI: 10.1021/ja0450408Google Scholar17https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptlSgtrw%253D&md5=6f4573960370cdded62c2ca77ad25b04Heterocyclic Peptide Backbone Modifications in an α-Helical Coiled CoilHorne, W. Seth; Yadav, Maneesh K.; Stout, C. David; Ghadiri, M. RezaJournal of the American Chemical Society (2004), 126 (47), 15366-15367CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)In this paper, we present 1,2,3-triazole ε2-amino acids incorporated as a dipeptide surrogate at three positions in the sequence of a known α-helical coiled coil. Biophys. characterization indicates that the modified peptides retain much of the helical structure of the parent sequence, and that the thermodn. stability of the coiled coil depends on the position of the incorporation of the ε-residue. Crystal structures obtained for each peptide give insight into the chem. behavior and conformational preferences of the non-natural amino acid and show that the triazole ring can participate in the backbone hydrogen bonding of the α-helix as well as template an interhelical crossing between chains in the bundle.
- 18(a) Reddy, D. M.; Srinivas, J.; Chashoo, G.; Saxena, A. K.; Sampath Kumar, H. M. 4β-[(4-Alkyl)-1,2,3-triazol-1-yl] podophyllotoxins as anticancer compounds: Design, synthesis and biological evaluation. Eur. J. Med. Chem. 2011, 46, 1983– 1991, DOI: 10.1016/j.ejmech.2011.02.016Google ScholarThere is no corresponding record for this reference.(b) Yoon, J.; Ryu, J.-S. A rapid synthesis of lavendustin-mimetic small molecules by click fragment assembly. Bioorg. Med. Chem. Lett. 2010, 20, 3930– 3935, DOI: 10.1016/j.bmcl.2010.05.014Google Scholar18bhttps://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsVWis7w%253D&md5=e07a4f2aa1c64bef25822ef2706547bfA rapid synthesis of lavendustin-mimetic small molecules by click fragment assemblyYoon, Jieun; Ryu, Jae-SangBioorganic & Medicinal Chemistry Letters (2010), 20 (13), 3930-3935CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Lavendustin-mimetic small mols. modifying the linker -CH2-NH- with an 1,2,3-triazole ring have been synthesized via a click chem. Two pharmacophoric fragments of lavendustin were varied to investigate chem. space and the auxophoric -CH2-NH- was altered to an 1,2,3-triazole for rapid click conjugation. The small mols. were evaluated against HCT116 colon cancer and CCRF-CEM leukemia cell lines. Among the 28 analogs, 3-phenylpropyl ester I inhibited CCRF-CEM leukemia cell growth with GI50 value of 0.9 μM.
- 19Giffin, M. J.; Heaslet, H.; Brik, A.; Lin, Y.-C.; Cauvi, G.; Wong, C.-H.; McRee, D. E.; Elder, J. H.; Stout, C. D.; Torbett, B. E. A Copper(I)-Catalyzed 1,2,3-Triazole Azide–Alkyne Click Compound Is a Potent Inhibitor of a Multidrug-Resistant HIV-1 Protease Variant. J. Med. Chem. 2008, 51, 6263– 6270, DOI: 10.1021/jm800149mGoogle Scholar19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtF2lsrvK&md5=c3ca2b1ae9e3a747ed7161715575f964A Copper(I)-Catalyzed 1,2,3-Triazole Azide-Alkyne Click Compound Is a Potent Inhibitor of a Multidrug-Resistant HIV-1 Protease VariantGiffin, Michael J.; Heaslet, Holly; Brik, Ashraf; Lin, Ying-Chuan; Cauvi, Gabrielle; Wong, Chi-Huey; McRee, Duncan E.; Elder, John H.; Stout, C. David; Torbett, Bruce E.Journal of Medicinal Chemistry (2008), 51 (20), 6263-6270CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Treatment with HIV-1 protease inhibitors, a component of highly active antiretroviral therapy (HAART), often results in viral resistance. Structural and biochem. characterization of a 6X protease mutant arising from in vitro selection with compd. 1, a C2-sym. diol protease inhibitor, has been previously described. We now show that compd. 2, a copper(I)-catalyzed 1,2,3-triazole derived compd. previously shown to be potently effective against wild-type protease (IC50 = 6.0 nM), has low nanomolar activity (IC50 = 15.7 nM) against the multidrug-resistant 6X protease mutant. Compd. 2 displays similar efficacy against wild-type and 6X HIV-1 in viral replication assays. While structural studies of compd. 1 bound to wild-type and mutant proteases revealed a progressive change in binding mode in the mutants, the 1.3 Å resoln. 6X protease-compd. 2 crystal structure reveals nearly identical interactions for 2 as in the wild-type protease complex with very little change in compd. 2 or protease conformation.
- 20Patpi, S. R.; Pulipati, L.; Yogeeswari, P.; Sriram, D.; Jain, N.; Sridhar, B.; Murthy, R.; Anjana Devi, T.; Kalivendi, S. V.; Kantevari, S. Design, Synthesis, and Structure-Activity Correlations of Novel Dibenzo[b,d]furan, Dibenzo[b,d]thiophene, and N-Methylcarbazole Clubbed 1,2,3-Triazoles as Potent Inhibitors of Mycobacterium tuberculosis. J. Med. Chem. 2012, 55, 3911– 3922, DOI: 10.1021/jm300125eGoogle Scholar20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XksVymurg%253D&md5=aa0a15ae22de7006b3a367fcaf49576bDesign, Synthesis, and Structure-Activity Correlations of Novel Dibenzo[b,d]furan, Dibenzo[b,d]thiophene, and N-Methylcarbazole Clubbed 1,2,3-Triazoles as Potent Inhibitors of Mycobacterium tuberculosisPatpi, Santhosh Reddy; Pulipati, Lokesh; Yogeeswari, Perumal; Sriram, Dharmarajan; Jain, Nishant; Sridhar, Balasubramanian; Murthy, Ramalinga; T, Anjana Devi; Kalivendi, Shasi Vardhan; Kantevari, SrinivasJournal of Medicinal Chemistry (2012), 55 (8), 3911-3922CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A mol. hybridization approach is an emerging structural modification tool to design new mols. with improved pharmacophoric properties. In this study, 1,2,3-triazole-based Mycobacterium tuberculosis inhibitors and synthetic and natural product-based tricyclic (carbazole, dibenzo[b,d]furan, and dibenzo[b,d]thiophene) antimycobacterial agents were integrated in one mol. platform to prep. various novel clubbed 1,2,3-triazole hybrids using click chem. Structure-activity correlations and in vitro activity against M. tuberculosis strain H37Rv of new analogs revealed the order: dibenzo[b,d]thiophene > dibenzo[b,d]furan > 9-methyl-9H-carbazole series. Some of the most potent M. tuberculosis inhibitors with MIC = 0.78 μg/mL (∼1.9 μM) displayed a low cytotoxicity and high selectivity index (50-255) against four different human cancer cell lines. These results together provided the potential importance of mol. hybridization and the development of triazole clubbed dibenzo[b,d]thiophene-based lead candidates to treat mycobacterial infections.
- 21De Simone, R.; Chini, M. G.; Bruno, I.; Riccio, R.; Mueller, D.; Werz, O.; Bifulco, G. Structure-Based Discovery of Inhibitors of Microsomal Prostaglandin E2Synthase–1, 5-Lipoxygenase and 5-Lipoxygenase-Activating Protein: Promising Hits for the Development of New Anti-inflammatory Agents. J. Med. Chem. 2011, 54, 1565– 1575, DOI: 10.1021/jm101238dGoogle Scholar21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXitVKqtbw%253D&md5=7e4379cce59ecf39290522c8238a3647Structure-Based Discovery of Inhibitors of Microsomal Prostaglandin E2 Synthase-1, 5-Lipoxygenase and 5-Lipoxygenase-Activating Protein: Promising Hits for the Development of New Anti-inflammatory AgentsDe Simone, Rosa; Chini, Maria Giovanna; Bruno, Ines; Riccio, Raffaele; Mueller, Daniela; Werz, Oliver; Bifulco, GiuseppeJournal of Medicinal Chemistry (2011), 54 (6), 1565-1575CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Microsomal prostaglandin E2 synthase (mPGES)-1 catalyzes the transformation of PGH2 to PGE2 that is involved in several pathologies like fever, pain, and inflammatory disorders. To identify novel mPGES-1 inhibitors, we used in silico screening to rapidly direct the synthesis, based on the copper-catalyzed 3 + 2 Huisgen's reaction (click chem.), of potential inhibitors. We designed 26 new triazole-based compds. in accordance with the pocket binding requirements of human mPGES-1. Docking results, in agreement with ligand efficiency values, suggested the synthesis of 15 compds. that at least in theory were shown to be more efficient in inhibiting mPGES-1. Biol. evaluation of these selected compds. has disclosed three new potential anti-inflammatory drugs: (I) compd. 4 displaying selectivity for mPGES-1 with an IC50 value of 3.2 μM, (II) compd. 20 that dually inhibits 5-lipoxygenase and mPGES-1, and (III) compd. 7 apparently acting as 5-lipoxygenase-activating protein inhibitor (IC50 = 0.4 μM).
- 22Banerji, B.; Killi, S. K.; Katarkar, A.; Chatterjee, S.; Tangella, Y.; Prodhan, C.; Chaudhuri, K. Neo-tanshinlactone D-ring modified novel analogues induce apoptosis in human breast cancer cell via DNA damage. Bioorg. Med. Chem. 2017, 25, 202– 212, DOI: 10.1016/j.bmc.2016.10.026Google ScholarThere is no corresponding record for this reference.
- 23Viegas-Junior, C.; Barreiro, E. J.; Fraga, C. A. M. Molecular hybridization: a useful tool in the design of new drug prototypes. Bioorg. Med. Chem. 2007, 14, 1829– 1852, DOI: 10.2174/092986707781058805Google ScholarThere is no corresponding record for this reference.
