1
|
Tankoua WLD, Nkwengoua EZT, Desiré S, Ndogo Eteme O, Tchana Satchet EM, de Araujo RSA, Nayarisseri A, de Lima MDCA, de Aquino TM, Barthélémy N, Mendonça-Junior FJB. Spectroscopic exploration of mode of binding of ctDNA and BSA with acridone alkaloids isolated from Zanthoxylum leprieurii (Rutaceae). Nat Prod Res 2024:1-15. [PMID: 39467251 DOI: 10.1080/14786419.2024.2421908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 09/30/2024] [Accepted: 10/20/2024] [Indexed: 10/30/2024]
Abstract
Zanthoxylum leprieurii is a medicinal plant widely studied due to its great phytochemical diversity, especially its acrinonic alkaloids, which have shown to be promising anticancer candidates. The aim of this work was to promote the isolation of acridonic alkaloids from fruits of Z. leprieurii and carried out absorption and fluorescence spectroscopy studies with calf thymus DNA and BSA. Five acridone alkaloids have been isolated, including the first description of 3-desmethoxy arborinine (2). In the study of interaction with biomacromolecules it was observed that all compounds show interaction with calf thymus DNA and BSA. Compound 2 promoted the bigger increase in BSA fluorescence (3.01%) with a lower fluorescence quenching constant (Ksv = 0.13 × 104). Taken together, these results reaffirm the great phytochemical diversity of Z. leprieurii, and show that acridonic alkaloids have an affinity with both DNA and BSA, therefore providing clues to their mechanisms of action related to their anticancer activities.
Collapse
Affiliation(s)
- Whistler Lucain Dibahteu Tankoua
- Laboratory of Medicinal Chemistry, Department of Organic Chemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Ernestine Zondegoumba T Nkwengoua
- Laboratory of Medicinal Chemistry, Department of Organic Chemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
- Laboratory of Synthesis and Drug Delivery, Department of Biological Science, State University of Paraiba, João Pessoa-Paraíba, Brazil
| | - Soh Desiré
- Laboratory of Medicinal Chemistry, Department of Organic Chemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Olivier Ndogo Eteme
- Laboratory of Medicinal Chemistry, Department of Organic Chemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Emmanuella Marthe Tchana Satchet
- Laboratory of Medicinal Chemistry, Department of Organic Chemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Rodrigo Santos A de Araujo
- Laboratory of Synthesis and Drug Delivery, Department of Biological Science, State University of Paraiba, João Pessoa-Paraíba, Brazil
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa-Paraíba, Brazil
| | - Anuraj Nayarisseri
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore, Madhya Pradesh India
| | | | - Thiago Mendonça de Aquino
- Grupo de Pesquisa em Estratégias Terapêuticas, Instituto de Química e Biotecnologia, Universidade Federal de Alagoas, Maceió, Brasil
| | - Nyassé Barthélémy
- Laboratory of Medicinal Chemistry, Department of Organic Chemistry, Faculty of Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Francisco J B Mendonça-Junior
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, João Pessoa-Paraíba, Brazil
- In silico Research Laboratory, Eminent Biosciences, Mahalakshmi Nagar, Indore, Madhya Pradesh India
| |
Collapse
|
2
|
Naguib BH, Elsebaie HA, Nafie MS, Mohamady S, Albujuq NR, Samir Ayed A, Nada D, Khalil AF, Hefny SM, Tawfik HO, Shaldam MA. Fragment-based design and synthesis of coumarin-based thiazoles as dual c-MET/STAT-3 inhibitors for potential antitumor agents. Bioorg Chem 2024; 151:107682. [PMID: 39137597 DOI: 10.1016/j.bioorg.2024.107682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 07/19/2024] [Accepted: 07/27/2024] [Indexed: 08/15/2024]
Abstract
c-MET and STAT-3 are significant targets for cancer treatments. Here, we describe a class of very effective dual STAT-3 and c-MET inhibitors with coumarin-based thiazoles (3a-o) as its scaffold. Spectroscopic evidence (NMR, HRMS, and HPLC) validated the structural discoveries of the new compounds. The cytotoxic activity of these compounds was also tested against a panel of cancer cells in accordance with US-NCI guidelines. Compound 3g proved to be active at 10 µM, thus it was automatically scheduled to be tested at five doses. Towards SNB-75 (CNS cancer cell line), compound 3g showed notable in vitro anti-cancer activity with GI50 = 1.43 μM. For the molecular targets, compound 3g displayed potent activity towards STAT-3 and c-MET having IC50 of 4.7 µM and 12.67, respectively, compared to Cabozantinib (IC50 = 15 nM of c-MET) and STAT-3-IN-3 (IC50 = 2.1 µM of STAT-3). Moreover, compound 3g significantly induced apoptosis in SNB-75 cells, causing a 3.04-fold increase in apoptotic cell death (treated cells exhibited 11.53 % overall apoptosis, against 3.04 % in reference cells) and a 3.58-fold increase in necrosis. Moreover, it arrests cells at the G2 phase. Dual inhibition of c-MET and STAT-3 protein kinase was further validated using RT-PCR. The target compound's binding mechanism was determined by the application of molecular docking.