- 24Stamos, J.; Sliwkowski, M. X.; Eigenbrot, C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J. Biol. Chem. 2002, 277, 46265– 46272, DOI: 10.1074/jbc.m207135200Google Scholar24https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XovFKhsL4%253D&md5=c63c60b93756bd45fb2375c56ba38941Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitorStamos, Jennifer; Sliwkowski, Mark X.; Eigenbrot, CharlesJournal of Biological Chemistry (2002), 277 (48), 46265-46272CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The crystal structure of the kinase domain from the epidermal growth factor receptor (EGFRK) including forty amino acids from the carboxyl-terminal tail has been detd. to 2.6-Å resoln., both with and without an EGFRK-specific inhibitor currently in Phase III clin. trials as an anti-cancer agent, erlotinib (OSI-774, CP-358,774, Tarceva). The EGFR family members are distinguished from all other known receptor tyrosine kinases in possessing constitutive kinase activity without a phosphorylation event within their kinase domains. Despite its lack of phosphorylation, we find that the EGFRK activation loop adopts a conformation similar to that of the phosphorylated active form of the kinase domain from the insulin receptor. Surprisingly, key residues of a putative dimerization motif lying between the EGFRK domain and carboxyl-terminal substrate docking sites are found in close contact with the kinase domain. Significant intermol. contacts involving the carboxyl-terminal tail are discussed with respect to receptor oligomerization.
- 25Towbin, H.; Staehelin, T.; Gordon, J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc. Natl. Acad. Sci. U. S. A. 1979, 76, 4350– 4354, DOI: 10.1073/pnas.76.9.4350Google Scholar25https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE1MXmtVKltLw%253D&md5=39eea3d100df7959e5d12e39e14cecbfElectrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applicationsTowbin, Harry; Staehelin, Theophil; Gordon, JulianProceedings of the National Academy of Sciences of the United States of America (1979), 76 (9), 4350-4CODEN: PNASA6; ISSN:0027-8424.A method was devised for the electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets. The method results in quant. transfer of ribosomal proteins from gels contg. urea. For Na dodecyl sulfate gels, the original band pattern was obtained with no loss of resoln., but the transfer was not quant. The method allows detection of proteins by autoradiog. and is simpler than conventional procedures. The immobilized proteins were detectable by immunol. procedures. All addnl. binding capacity on the nitrocellulose was blocked with excess protein; then, a specific antibody was bound and, finally, a 2nd antibody directed against the 1st antibody. The 2nd antibody was wither radioactively labeled or conjugated to fluorescein or to peroxidase. The specific protein was then detected by either autoradiog., under UV light, or by the peroxidase reaction product, resp. In the latter case, as little as 100 pg of protein was clearly detectable.
- 26Lambert, A. J.; Brand, M. D. Superoxide production by NADH:ubiquinone oxidoreductase (complex I) depends on the pH gradient across the mitochondrial inner membrane. Biochem. J. 2004, 382, 511– 517, DOI: 10.1042/bj20040485Google ScholarThere is no corresponding record for this reference.
- 27Dey, S. K.; Bose, D.; Hazra, A.; Naskar, S.; Nandy, A.; Munda, R. N.; Das, S.; Chatterjee, N.; Mondal, N. B.; Banerjee, S.; Saha, K. D. Cytotoxic activity and apoptosis-inducing potential of di-spiropyrrolidino and di-spiropyrrolizidino oxindole andrographolide derivatives. PLoS One 2013, 8, e58055, DOI: 10.1371/journal.pone.0058055Google Scholar27https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXktlSrsLg%253D&md5=64ae726bf948e1fc8225ad1a9ae8f516Cytotoxic activity and apoptosis-inducing potential of di-spiropyrrolidino and di-spiropyrrolizidino oxindole andrographolide derivativesDey, Sumit Kumar; Bose, Dipayan; Hazra, Abhijit; Naskar, Subhendu; Nandy, Abhishek; Munda, Rudra Narayan; Das, Subhadip; Chatterjee, Nabanita; Mondal, Nirup Bikash; Banerjee, Sukdeb; Das Saha, KrishnaPLoS One (2013), 8 (3), e58055CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Anticancer role of andrographolide is well documented. To find novel potent derivs. with improved cytotoxicity than andrographolide on cancer cells, two series of di-spiropyrrolidino- and di-spiropyrrolizidino oxindole andrographolide derivs. prepd. by cyclo-addn. of azomethine ylide along with sarcosine or proline (viz. sarcosine and proline series resp.) and substitution of different functional groups (-CH3, -OCH3 and halogens) were examd. for their cytotoxic effect on a panel of six human cancer cell lines (colorectal carcinoma HCT116 cells, pancreatic carcinoma MiaPaCa-2 cells, hepatocarcinoma HepG2 cells, cervical carcinoma HeLa cells, lung carcinoma A549 and melanoma A375 cells). Except halogen substituted derivs. of proline series (viz. CY2, CY14 and CY15 for Br, Cl and I substitution resp.), none of the other derivs. showed improved cytotoxicity than andrographolide in the cancer cell lines examd. Order of cytotoxicity of the potent compds. is CY2>CY14>CY15>andrographolide. Higher toxicity was obsd. in HCT116, MiaPaCa-2 and HepG2 cells. CY2, induced death of HCT116 (GI50 10.5), MiaPaCa-2 (GI50 11.2) and HepG2 (GI50 16.6) cells were assocd. with cell rounding, nuclear fragmentation and increased percentage of apoptotic cells, cell cycle arrest at G1 phase, ROS generation and involvement of mitochondrial pathway. Upregulation of Bax, Bad, p53, caspases-3,-9 and cleaved PARP; downregulation of Bcl-2, cytosolic NF-κB p65, PI3K and p-Akt; translocation of P53/P21, NF-κB p65 were seen in CY2 treated HCT116 cells. Thus, three halogenated di-spiropyrrolizidino oxindole derivs. of andrographolide are found to be more cytotoxic than andrographolide in some cancer cells. The most potent deriv., CY2 induced death of the cancer cells involves ROS dependent mitochondrial pathway like andrographolide.
- 28Nandy, A.; Dey, S.; Das, S.; Munda, R.; Dinda, J.; Saha, K. Gold (I) N-heterocyclic carbene complex inhibits mouse melanoma growth by p53 upregulation. Mol. Cancer 2014, 13, 57, DOI: 10.1186/1476-4598-13-57Google Scholar28https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXivFWhtbc%253D&md5=40df17b4bddf6dce687596250172e457Gold (I) N-heterocyclic carbene complex inhibits mouse melanoma growth by p53 upregulationNandy, Abhishek; Dey, Sumit Kumar; Das, Sujata; Munda, Rudra Narayan; Dinda, Joydev; Das Saha, KrishnaMolecular Cancer (2014), 13 (), 57/1-57/14, 14 pp.CODEN: MCOACG; ISSN:1476-4598. (BioMed Central Ltd.)Background: Cancer treatment using gold (I) complexes is becoming popular. In this study, a gold (I) N-heterocyclic complex designated as complex 3 was synthesized, its cytotoxicity was examd., and its anti-melanoma activity was evaluated in vitro and in vivo. Methods: Viability of cancer cells was detd. by MTT assay upon treatment with various concns. of a gold (I) N-heterocyclic carbene complex (complex 3) in a dose and time dependent manner. Mouse melanoma cells B16F10 were selected for further apoptotic studies, including flowcytometric anal. of annexin binding, cell cycle arrest, intracellular ROS generation and loss in the mitochondrial membrane potential. ELISA based assays were done for caspase activities and western blots for detg. the expression of various survival and apoptotic proteins. Immunocytol. was performed to visualize the translocation of p53 to the nucleus. B16F10 cells were inoculated into mice and post tumor formation, complex 3 was administered. Immunohistol. was performed to det. the expressions of p53, p21, NF-κB (p65 and p50), MMP-9 and VEGF. Student's t test was used for detg. statistical significance. The survival rate data were analyzed by Kaplan-Meier plots. Results: Complex 3 markedly inhibited the growth of HCT 116, HepG2, and A549, and induced apoptosis in B16F10 cells with nuclear condensation, DNA fragmentation, externalization of phosphatidylserine, activation of caspase 3 and caspase 9, PARP cleavage, downregulation of Bcl-2, upregulation of Bax, cytosolic cytochrome c elevation, ROS generation, and mitochondrial membrane potential loss indicating the involvement of an intrinsic mitochondrial death pathway. Further, upregulation of p53, p-p53 (ser 15) and p21 indicated the role of p53 in complex 3 mediated apoptosis. The complex reduced tumor size, and caused upregulation of p53 and p21 along with downregulation of NF-κB (p65 and p50), VEGF and MMP-9. These results suggest that it induced anti-melanoma effect in vitro and in vivo by modulating p53 and other apoptotic factors. Conclusions: The gold (I) N-heterocyclic carbene complex (C22H26N6AuO2PF6) designated as complex 3 induced ROS and p53 dependent apoptosis in B16F10 cells involving the mitochondrial death pathway along with suppression of melanoma tumor growth by regulating the levels of pro and anti apoptotic factors (p53, p21, NF-κB, VEGF and MMP-9).
Cited By
This article is cited by 53 publications.
- Rajubai D. Bakale, Amruta N. Bhagat, Udhav V. Mhetre, Shyam V. Londhe, Sanket S. Rathod, Prafulla B. Choudhari, Kishan P. Haval. Design, synthesis and molecular docking study of novel quinoline–triazole molecular hybrids as anticancer agents. Journal of Molecular Structure 2025, 1321 , 140072. https://doi.org/10.1016/j.molstruc.2024.140072
- Ibrahim A. Bala, Abdullah M. Asiri, Reda M. El-Shishtawy. Quinazoline derivatives and hybrids: recent structures with potent bioactivity. Medicinal Chemistry Research 2024, 33
(12)
, 2372-2419. https://doi.org/10.1007/s00044-024-03318-9
- Venkat Reddy Dodlapati, E. Ramya Sucharitha, Rambabu Palabindela, Ravikumar Kapavarapu, Sridhar Kavela, Sirassu Narsimha. Organocatalytic[3 + 2]Cycloaddition: Synthesis of Quinazoline Containing Sulfonyl 1,2,3‐Triazoles as Potent
EGFR
Targeting Anti‐Breast Cancer Agents. Journal of Heterocyclic Chemistry 2024, 61
(11)
, 1762-1776. https://doi.org/10.1002/jhet.4905
- Kotni Hari Gangadhar, Velaga Benarjee, Annapragada Ratnamala. Coumarin-Piperazine Tethered 1,2,3-Triazoles: EGFR Targeting Anti-Breast Cancer Evaluation and Molecular Docking Studies. Polycyclic Aromatic Compounds 2024, 44
(8)
, 5487-5503. https://doi.org/10.1080/10406638.2023.2265026
- Narahari Udayasree, Ramesh Babu Haridasyam, Rambabu Palabindela, Thupurani Murali Krishna, Sirassu Narsimha. One-pot synthesis, anticancer, EGFR and caspases assays of novel fused [1,2,3]triazolo-pyrrolo[2,1-b]quinazolinones. Journal of Molecular Structure 2024, 97 , 139570. https://doi.org/10.1016/j.molstruc.2024.139570
- Afaf El-Malah, Azizah M. Malebari, Ahdab N. Khayyat, Khadijah A. Mohammad, Magdy M. Gineinah, Zeinab Mahmoud. Design, synthesis, and antiproliferative activities of novel substitutedhydrazone/triazolo-linked quinazoline derivatives. Journal of Molecular Structure 2024, 1306 , 137822. https://doi.org/10.1016/j.molstruc.2024.137822
- Rabecca Jenifer Vasanthan, Sheersha Pradhan, Mohan Das Thangamuthu. Emerging Aspects of Triazole in Organic Synthesis: Exploring its
Potential as a Gelator. Current Organic Synthesis 2024, 21
(4)
, 456-512. https://doi.org/10.2174/1570179420666221010094531
- Annapurna Mamidala, Narasimha Swamy Thirukovela, Ashok Kumar Bapuram, Karthik Bokkala, Satheesh Kumar Nukala. Synthesis of Some New Barbituric Acid Linked Quinoline-1,2,3-triazole Hybrids as Dual EGFR/VEGFR-2 Inhibitors. Russian Journal of General Chemistry 2024, 94
(6)
, 1399-1411. https://doi.org/10.1134/S1070363224060173
- Acácio S. de Souza, Deborah S. Dias, Ruan C.B. Ribeiro, Dora C.S. Costa, Matheus G. de Moraes, David R. Pinho, Maria E.G. Masset, Laís M. Marins, Sandy P. Valle, Cláudio J.C. de Carvalho, Gustavo S.G. de Carvalho, Angélica Lauria N. Mello, Mauro Sola-Penna, Marcos V. Palmeira-Mello, Raissa A. Conceição, Carlos R. Rodrigues, Alessandra M.T. Souza, Luana da S.M. Forezi, Patricia Zancan, Vitor F. Ferreira, Fernando de C. da Silva. Novel naphthoquinone-1H-1,2,3-triazole hybrids: Design, synthesis and evaluation as inductors of ROS-mediated apoptosis in the MCF-7 cells. Bioorganic & Medicinal Chemistry 2024, 102 , 117671. https://doi.org/10.1016/j.bmc.2024.117671
- T. Vishnu, M. Veerabhadraiah, V. Krishna Chaitanya, M. Nagamani, M. Raghavender, P. Jalapathi. Design, synthesis and anticancer activity of 5-((2-(4-bromo/chloro benzoyl) benzofuran-5-yl) methyl)-2-((1-(substituted)-1H-1,2,3-triazol-4-yl)methoxy)benzaldehyde analogues. Molecular Diversity 2023, 27
(6)
, 2695-2713. https://doi.org/10.1007/s11030-022-10575-6
- Randhir Rai. Copper-based laboratory waste: Its application as a catalyst and recovery. Journal of Cleaner Production 2023, 432 , 139775. https://doi.org/10.1016/j.jclepro.2023.139775
- V. K. Vashistha, A. Mittal, R. Bala, R. V. S. R. Pullabhotla. Triazole Moieties as Potent Drug Molecules: Synthetic Approaches and Application as Promising Candidates as Anticancer Agents (A Review). Russian Journal of Bioorganic Chemistry 2023, 49
(S1)
, S13-S30. https://doi.org/10.1134/S1068162023080034
- Sandhya Rani Bandi, Ravikumar Kapavarapu, Rambabu Palabindela, Mohammad Azam, Kim Min, Sirassu Narsimha. Synthesis of novel pyrido[2,3-d]pyrimidine-thiazolidine-1,2,3-triazoles: Potent EGFR targeting anticancer agents. Journal of Molecular Structure 2023, 1294 , 136451. https://doi.org/10.1016/j.molstruc.2023.136451
- Bakr F. Abdel-Wahab, Benson M. Kariuki, Hanan A. Mohamed, Mohamed S. Bekheit, Hanem M. Awad, Gamal A. El-Hiti. Synthesis and anticancer activity of 3-(1-aryl-5-methyl-1H-1,2,3-triazol-4-yl)-1-phenyl-1H-pyrazole-4-carbaldehydes. Journal of Molecular Structure 2023, 1294 , 136528. https://doi.org/10.1016/j.molstruc.2023.136528
- Jasneet Kaur, Sukhmeet Kaur, Amit Anand, Nasarul Islam, Simrandeep Singh, Akshdeep Singh. An Updated Overview on the Synthesis and Anticancer Evaluation of Quinazoline Derivatives. ChemistrySelect 2023, 8
(30)
https://doi.org/10.1002/slct.202302778
- Navneet P. Mori, Priti K. Parmar, Vijay M. Khedkar, Ranjan C. Khunt. Synthesis of N-Methylene Linker Containing Phthalimide Bearing-1
H
-1,2,3-Triazole by Click Chemistry Approach: Anticancer Activity in Human Cells. Polycyclic Aromatic Compounds 2023, 43
(6)
, 5354-5374. https://doi.org/10.1080/10406638.2022.2101487
- Mohamed A. Mahmoud, Anber F. Mohammed, Ola I.A. Salem, Safwat M. Rabea, Bahaa G.M. Youssif. Design, synthesis, and antiproliferative properties of new 1,2,3-triazole-carboximidamide derivatives as dual EGFR/VEGFR-2 inhibitors. Journal of Molecular Structure 2023, 1282 , 135165. https://doi.org/10.1016/j.molstruc.2023.135165
- Ashraf A. Aly, Mohammed B. Alshammari, Akil Ahmad, Hesham A. M. Gomaa, Bahaa G. M. Youssif, Stefan Bräse, Mahmoud A. A. Ibrahim, Asmaa H. Mohamed. Design, synthesis, docking and mechanistic studies of new thiazolyl/thiazolidinylpyrimidine-2,4-dione antiproliferative agents. Arabian Journal of Chemistry 2023, 16
(4)
, 104612. https://doi.org/10.1016/j.arabjc.2023.104612
- Mohamed T.‐E. Maghraby, Ola I. A. Salem, Bahaa G. M. Youssif, Mahmoud M. Sheha. Design, synthesis, and modelling study of new 1,2,3‐triazole/chalcone hybrids with antiproliferative action as epidermal growth factor receptor inhibitors. Chemical Biology & Drug Design 2023, 101
(3)
, 749-759. https://doi.org/10.1111/cbdd.14178
- Naman Jain, Tanvi Goel, Snehal Thakar, Madhav Jadhav, Deepali Bansode. An Explicative Review on the Progress of Quinazoline Scaffold as
Bioactive Agents in the Past Decade. Medicinal Chemistry 2023, 19
(3)
, 211-245. https://doi.org/10.2174/1573406418666220606093202
- Hanan A. Mohamed, Mohamed S. Bekheit, Ewies F. Ewies, Hanem M. Awad, Richard Betz, Eric C. Hosten, Bakr F. Abdel-Wahab. Design of new hybrids indole/phthalimide/oxadiazole-1,2,3 triazole agents and their anticancer properties. Journal of Molecular Structure 2023, 1274 , 134415. https://doi.org/10.1016/j.molstruc.2022.134415
- Mohammed A. Baashen. Synthesis and antibacterial evaluation of novel hydrazones and bis-hydrazones containing 1,2,3-triazole moiety. Journal of Taibah University for Science 2022, 16
(1)
, 1157-1164. https://doi.org/10.1080/16583655.2022.2151297
- Veerabhadraiah Mallikanti, Vishnu Thumma, Krishna Chaitanya Veeranki, Srinivas Gali, Jalapathi Pochampally. Synthesis, Cytotoxicity, Molecular Docking and ADME Assay of Novel Morpholine appended 1,2,3‐Triazole Analogues. ChemistrySelect 2022, 7
(48)
https://doi.org/10.1002/slct.202204020
- Annapurna Mamidala, Karthik Bokkala, Narasimha Swamy Thirukovela, Narsimha Sirassu, Srinivas Bandari, Satheesh Kumar Nukala. Synthesis of Quinoline‐Morpholine‐Coupled 1,2,3‐Triazole Hybrids as
In vitro
EGFR inhibitors. ChemistrySelect 2022, 7
(47)
https://doi.org/10.1002/slct.202203763
- Dinh Thanh Nguyen, Son Hai Do, Thi Huyen Le, Thi Hanh Nguyen, Minh Huyen Nguyen, Thi Ngoc Bich Vu, Thi Thu Hien Pham, Ngoc Toan Vu, Thi Kim Van Hoàng, Thi Kim Giang Nguyen. Synthesis and antiproliferative activity of 1
H
-1,2,3-triazole-4
H
-chromene-
d
-glucose hybrid compounds with dual inhibitory activity against EGFR/VEGFR-2 and molecular docking studies. New Journal of Chemistry 2022, 46
(48)
, 23179-23197. https://doi.org/10.1039/D2NJ04373D
- Eman A. Madbouly, El-Sayed M. Lashine, Ahmed A. Al-Karmalawy, Mahmoud M. Sebaiy, Harris Pratsinis, Dimitris Kletsas, Kamel Metwally. Design and synthesis of novel quinazolinone–chalcone hybrids as potential apoptotic candidates targeting caspase-3 and PARP-1:
in vitro
, molecular docking, and SAR studies. New Journal of Chemistry 2022, 46
(46)
, 22013-22029. https://doi.org/10.1039/D2NJ04053K