Collapse
Affiliation(s)
- Bassem H Naguib
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Heba A Elsebaie
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Mohamed S Nafie
- Department of Chemistry, College of Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; Chemistry Department, Faculty of Science, Suez Canal University, Ismailia 41522, Egypt
| | - Samy Mohamady
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt; The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo 11837, Egypt
| | - Nader R Albujuq
- Department of Chemistry, School of Science, The University of Jordan, Amman 11942, Jordan
| | - Aya Samir Ayed
- Zoology Department, Faculty of Science, Suez Canal University, P.O. 41522, Ismailia, Egypt
| | - Dina Nada
- Pharmacology and Biochemistry Department, Faculty of Pharmacy, The British University, Egypt
| | - Ahmed F Khalil
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Salma M Hefny
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt
| | - Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta 31527, Egypt.
| | - Moataz A Shaldam
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, P.O. Box 33516, Kafrelsheikh, Egypt
| |
Collapse
|
3
|
Hagar FF, Abbas SH, Atef E, Abdelhamid D, Abdel-Aziz M. Benzimidazole scaffold as a potent anticancer agent with different mechanisms of action (2016-2023). Mol Divers 2024:10.1007/s11030-024-10907-8. [PMID: 39031290 DOI: 10.1007/s11030-024-10907-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Accepted: 05/30/2024] [Indexed: 07/22/2024]
Abstract
Benzimidazole scaffolds have potent anticancer activity due to their structure similarity to nucleoside. In addition, benzimidazoles could function as hydrogen donors or acceptors and bind to different drug targets that participate in cancer progression. The literature had many anticancer agents containing benzimidazole cores that gained much interest. Provoked by our endless interest in benzimidazoles as anticancer agents, we summarized the successful trials of the benzimidazole scaffolds in this concern. Moreover, we discuss the substantial opportunities in cancer treatment using benzimidazole-based drugs that may direct medicinal chemists for a compelling future design of more active chemotherapeutic agents with potential clinical applications. The uniqueness of this work lies in the highlighted benzimidazole scaffold hybridization with different molecules and benzimidazole-metal complexes, detailed mechanisms of action, and the IC50 of the developed compounds determined by different laboratories after 2015.
Collapse
Affiliation(s)
- Fatma Fouad Hagar
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| | - Samar H Abbas
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
| | - Eman Atef
- College of Pharmacy, West Coast University, Los Angeles, CA, USA
| | - Dalia Abdelhamid
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt.
- Raabe College of Pharmacy, Ohio Northern University, Ohio, USA.
| | - Mohamed Abdel-Aziz
- Medicinal Chemistry Department, Faculty of Pharmacy, Minia University, Minia, Egypt
| |
Collapse
|
4
|
Gogoi NG, Rahman A, Dutta P, Saikia J, Baruah A, Handique JG. Design, Synthesis, Biological Evaluation and in Silico Studies of Curcumin Pyrrole Conjugates. Chem Biodivers 2024; 21:e202301605. [PMID: 38488861 DOI: 10.1002/cbdv.202301605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 03/13/2024] [Indexed: 04/12/2024]
Abstract
Curcumin conjugated heterocyclic compounds are potent candidates with drug likeness against various bacterial pathogens. A set of curcumin-based pyrrole conjugates (CPs) were synthesized and characterized by FT-IR, 1H and 13C NMR and HR-MS techniques. The results of free radical scavenging activity of the synthesized CPs, evaluated by FRAP and CUPRAC assays, showed the potency of these compounds as effective antioxidants. CP3 exhibits the highest antioxidant activity amongst the CPs. The bactericidal efficacy of CPs was screened against ESKAP bacterial pathogens, and CPs were found to possess better antibacterial property than curcumin, specifically against staphylococcus aureus bacteria. In addition, serum albumin (BSA and HSA) binding interaction of these CPs were determined by UV-visible and fluorescence spectrophotometric techniques. In-silico molecular docking study was performed to determine the binding patterns of molecular targets against Staphylococcus aureus tyrosyl tRNA synthetase, and serum albumin proteins. The structure-activity relationship showed that the presence of multiple phenolic hydroxyl groups, and electron withdrawing groups on the structure of CP molecule, enhances its antioxidant and antibacterial activity, respectively.