- Yanyan Wang, Ruicheng Chen, Yiping Hu, Shulin Jiao, Zhihong Zou. Synthesis, Molecular Docking Study, and Anticancer Activity of 7-Azaindole-1,2,3-triazol Bearing N-Benzamide Derivatives. Russian Journal of General Chemistry 2022, 92
(10)
, 2119-2131. https://doi.org/10.1134/S1070363222100255
- Devidas S. Bhagat, Gurvinder S. Bumbrah, Pooja A. Chawla, Wasudeo B. Gurnule, Sampada K. Shejul. Recent Advances in Synthesis and Anticancer Potential of Triazole-Containing
Scaffolds. Anti-Cancer Agents in Medicinal Chemistry 2022, 22
(16)
, 2852-2875. https://doi.org/10.2174/1871520622666220217161346
- Marius Mioc, Alexandra Mioc, Alexandra Prodea, Andreea Milan, Mihaela Balan-Porcarasu, Roxana Racoviceanu, Roxana Ghiulai, Gheorghe Iovanescu, Ioana Macasoi, George Draghici, Cristina Dehelean, Codruta Soica. Novel Triterpenic Acid—Benzotriazole Esters Act as Pro-Apoptotic Antimelanoma Agents. International Journal of Molecular Sciences 2022, 23
(17)
, 9992. https://doi.org/10.3390/ijms23179992
- Gouthami Dasari, Srinivas Bandari, Satheesh Kumar Nukala, Narasimha Swamy Thirukovela, Narsimha Sirassu, Vinitha Badithapuram, Ravinder Manchal. In vitro Anticancer and Insilico Studies of Quinoxaline‐sulfonyl‐1,2,4‐triazole Hybrids. ChemistrySelect 2022, 7
(29)
https://doi.org/10.1002/slct.202200681
- Manoj Kumar N, Satheesh Kumar Nukala, Narasimha Swamy Thirukovela, Rakesh Sreerama, Ramya Sucharitha E, Pavan Kamarajugadda, Sirassu Narsimha. Ramachary-Bressy-Wang [3+2]cycloaddition reaction: Synthesis of fully decorated 1,2,3-triazoles as potent anticancer and EGFR inhibitors. Journal of Molecular Structure 2022, 1262 , 132975. https://doi.org/10.1016/j.molstruc.2022.132975
- Cauê Benito Scarim, Chung Man Chin. Recent Trends in Drug Development for the Treatment of Adenocarcinoma Breast
Cancer: Thiazole, Triazole, and Thiosemicarbazone Analogues as Efficient Scaffolds. Anti-Cancer Agents in Medicinal Chemistry 2022, 22
(12)
, 2204-2240. https://doi.org/10.2174/1871520621666211201152815
- Kashif Haider, Subham Das, Alex Joseph, M. Shahar Yar. An appraisal of anticancer activity with structure–activity relationship of quinazoline and quinazolinone analogues through EGFR and VEGFR inhibition: A review. Drug Development Research 2022, 83
(4)
, 859-890. https://doi.org/10.1002/ddr.21925
- Shital Patil, Shashikant Bhandari. A Review: Discovering 1,3,4-oxadiazole and Chalcone Nucleus for Cytotoxicity
/ EGFR Inhibitory Anticancer Activity. Mini-Reviews in Medicinal Chemistry 2022, 22
(5)
, 805-820. https://doi.org/10.2174/1389557521666210902160644
- Amita J. Jivani, Kaushik S. Lunagariya, Khushal M. Kapadiya, Piyush V. Dholaria, Vijay M. Khedkar, Ranjan C. Khunt. A Step-By-Step Synthesis of S-Methylene Linker Containing Triazole-Benzoxazole Hybrids by Click Chemistry Approach: Anticancer Activity in Human Cells. Analytical Chemistry Letters 2022, 12
(1)
, 44-57. https://doi.org/10.1080/22297928.2021.1894604
- Mohammad Mahboob Alam. 1,2,3‐Triazole hybrids as anticancer agents: A review. Archiv der Pharmazie 2022, 355
(1)
https://doi.org/10.1002/ardp.202100158
- David Samuvel Michael, M. Krishna Priya, J. Sidharthan, M. Kumar, Rajadurai Vijay Solomon, D. Reuben Jonathan. Synthesis, crystallography, DFT, MTT assay, and molecular docking studies of an exocyclic double-bonded crystalline chalcone. Chemical Data Collections 2021, 36 , 100773. https://doi.org/10.1016/j.cdc.2021.100773
- X Ge, Y Zhang, F Huang, Y Wu, J Pang, X Li, F Fan, H Liu, S Li. EGFR tyrosine kinase inhibitor Almonertinib induces apoptosis and autophagy mediated by reactive oxygen species in non-small cell lung cancer cells. Human & Experimental Toxicology 2021, 40
(12_suppl)
, S49-S62. https://doi.org/10.1177/09603271211030554
- Paulina Wdowiak, Joanna Matysiak, Piotr Kuszta, Katarzyna Czarnek, Ewa Niezabitowska, Tomasz Baj. Quinazoline Derivatives as Potential Therapeutic Agents in Urinary Bladder Cancer Therapy. Frontiers in Chemistry 2021, 9 https://doi.org/10.3389/fchem.2021.765552
- Sumathi Vodnala, Anagani Kanaka Durga Bhavani, Shankaraiah Pagilla, Muralidhar Allam, Nagamani Rayala, Anwita Mudiraj, Phanithi Prakash Babu. Synthesis and Cytotoxic Studies of Quinazoline-Triazole Hybrid Aza Heterocycles. Russian Journal of General Chemistry 2021, 91
(11)
, 2304-2310. https://doi.org/10.1134/S1070363221110189
- Teodora Constantinescu, Claudiu N. Lungu. Anticancer Activity of Natural and Synthetic Chalcones. International Journal of Molecular Sciences 2021, 22
(21)
, 11306. https://doi.org/10.3390/ijms222111306
- Saleh K. Ihmaid, Shaya Yahya Alraqa, Mohamed R. Aouad, Ateyatallah Aljuhani, Hossein M. Elbadawy, Samir A. Salama, Nadjet Rezki, Hany E.A. Ahmed. Design of molecular hybrids of phthalimide-triazole agents with potent selective MCF-7/HepG2 cytotoxicity: Synthesis, EGFR inhibitory effect, and metabolic stability. Bioorganic Chemistry 2021, 111 , 104835. https://doi.org/10.1016/j.bioorg.2021.104835
- Pedro Alves Bezerra Morais, Carla Santana Francisco, Heberth de Paula, Rayssa Ribeiro, Mariana Alves Eloy, Clara Lirian Javarini, Álvaro Cunha Neto, Valdemar Lacerda Júnior. Semisynthetic Triazoles as an Approach in the Discovery of Novel Lead Compounds. Current Organic Chemistry 2021, 25
(10)
, 1097-1179. https://doi.org/10.2174/1385272825666210126100227
- Channa Basappa Vagish, Karthik Kumara, Hamse Kameshwar Vivek, Srinivasan Bharath, Neratur Krishnappagowda Lokanath, Kariyappa Ajay Kumar. Coumarin-triazole hybrids: Design, microwave-assisted synthesis, crystal and molecular structure, theoretical and computational studies and screening for their anticancer potentials against PC-3 and DU-145. Journal of Molecular Structure 2021, 1230 , 129899. https://doi.org/10.1016/j.molstruc.2021.129899
- Parul Grover, Monika Bhardwaj, Garima Kapoor, Lovekesh Mehta, Roma Ghai, K. Nagarajan. Advances on Quinazoline Based Congeners for Anticancer Potential. Current Organic Chemistry 2021, 25
(6)
, 695-723. https://doi.org/10.2174/1385272825666210212121056
- Muhammad Kumayl Abdulwahab, Ke Han Tan, Rashidi Dzulkeflee, Kok Hoong Leong, Choon Han Heh, Azhar Ariffin. In-silico Studies of the Antiproliferative Activity of New Anilinoquinazoline Derivatives Against NSCLC Cells. Journal of Molecular Structure 2021, 1228 , 129786. https://doi.org/10.1016/j.molstruc.2020.129786
- Sumit Kumar, Bharvi Sharma, Vishu Mehra, Vipan Kumar. Recent accomplishments on the synthetic/biological facets of pharmacologically active 1H-1,2,3-triazoles. European Journal of Medicinal Chemistry 2021, 212 , 113069. https://doi.org/10.1016/j.ejmech.2020.113069
- Fanyong Yan, Hao Zhang, Xing Li, Xiaodong Sun, Yingxia Jiang, Yali Cui. A fluorescein-coumarin based ratiometric fluorescent probe for detecting hydrazine and its real applications in cells imaging. Talanta 2021, 223 , 121779. https://doi.org/10.1016/j.talanta.2020.121779
- Luiz Claudio Ferreira Pimentel, Lucas Villas Boas Hoelz, Henayle Fernandes Canzian, Frederico Silva Castelo Branco, Andressa Paula de Oliveira, Vinicius Rangel Campos, Floriano Paes Silva Júnior, Rafael Ferreira Dantas, Jackson Antônio Lamounier Camargos Resende, Anna Claudia Cunha, Nubia Boechat, Mônica Macedo Bastos. (Phenylamino)pyrimidine-1,2,3-triazole derivatives as analogs of imatinib: searching for novel compounds against chronic myeloid leukemia. Beilstein Journal of Organic Chemistry 2021, 17 , 2260-2269. https://doi.org/10.3762/bjoc.17.144
- Prashant S. Auti, Ginson George, Atish T. Paul. Recent advances in the pharmacological diversification of quinazoline/quinazolinone hybrids. RSC Advances 2020, 10
(68)
, 41353-41392. https://doi.org/10.1039/D0RA06642G
- Adileh Ayati, Setareh Moghimi, Somayeh Salarinejad, Maliheh Safavi, Behjat Pouramiri, Alireza Foroumadi. A review on progression of epidermal growth factor receptor (EGFR) inhibitors as an efficient approach in cancer targeted therapy. Bioorganic Chemistry 2020, 99 , 103811. https://doi.org/10.1016/j.bioorg.2020.103811
- Koroush Mashayekh, Pezhman Shiri. An Overview of Recent Advances in the Applications of Click Chemistry in the Synthesis of Bioconjugates with Anticancer Activities. ChemistrySelect 2019, 4
(46)
, 13459-13478. https://doi.org/10.1002/slct.201902362
- Zhi Xu, Shi-Jia Zhao, Yi Liu. 1,2,3-Triazole-containing hybrids as potential anticancer agents: Current developments, action mechanisms and structure-activity relationships. European Journal of Medicinal Chemistry 2019, 183 , 111700. https://doi.org/10.1016/j.ejmech.2019.111700
Article Views are the COUNTER-compliant sum of full text article downloads since November 2008 (both PDF and HTML) across all institutions and individuals. These metrics are regularly updated to reflect usage leading up to the last few days.
Citations are the number of other articles citing this article, calculated by Crossref and updated daily. Find more information about Crossref citation counts.
The Altmetric Attention Score is a quantitative measure of the attention that a research article has received online. Clicking on the donut icon will load a page at altmetric.com with additional details about the score and the social media presence for the given article. Find more information on the Altmetric Attention Score and how the score is calculated.
Recommended Articles
References
This article references 28 other publications.
- 1Fidler, M. M.; Bray, F.; Soerjomataram, I. The global cancer burden and human development: A review. Scand. J. Public Health 2018, 46, 27– 36, DOI: 10.1177/14034948177154001https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC1cjjslentg%253D%253D&md5=94ce8f48e941130e946974bb5d60e747The global cancer burden and human development: A reviewFidler Miranda M; Bray Freddie; Soerjomataram IsabelleScandinavian journal of public health (2018), 46 (1), 27-36 ISSN:.AIMS: This review examines the links between human development and cancer overall and for specific types of cancer, as well as cancer-related risk-factors and outcomes, such as disability and life expectancy. METHODS: To assess human development, the Human Development Index was utilized continuously and according to four levels (low, medium, high, very high), where the low and very high categories include the least and most developed countries, respectively. All studies that assessed aspects of the global cancer burden using this measure were reviewed. RESULTS: Although the present cancer incidence burden is greater in higher Human Development Index countries, a greater proportion of the global mortality burden is observed in less developed countries, with a higher mean fatality rate in the latter countries. Further, the future cancer burden is expected to disproportionally affect less developed regions; in particular, it has been estimated that low and medium Human Development Index countries will experience a 100% and 81% increase in cancer incidence from 2008 to 2030, respectively. Disparities were also observed in risk factors and average health outcomes, such as a greater number of years of life lost prematurely and fewer cancer-related gains in life expectancy observed in lower versus higher Human Development Index settings. CONCLUSIONS: From a global perspective, there remain clear disparities in the cancer burden according to national Human Development Index scores. International efforts are needed to aid countries in social and economic transition in order to efficiently plan, implement and evaluate cancer control initiatives as a means to reduce the widening gap in cancer occurrence and survival worldwide.
- 2Ciardiello, F.; Tortora, G. Epidermal growth factor receptor (EGFR) as a target in cancer therapy: understanding the role of receptor expression and other molecular determinants that could influence the response to anti-EGFR drugs. Eur. J. Cancer 2003, 39, 1348– 1354, DOI: 10.1016/s0959-8049(03)00235-1There is no corresponding record for this reference.
- 3Cohen, M. H.; Johnson, J. R.; Chen, Y. F.; Sridhara, R.; Pazdur, R. FDA Drug Approval Summary: Erlotinib (Tarceva(R)) Tablets. Oncologist 2005, 10, 461– 466, DOI: 10.1634/theoncologist.10-7-461There is no corresponding record for this reference.
- 4Cohen, M. H.; Williams, G. A.; Sridhara, R.; Chen, G.; Pazdur, R. FDA Drug Approval Summary: Gefitinib (ZD1839) (Iressa(R)) Tablets. Oncologist 2003, 8, 303– 306, DOI: 10.1634/theoncologist.8-4-303There is no corresponding record for this reference.
- 5Tan, F.; Shi, Y.; Wang, Y.; Ding, L.; Yuan, X.; Sun, Y. Icotinib, a selective EGF receptor tyrosine kinase inhibitor, for the treatment of non-small-cell lung cancer. Future Oncol. 2015, 11, 385– 397, DOI: 10.2217/fon.14.2495https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXisF2ls7s%253D&md5=a19fbbc42fd06b8971bc123155d180c0Icotinib, a selective EGF receptor tyrosine kinase inhibitor, for the treatment of non-small-cell lung cancerTan, Fenlai; Shi, Yuankai; Wang, Yinxiang; Ding, Lieming; Yuan, Xiaobin; Sun, YanFuture Oncology (2015), 11 (3), 385-397CODEN: FOUNBN; ISSN:1479-6694. (Future Medicine Ltd.)A review. Advanced non-small-cell lung cancer (NSCLC) is the main cause for cancer-related mortality. Treatments for advanced NSCLC are largely palliative and a benefit plateau appears to have reached with the platinum-based chemotherapy regimens. EGF receptor (EGFR) tyrosine kinase inhibitors gefitinib, erlotinib and afatinib came up with prolonged progression-free survival and improved quality of life, esp. in EGFR-mutated patients. Icotinib is an oral selective EGFR tyrosine kinase, which was approved by China Food and Drug administration in June 2011 for treating advanced NSCLC. Its approval was based on the registered Phase III trial (ICOGEN), which showed icotinib is noninferior to gefitinib. This review will discuss the role of icotinib in NSCLC, and its potential application and ongoing investigations.
- 6Wu, P.; Nielsen, T. E.; Clausen, M. H. FDA-approved small-molecule kinase inhibitors. Trends Pharmacol. Sci. 2015, 36, 422– 439, DOI: 10.1016/j.tips.2015.04.0056https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXns1KisLg%253D&md5=a6aa3dbb11e6fe3001fa5fa03a4c950aFDA-approved small-molecule kinase inhibitorsWu, Peng; Nielsen, Thomas E.; Clausen, Mads H.Trends in Pharmacological Sciences (2015), 36 (7), 422-439CODEN: TPHSDY; ISSN:0165-6147. (Elsevier Ltd.)A review. Kinases have emerged as one of the most intensively pursued targets in current pharmacol. research, esp. for cancer, due to their crit. roles in cellular signaling. To date, the US FDA has approved 28 small-mol. kinase inhibitors, half of which were approved in the past 3 years. While the clin. data of these approved mols. are widely presented and structure-activity relationship (SAR) has been reported for individual mols., an updated review that analyzes all approved mols. and summarizes current achievements and trends in the field has yet to be found. Here we present all approved small-mol. kinase inhibitors with an emphasis on binding mechanism and structural features, summarize current challenges, and discuss future directions in this field.
- 7Dungo, R. T.; Keating, G. M. Afatinib: first global approval. Drugs 2013, 73, 1503– 1515, DOI: 10.1007/s40265-013-0111-67https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXhvVWmurnM&md5=21851cbfac56e2fffcba6c731f123cffAfatinib: First Global ApprovalDungo, Rosselle T.; Keating, Gillian M.Drugs (2013), 73 (13), 1503-1515CODEN: DRUGAY; ISSN:0012-6667. (Springer International Publishing AG)A review. Afatinib, an irreversible inhibitor of the ErbB family of tyrosine kinases, is under development with Boehringer Ingelheim for the once-daily, oral treatment of cancer. Afatinib downregulates ErbB signalling by covalently binding to epidermal growth factor receptor (EGFR), human epidermal growth factor receptor (HER) 2 and HER4, irreversibly inhibiting tyrosine kinase autophosphorylation. It also inhibits transphosphorylation of HER3. Oral afatinib (Gilotrif®) has been approved in the US for the first-line treatment of patients with metastatic non-small-cell lung cancer (NSCLC) who have tumors with EGFR exon 19 deletions or exon 21 (L858R) substitution mutations as detected by a US FDA-approved test. Afatinib has also been approved in Taiwan for the first-line treatment of patients with EGFR mutation-pos. NSCLC. In addn., the European Medicines Agency's Committee for Medicinal Products for Human Use has recommended the approval of afatinib (Giotrif) for the treatment of patients with locally advanced or metastatic NSCLC with activating EGFR mutations who are EGFR tyrosine kinase inhibitor naive. Afatinib is also under regulatory review in Canada, Japan and other Asian countries. This article summarizes the milestones in the development of afatinib, leading to this first approval in patients with metastatic NSCLC.
- 8Leung, E. L.-H.; Fan, X.-X.; Wong, M. P.; Jiang, Z.-H.; Liu, Z.-Q.; Yao, X.-J.; Lu, L.-L.; Zhou, Y.-L.; Yau, L.-F.; Tin, V. P.-C.; Liu, L. Targeting tyrosine kinase inhibitor-resistant non-small cell lung cancer by inducing epidermal growth factor receptor degradation via methionine 790 oxidation. Antioxid. Redox Signaling 2016, 24, 263– 279, DOI: 10.1089/ars.2015.64208https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC28XhvV2ktrs%253D&md5=3eefc008e2ebac6a82122f3d87e849c2Targeting Tyrosine Kinase Inhibitor-Resistant Non-Small Cell Lung Cancer by Inducing Epidermal Growth Factor Receptor Degradation via Methionine 790 OxidationLeung, Elaine Lai-Han; Fan, Xing-Xing; Wong, Maria Pik; Jiang, Zhi-Hong; Liu, Zhong-Qiu; Yao, Xiao-Jun; Lu, Lin-Lin; Zhou, Yan-Ling; Yau, Li-Fong; Tin, Vicky Pui-Chi; Liu, LiangAntioxidants & Redox Signaling (2016), 24 (5), 263-279CODEN: ARSIF2; ISSN:1523-0864. (Mary Ann Liebert, Inc.)Aims: Epidermal growth factor receptor (EGFR) tyrosine kinase inhibitors (TKIs) have been developed to treat non-small cell lung cancer (NSCLC) patients with EGFR mutation, but TKI resistance is common. Almost half of the acquired resistance patients are due to addnl. T790M mutation on EGFR (EGFRT790M), thus overcoming TKI resistance is important. In this study, we aim to investigate the role of reactive oxygen species (ROS) in TKI resistance as well as the mol. and biol. effects of EGFRT790M after redox manipulation. Results: The basal ROS levels in EGFRT790M-contg. TKI-resistant NSCLC cell lines were substantially high. Sixty-three human lung tumors showed higher NADPH oxidase isoform 2 (NOX2) expression than normal lung tissues, which may contribute to high basal ROS in cancer and poor survival. Interestingly, only NOX3 was upregulated by sanguinarine, a pharmacol. agent to elevate ROS, and resulted in EGFR overoxidn., degrdn., and apoptosis. By contrast, such responses were lacking in EGFRWT cells. Selective EGFRT790M degrdn. was manipulated by redox imbalance between NOX3 and methionine reductase A (MsrA). Furthermore, the in vivo tumor suppression effect of sanguinarine, NOX3 upregulation, and EGFR degrdn. were confirmed. Innovation: We have found a new treatment strategy to overcome TKI resistance by selectively inducing EGFRT790M degrdn. via specific stimulation of methionine 790 (M790) oxidn. It can be achieved via manipulating redox imbalance between NOX3 and MsrA. Conclusion: Targeting EGFR by elevating ROS and redox imbalance is a potential new strategy to develop a new EGFR inhibitor for TKI-resistant patients with a wide therapeutic window between EGFRT790M and EGFRWT. Antioxid. Redox Signal. 24, 263-279.
- 9Schacher-Kaufmann, S.; Pless, M. Acute fatal liver toxicity under erlotinib. Case Rep. Oncol. 2010, 3, 182– 188, DOI: 10.1159/0003153669https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A280%3ADC%252BC2sbos1yqsQ%253D%253D&md5=3180e9e98456bd0eacd6dc721c7eb932Acute Fatal Liver Toxicity under ErlotinibSchacher-Kaufmann Sabina; Pless MiklosCase reports in oncology (2010), 3 (2), 182-188 ISSN:1662-6575.We describe the case of a never-smoker who received second-line erlotinib as a treatment for his non-small cell lung cancer. Within one month, acute hepatic failure developed as well as a thrombotic-thrombocytopenic microangiopathy, with fatal outcome. In patients with non-small cell lung cancer, hepatic toxicity of erlotinib is a rare but severe complication; so far three fatal cases have been reported. Patients' liver function should be assessed before starting erlotinib and special care is recommended if pretreatment bilirubin is elevated.
- 10Stathopoulos, G.; Trafalis, D.; Athanasiou, A.; Bardi, G.; Chandrinou, H. Serious hematologic complications following erlotinib treatment. Anticancer Res. 2010, 30, 973– 97610https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXlslelu7s%253D&md5=bf3eb9980434cddda198562f4a226716Serious hematologic complications following erlotinib treatmentStathopoulos, G. P.; Trafalis, Dim; Athanasiou, A.; Bardi, G.; Chandrinou, H.Anticancer Research (2010), 30 (3), 973-976CODEN: ANTRD4; ISSN:0250-7005. (International Institute of Anticancer Research)Background: Erlotinib is an oral, small-mol. targeting therapy which inhibits epidermal growth factor tyrosine kinase receptors. Erlotinib has been administered for the treatment of advanced pancreatic cancer and non-small cell lung cancer. Patients and Methods: In the present report, unusual hematol. complications were detected after erlotinib was administered as second-line monotherapy in pretreated patients with advanced non-small cell lung cancer. Four patients pre-treated with cisplatin or its analog-based combinations, were evaluated. Erlotinib was given at a dose of 150 mg daily. In cases of intolerable adverse reactions, the dose was either reduced to 100 mg daily or treatment was interrupted for a max. of two weeks. Results: Serious hematol. toxicity (or complications) developed in these 4 patients after 4-8.5 mo of treatment. Two patients developed leukemias (AML, CML) and two, myelodysplastic syndrome. Conclusion: Whether or not these hematol. complications were related to erlotinib treatment is comprehensively discussed.
- 11Rajput, R.; Mishra, A. P. A review on biological activity of quinazolinones. Int. J. Pharm. Pharm. Sci. 2012, 4, 66– 7011https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38Xms1Cmtrg%253D&md5=253d611f1b2c3e887bf4d403d261026cA review on biological activity of quinazolinonesRajput, Rakhi; Mishra, Abhinav PrasoonInternational Journal of Pharmacy and Pharmaceutical Sciences (2012), 4 (2), 66-70CODEN: IJPPKB; ISSN:0975-1491. (International Journal of Pharmacy and Pharmaceutical Sciences)A review. Quinazolinones is considered as an important chem. synthesis of various physiol. significance and pharmacol. utility. Quinazolinones are a large class of active chem. compds. exhibiting a broad spectrum of biol. activities in animals as well as in humans. Literature studies on quinazolinones have shown that these derivs. possess a wide variety of biol. activities such as anti HIV, anticancer, antifungal, antibacterial, antimutagenic, anticoccidial, anticonvulsant, anti-inflammatory, CNS depressant, antimalarial, antioxidant, antileukemic activity, antileishmanial activity. This review focused on the various biol. activities of quinazolinones.
- 12(a) Cheng, C.-M.; Lee, Y.-J.; Wang, W.-T.; Hsu, C.-T.; Tsai, J.-S.; Wu, C.-M.; Ou, K.-L.; Yang, T.-S. Determining the binding mode and binding affinity constant of tyrosine kinase inhibitor PD153035 to DNA using optical tweezers. Biochem. Biophys. Res. Commun. 2011, 404, 297– 301, DOI: 10.1016/j.bbrc.2010.11.110There is no corresponding record for this reference.(b) Selvam, T. P.; Kumar, P. V. Quinazoline marketed drugs. Res. Pharm. 2011, 1 ().There is no corresponding record for this reference.
- 13Medapi, B.; Suryadevara, P.; Renuka, J.; Sridevi, J. P.; Yogeeswari, P.; Sriram, D. 4-Aminoquinoline derivatives as novel Mycobacterium tuberculosis GyrB inhibitors: structural optimization, synthesis and biological evaluation. Eur. J. Med. Chem. 2015, 103, 1– 16, DOI: 10.1016/j.ejmech.2015.06.032There is no corresponding record for this reference.
- 14Christopher, E.; Bedir, E.; Dunbar, C.; Khan, I. A.; Okunji, C. O.; Schuster, B. M.; Iwu, M. M. Indoloquinazoline Alkaloids fromAraliopsis tabouensis. Helv. Chim. Acta 2003, 86, 2914– 2918, DOI: 10.1002/hlca.200390239There is no corresponding record for this reference.
- 15Mendoza-Martínez, C.; Galindo-Sevilla, N.; Correa-Basurto, J.; Ugalde-Saldivar, V. M.; Rodríguez-Delgado, R. G.; Hernández-Pineda, J.; Padierna-Mota, C.; Flores-Alamo, M.; Hernández-Luis, F. Antileishmanial activity of quinazoline derivatives: Synthesis, docking screens, molecular dynamic simulations and electrochemical studies. Eur. J. Med. Chem. 2015, 92, 314– 331, DOI: 10.1016/j.ejmech.2014.12.051There is no corresponding record for this reference.
- 16Antipenko, L. N.; Karpenko, A. V.; Kovalenko, S. I.; Katsev, A. M.; Komarovska-Porokhnyavets, E. Z.; Novikov, V. P. Synthesis, Cytotoxicity by Bioluminescence Inhibition, Antibacterial and Antifungal Activity of ([1,2,4]Triazolo[1,5-c]quinazolin-2-ylthio)carboxylic Acid Amides. Arch. Pharm. 2009, 342, 651– 662, DOI: 10.1002/ardp.20090007716https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1MXhtlGgsLnM&md5=64f2172cb9615f7a320ad9fb559987cbSynthesis, Cytotoxicity by Bioluminescence Inhibition, Antibacterial and Antifungal Activity of ([1,2,4]Triazolo[1,5-c]quinazolin-2-ylthio)carboxylic Acid AmidesAntipenko, Lyudmila N.; Karpenko, Alexander V.; Kovalenko, Sergey I.; Katsev, Andrew M.; Komarovska-Porokhnyavets, Elena Z.; Novikov, Vladimir P.Archiv der Pharmazie (Weinheim, Germany) (2009), 342 (11), 651-662CODEN: ARPMAS; ISSN:0365-6233. (Wiley-VCH Verlag GmbH & Co. KGaA)We report in this work the synthesis, cytotoxicity, and antimicrobial activity of ([1,2,4]triazolo[1,5-c]quinazolin-2-ylthio)carboxylic acid amides 4-7 in connection with our previous research in the prepn. of triazoloquinazoline derivs. Due to simplicity, general availability of starting materials, and high yields, the most reliable method of synthesis appeared to be the one with N,N-carbonyldiimidazole activation stage. The chem. structures of all obtained substances were deduced from FT-IR, 1H-NMR, EI-MS, and LC-MS spectral data. The results of cytotoxicity evaluated by bioluminescence inhibition of bacterium Photobacterium leiognathi, strain Sh1 showed that compds.4.1, 4.6, and 6.1 were the most cytotoxic. Investigation of the antimicrobial and antifungal activity of amides 4-7 (concn. 5 mg/mL) was carried out by the stiff-plate agar-diffusion method. We found that the compds. possessed low (4.1, 4.7) antifungal activity against Candida tenuis and strong (4.21, 5.1, 5.9) or inefficient (4.7, 4.12, 4.16) activity against Aspergillus niger. Substances 5.1 and 5.9 slightly affected Mycobacterium luteum. Staphylococcus aureus was resistant to all obtained substances, and only the n-butyramide derivs. 7.1 and 7.5 inhibited the growth of Escherichia coli. Hence, there was no strong correlation between bioluminescence inhibition and antimicrobial activity of the investigated substances.
- 17Horne, W. S.; Yadav, M. K.; Stout, C. D.; Ghadiri, M. R. Heterocyclic Peptide Backbone Modifications in an α-Helical Coiled Coil. J. Am. Chem. Soc. 2004, 126, 15366– 15367, DOI: 10.1021/ja045040817https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD2cXptlSgtrw%253D&md5=6f4573960370cdded62c2ca77ad25b04Heterocyclic Peptide Backbone Modifications in an α-Helical Coiled CoilHorne, W. Seth; Yadav, Maneesh K.; Stout, C. David; Ghadiri, M. RezaJournal of the American Chemical Society (2004), 126 (47), 15366-15367CODEN: JACSAT; ISSN:0002-7863. (American Chemical Society)In this paper, we present 1,2,3-triazole ε2-amino acids incorporated as a dipeptide surrogate at three positions in the sequence of a known α-helical coiled coil. Biophys. characterization indicates that the modified peptides retain much of the helical structure of the parent sequence, and that the thermodn. stability of the coiled coil depends on the position of the incorporation of the ε-residue. Crystal structures obtained for each peptide give insight into the chem. behavior and conformational preferences of the non-natural amino acid and show that the triazole ring can participate in the backbone hydrogen bonding of the α-helix as well as template an interhelical crossing between chains in the bundle.
- 18(a) Reddy, D. M.; Srinivas, J.; Chashoo, G.; Saxena, A. K.; Sampath Kumar, H. M. 4β-[(4-Alkyl)-1,2,3-triazol-1-yl] podophyllotoxins as anticancer compounds: Design, synthesis and biological evaluation. Eur. J. Med. Chem. 2011, 46, 1983– 1991, DOI: 10.1016/j.ejmech.2011.02.016There is no corresponding record for this reference.(b) Yoon, J.; Ryu, J.-S. A rapid synthesis of lavendustin-mimetic small molecules by click fragment assembly. Bioorg. Med. Chem. Lett. 2010, 20, 3930– 3935, DOI: 10.1016/j.bmcl.2010.05.01418bhttps://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3cXnsVWis7w%253D&md5=e07a4f2aa1c64bef25822ef2706547bfA rapid synthesis of lavendustin-mimetic small molecules by click fragment assemblyYoon, Jieun; Ryu, Jae-SangBioorganic & Medicinal Chemistry Letters (2010), 20 (13), 3930-3935CODEN: BMCLE8; ISSN:0960-894X. (Elsevier B.V.)Lavendustin-mimetic small mols. modifying the linker -CH2-NH- with an 1,2,3-triazole ring have been synthesized via a click chem. Two pharmacophoric fragments of lavendustin were varied to investigate chem. space and the auxophoric -CH2-NH- was altered to an 1,2,3-triazole for rapid click conjugation. The small mols. were evaluated against HCT116 colon cancer and CCRF-CEM leukemia cell lines. Among the 28 analogs, 3-phenylpropyl ester I inhibited CCRF-CEM leukemia cell growth with GI50 value of 0.9 μM.
- 19Giffin, M. J.; Heaslet, H.; Brik, A.; Lin, Y.-C.; Cauvi, G.; Wong, C.-H.; McRee, D. E.; Elder, J. H.; Stout, C. D.; Torbett, B. E. A Copper(I)-Catalyzed 1,2,3-Triazole Azide–Alkyne Click Compound Is a Potent Inhibitor of a Multidrug-Resistant HIV-1 Protease Variant. J. Med. Chem. 2008, 51, 6263– 6270, DOI: 10.1021/jm800149m19https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD1cXhtF2lsrvK&md5=c3ca2b1ae9e3a747ed7161715575f964A Copper(I)-Catalyzed 1,2,3-Triazole Azide-Alkyne Click Compound Is a Potent Inhibitor of a Multidrug-Resistant HIV-1 Protease VariantGiffin, Michael J.; Heaslet, Holly; Brik, Ashraf; Lin, Ying-Chuan; Cauvi, Gabrielle; Wong, Chi-Huey; McRee, Duncan E.; Elder, John H.; Stout, C. David; Torbett, Bruce E.Journal of Medicinal Chemistry (2008), 51 (20), 6263-6270CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Treatment with HIV-1 protease inhibitors, a component of highly active antiretroviral therapy (HAART), often results in viral resistance. Structural and biochem. characterization of a 6X protease mutant arising from in vitro selection with compd. 1, a C2-sym. diol protease inhibitor, has been previously described. We now show that compd. 2, a copper(I)-catalyzed 1,2,3-triazole derived compd. previously shown to be potently effective against wild-type protease (IC50 = 6.0 nM), has low nanomolar activity (IC50 = 15.7 nM) against the multidrug-resistant 6X protease mutant. Compd. 2 displays similar efficacy against wild-type and 6X HIV-1 in viral replication assays. While structural studies of compd. 1 bound to wild-type and mutant proteases revealed a progressive change in binding mode in the mutants, the 1.3 Å resoln. 6X protease-compd. 2 crystal structure reveals nearly identical interactions for 2 as in the wild-type protease complex with very little change in compd. 2 or protease conformation.
- 20Patpi, S. R.; Pulipati, L.; Yogeeswari, P.; Sriram, D.; Jain, N.; Sridhar, B.; Murthy, R.; Anjana Devi, T.; Kalivendi, S. V.; Kantevari, S. Design, Synthesis, and Structure-Activity Correlations of Novel Dibenzo[b,d]furan, Dibenzo[b,d]thiophene, and N-Methylcarbazole Clubbed 1,2,3-Triazoles as Potent Inhibitors of Mycobacterium tuberculosis. J. Med. Chem. 2012, 55, 3911– 3922, DOI: 10.1021/jm300125e20https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC38XksVymurg%253D&md5=aa0a15ae22de7006b3a367fcaf49576bDesign, Synthesis, and Structure-Activity Correlations of Novel Dibenzo[b,d]furan, Dibenzo[b,d]thiophene, and N-Methylcarbazole Clubbed 1,2,3-Triazoles as Potent Inhibitors of Mycobacterium tuberculosisPatpi, Santhosh Reddy; Pulipati, Lokesh; Yogeeswari, Perumal; Sriram, Dharmarajan; Jain, Nishant; Sridhar, Balasubramanian; Murthy, Ramalinga; T, Anjana Devi; Kalivendi, Shasi Vardhan; Kantevari, SrinivasJournal of Medicinal Chemistry (2012), 55 (8), 3911-3922CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)A mol. hybridization approach is an emerging structural modification tool to design new mols. with improved pharmacophoric properties. In this study, 1,2,3-triazole-based Mycobacterium tuberculosis inhibitors and synthetic and natural product-based tricyclic (carbazole, dibenzo[b,d]furan, and dibenzo[b,d]thiophene) antimycobacterial agents were integrated in one mol. platform to prep. various novel clubbed 1,2,3-triazole hybrids using click chem. Structure-activity correlations and in vitro activity against M. tuberculosis strain H37Rv of new analogs revealed the order: dibenzo[b,d]thiophene > dibenzo[b,d]furan > 9-methyl-9H-carbazole series. Some of the most potent M. tuberculosis inhibitors with MIC = 0.78 μg/mL (∼1.9 μM) displayed a low cytotoxicity and high selectivity index (50-255) against four different human cancer cell lines. These results together provided the potential importance of mol. hybridization and the development of triazole clubbed dibenzo[b,d]thiophene-based lead candidates to treat mycobacterial infections.
- 21De Simone, R.; Chini, M. G.; Bruno, I.; Riccio, R.; Mueller, D.; Werz, O.; Bifulco, G. Structure-Based Discovery of Inhibitors of Microsomal Prostaglandin E2Synthase–1, 5-Lipoxygenase and 5-Lipoxygenase-Activating Protein: Promising Hits for the Development of New Anti-inflammatory Agents. J. Med. Chem. 2011, 54, 1565– 1575, DOI: 10.1021/jm101238d21https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3MXitVKqtbw%253D&md5=7e4379cce59ecf39290522c8238a3647Structure-Based Discovery of Inhibitors of Microsomal Prostaglandin E2 Synthase-1, 5-Lipoxygenase and 5-Lipoxygenase-Activating Protein: Promising Hits for the Development of New Anti-inflammatory AgentsDe Simone, Rosa; Chini, Maria Giovanna; Bruno, Ines; Riccio, Raffaele; Mueller, Daniela; Werz, Oliver; Bifulco, GiuseppeJournal of Medicinal Chemistry (2011), 54 (6), 1565-1575CODEN: JMCMAR; ISSN:0022-2623. (American Chemical Society)Microsomal prostaglandin E2 synthase (mPGES)-1 catalyzes the transformation of PGH2 to PGE2 that is involved in several pathologies like fever, pain, and inflammatory disorders. To identify novel mPGES-1 inhibitors, we used in silico screening to rapidly direct the synthesis, based on the copper-catalyzed 3 + 2 Huisgen's reaction (click chem.), of potential inhibitors. We designed 26 new triazole-based compds. in accordance with the pocket binding requirements of human mPGES-1. Docking results, in agreement with ligand efficiency values, suggested the synthesis of 15 compds. that at least in theory were shown to be more efficient in inhibiting mPGES-1. Biol. evaluation of these selected compds. has disclosed three new potential anti-inflammatory drugs: (I) compd. 4 displaying selectivity for mPGES-1 with an IC50 value of 3.2 μM, (II) compd. 20 that dually inhibits 5-lipoxygenase and mPGES-1, and (III) compd. 7 apparently acting as 5-lipoxygenase-activating protein inhibitor (IC50 = 0.4 μM).
- 22Banerji, B.; Killi, S. K.; Katarkar, A.; Chatterjee, S.; Tangella, Y.; Prodhan, C.; Chaudhuri, K. Neo-tanshinlactone D-ring modified novel analogues induce apoptosis in human breast cancer cell via DNA damage. Bioorg. Med. Chem. 2017, 25, 202– 212, DOI: 10.1016/j.bmc.2016.10.026There is no corresponding record for this reference.
- 23Viegas-Junior, C.; Barreiro, E. J.; Fraga, C. A. M. Molecular hybridization: a useful tool in the design of new drug prototypes. Bioorg. Med. Chem. 2007, 14, 1829– 1852, DOI: 10.2174/092986707781058805There is no corresponding record for this reference.
- 24Stamos, J.; Sliwkowski, M. X.; Eigenbrot, C. Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitor. J. Biol. Chem. 2002, 277, 46265– 46272, DOI: 10.1074/jbc.m20713520024https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BD38XovFKhsL4%253D&md5=c63c60b93756bd45fb2375c56ba38941Structure of the epidermal growth factor receptor kinase domain alone and in complex with a 4-anilinoquinazoline inhibitorStamos, Jennifer; Sliwkowski, Mark X.; Eigenbrot, CharlesJournal of Biological Chemistry (2002), 277 (48), 46265-46272CODEN: JBCHA3; ISSN:0021-9258. (American Society for Biochemistry and Molecular Biology)The crystal structure of the kinase domain from the epidermal growth factor receptor (EGFRK) including forty amino acids from the carboxyl-terminal tail has been detd. to 2.6-Å resoln., both with and without an EGFRK-specific inhibitor currently in Phase III clin. trials as an anti-cancer agent, erlotinib (OSI-774, CP-358,774, Tarceva). The EGFR family members are distinguished from all other known receptor tyrosine kinases in possessing constitutive kinase activity without a phosphorylation event within their kinase domains. Despite its lack of phosphorylation, we find that the EGFRK activation loop adopts a conformation similar to that of the phosphorylated active form of the kinase domain from the insulin receptor. Surprisingly, key residues of a putative dimerization motif lying between the EGFRK domain and carboxyl-terminal substrate docking sites are found in close contact with the kinase domain. Significant intermol. contacts involving the carboxyl-terminal tail are discussed with respect to receptor oligomerization.
- 25Towbin, H.; Staehelin, T.; Gordon, J. Electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: procedure and some applications. Proc. Natl. Acad. Sci. U. S. A. 1979, 76, 4350– 4354, DOI: 10.1073/pnas.76.9.435025https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADyaE1MXmtVKltLw%253D&md5=39eea3d100df7959e5d12e39e14cecbfElectrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets: Procedure and some applicationsTowbin, Harry; Staehelin, Theophil; Gordon, JulianProceedings of the National Academy of Sciences of the United States of America (1979), 76 (9), 4350-4CODEN: PNASA6; ISSN:0027-8424.A method was devised for the electrophoretic transfer of proteins from polyacrylamide gels to nitrocellulose sheets. The method results in quant. transfer of ribosomal proteins from gels contg. urea. For Na dodecyl sulfate gels, the original band pattern was obtained with no loss of resoln., but the transfer was not quant. The method allows detection of proteins by autoradiog. and is simpler than conventional procedures. The immobilized proteins were detectable by immunol. procedures. All addnl. binding capacity on the nitrocellulose was blocked with excess protein; then, a specific antibody was bound and, finally, a 2nd antibody directed against the 1st antibody. The 2nd antibody was wither radioactively labeled or conjugated to fluorescein or to peroxidase. The specific protein was then detected by either autoradiog., under UV light, or by the peroxidase reaction product, resp. In the latter case, as little as 100 pg of protein was clearly detectable.
- 26Lambert, A. J.; Brand, M. D. Superoxide production by NADH:ubiquinone oxidoreductase (complex I) depends on the pH gradient across the mitochondrial inner membrane. Biochem. J. 2004, 382, 511– 517, DOI: 10.1042/bj20040485There is no corresponding record for this reference.
- 27Dey, S. K.; Bose, D.; Hazra, A.; Naskar, S.; Nandy, A.; Munda, R. N.; Das, S.; Chatterjee, N.; Mondal, N. B.; Banerjee, S.; Saha, K. D. Cytotoxic activity and apoptosis-inducing potential of di-spiropyrrolidino and di-spiropyrrolizidino oxindole andrographolide derivatives. PLoS One 2013, 8, e58055, DOI: 10.1371/journal.pone.005805527https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC3sXktlSrsLg%253D&md5=64ae726bf948e1fc8225ad1a9ae8f516Cytotoxic activity and apoptosis-inducing potential of di-spiropyrrolidino and di-spiropyrrolizidino oxindole andrographolide derivativesDey, Sumit Kumar; Bose, Dipayan; Hazra, Abhijit; Naskar, Subhendu; Nandy, Abhishek; Munda, Rudra Narayan; Das, Subhadip; Chatterjee, Nabanita; Mondal, Nirup Bikash; Banerjee, Sukdeb; Das Saha, KrishnaPLoS One (2013), 8 (3), e58055CODEN: POLNCL; ISSN:1932-6203. (Public Library of Science)Anticancer role of andrographolide is well documented. To find novel potent derivs. with improved cytotoxicity than andrographolide on cancer cells, two series of di-spiropyrrolidino- and di-spiropyrrolizidino oxindole andrographolide derivs. prepd. by cyclo-addn. of azomethine ylide along with sarcosine or proline (viz. sarcosine and proline series resp.) and substitution of different functional groups (-CH3, -OCH3 and halogens) were examd. for their cytotoxic effect on a panel of six human cancer cell lines (colorectal carcinoma HCT116 cells, pancreatic carcinoma MiaPaCa-2 cells, hepatocarcinoma HepG2 cells, cervical carcinoma HeLa cells, lung carcinoma A549 and melanoma A375 cells). Except halogen substituted derivs. of proline series (viz. CY2, CY14 and CY15 for Br, Cl and I substitution resp.), none of the other derivs. showed improved cytotoxicity than andrographolide in the cancer cell lines examd. Order of cytotoxicity of the potent compds. is CY2>CY14>CY15>andrographolide. Higher toxicity was obsd. in HCT116, MiaPaCa-2 and HepG2 cells. CY2, induced death of HCT116 (GI50 10.5), MiaPaCa-2 (GI50 11.2) and HepG2 (GI50 16.6) cells were assocd. with cell rounding, nuclear fragmentation and increased percentage of apoptotic cells, cell cycle arrest at G1 phase, ROS generation and involvement of mitochondrial pathway. Upregulation of Bax, Bad, p53, caspases-3,-9 and cleaved PARP; downregulation of Bcl-2, cytosolic NF-κB p65, PI3K and p-Akt; translocation of P53/P21, NF-κB p65 were seen in CY2 treated HCT116 cells. Thus, three halogenated di-spiropyrrolizidino oxindole derivs. of andrographolide are found to be more cytotoxic than andrographolide in some cancer cells. The most potent deriv., CY2 induced death of the cancer cells involves ROS dependent mitochondrial pathway like andrographolide.
- 28Nandy, A.; Dey, S.; Das, S.; Munda, R.; Dinda, J.; Saha, K. Gold (I) N-heterocyclic carbene complex inhibits mouse melanoma growth by p53 upregulation. Mol. Cancer 2014, 13, 57, DOI: 10.1186/1476-4598-13-5728https://chemport.cas.org/services/resolver?origin=ACS&resolution=options&coi=1%3ACAS%3A528%3ADC%252BC2MXivFWhtbc%253D&md5=40df17b4bddf6dce687596250172e457Gold (I) N-heterocyclic carbene complex inhibits mouse melanoma growth by p53 upregulationNandy, Abhishek; Dey, Sumit Kumar; Das, Sujata; Munda, Rudra Narayan; Dinda, Joydev; Das Saha, KrishnaMolecular Cancer (2014), 13 (), 57/1-57/14, 14 pp.CODEN: MCOACG; ISSN:1476-4598. (BioMed Central Ltd.)Background: Cancer treatment using gold (I) complexes is becoming popular. In this study, a gold (I) N-heterocyclic complex designated as complex 3 was synthesized, its cytotoxicity was examd., and its anti-melanoma activity was evaluated in vitro and in vivo. Methods: Viability of cancer cells was detd. by MTT assay upon treatment with various concns. of a gold (I) N-heterocyclic carbene complex (complex 3) in a dose and time dependent manner. Mouse melanoma cells B16F10 were selected for further apoptotic studies, including flowcytometric anal. of annexin binding, cell cycle arrest, intracellular ROS generation and loss in the mitochondrial membrane potential. ELISA based assays were done for caspase activities and western blots for detg. the expression of various survival and apoptotic proteins. Immunocytol. was performed to visualize the translocation of p53 to the nucleus. B16F10 cells were inoculated into mice and post tumor formation, complex 3 was administered. Immunohistol. was performed to det. the expressions of p53, p21, NF-κB (p65 and p50), MMP-9 and VEGF. Student's t test was used for detg. statistical significance. The survival rate data were analyzed by Kaplan-Meier plots. Results: Complex 3 markedly inhibited the growth of HCT 116, HepG2, and A549, and induced apoptosis in B16F10 cells with nuclear condensation, DNA fragmentation, externalization of phosphatidylserine, activation of caspase 3 and caspase 9, PARP cleavage, downregulation of Bcl-2, upregulation of Bax, cytosolic cytochrome c elevation, ROS generation, and mitochondrial membrane potential loss indicating the involvement of an intrinsic mitochondrial death pathway. Further, upregulation of p53, p-p53 (ser 15) and p21 indicated the role of p53 in complex 3 mediated apoptosis. The complex reduced tumor size, and caused upregulation of p53 and p21 along with downregulation of NF-κB (p65 and p50), VEGF and MMP-9. These results suggest that it induced anti-melanoma effect in vitro and in vivo by modulating p53 and other apoptotic factors. Conclusions: The gold (I) N-heterocyclic carbene complex (C22H26N6AuO2PF6) designated as complex 3 induced ROS and p53 dependent apoptosis in B16F10 cells involving the mitochondrial death pathway along with suppression of melanoma tumor growth by regulating the levels of pro and anti apoptotic factors (p53, p21, NF-κB, VEGF and MMP-9).
Supporting Information
Supporting Information
The Supporting Information is available free of charge on the ACS Publications website at DOI: 10.1021/acsomega.8b01960.
1H NMR and 13C NMR spectra of all compounds and crystal data of compound 5c (PDF)
Crystallographic data of compound 5c (CIF)
Terms & Conditions
Most electronic Supporting Information files are available without a subscription to ACS Web Editions. Such files may be downloaded by article for research use (if there is a public use license linked to the relevant article, that license may permit other uses). Permission may be obtained from ACS for other uses through requests via the RightsLink permission system: http://pubs.acs.org/page/copyright/permissions.html.