Collapse
Affiliation(s)
- Nishi Gandha Gogoi
- Department of Chemistry, Dibrugarh University, Dibrugarh, 786004, Assam, India
- Department of Chemistry, Manohari Devi Kanoi Girls College, Dibrugarh, 786001, Assam, India
| | - Aziza Rahman
- Department of Chemistry, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Pankaj Dutta
- Department of Physics, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Jiban Saikia
- Department of Chemistry, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | - Anupaul Baruah
- Department of Chemistry, Dibrugarh University, Dibrugarh, 786004, Assam, India
| | | |
Collapse
|
5
|
Bareth D, Jain S, Kumawat J, Kishore D, Dwivedi J, Hashmi SZ. Synthetic and pharmacological developments in the hybrid s-triazine moiety: A review. Bioorg Chem 2024; 143:106971. [PMID: 38016395 DOI: 10.1016/j.bioorg.2023.106971] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/03/2023] [Accepted: 11/14/2023] [Indexed: 11/30/2023]
Abstract
This article summarizes the most recent advancements in the synthetic and pharmacological approaches along with the structure activity relationship towards the s-triazine and its derivatives. Much attention has been given to s-triazine core due to its facile synthesis, interesting pharmacology, high reactivity, and binding characteristics towards various enzymes. An array of biological applications has been demonstrated by s-triazines including antimalarial, anti-HIV, anti-viral, antimicrobial, anti-tuberculosis to name a few. In the present investigation s-triazine based molecular structures have been assembled in respect to their synthesis and medicinal properties. Further, the competence of s-triazine has been correlated and compared with the other heterocyclic moieties to substantiates-triazine a privileged scaffold. From the literature it is revealed that nucleophilic substitution at 2, 4, and 6 positions is significant for various biological applications. This article would help in assisting the chemists in designing novel molecular entities with high medicinal value.
Collapse
Affiliation(s)
- Diksha Bareth
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sonika Jain
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Jyoti Kumawat
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Dharma Kishore
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India
| | - Sonia Zeba Hashmi
- Department of Chemistry, Banasthali Vidyapith, Rajasthan 304022, India.
| |
Collapse
|
6
|
Kumawat J, Jain S, Misra N, Dwivedi J, Kishore D. 1,3,5-Triazine: Recent Development in Synthesis of its Analogs and Biological Profile. Mini Rev Med Chem 2024; 24:2019-2071. [PMID: 38847171 DOI: 10.2174/0113895575309800240526180356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 10/25/2024]
Abstract
Triazine is an important pharmacophore in the field of research for the development of novel medications due to its presence in numerous powerful physiologically active compounds with significant medical potential, such as anti-tumor, anti-viral, anti-inflammatory, anti-microbial, anti- HIV, anti-leishmanial and others. The easy availability of triazine, high reactivity, simple synthesis of their analog, and their notable broad range of biological activities have garnered chemist interest in designing s-triazine-based drugs. The interest of medicinal chemists has been sparked by the structure-activity relationship of these biologically active entities, leading to the discovery of several promising lead molecules. Its importance for medicinal chemistry research is demonstrated by the remarkable progress made with triazine derivatives in treating a variety of disorders in a very short period. Authors have collated and reviewed the medicinal potential of s-triazine analogous to afford medicinal chemists with a thorough and target-oriented overview of triazine-derived compounds. We hope the present compilation will help people from the industry and research working in the medicinal chemistry area.
Collapse
Affiliation(s)
- Jyoti Kumawat
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Sonika Jain
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Namita Misra
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Jaya Dwivedi
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| | - Dharma Kishore
- Department of Chemistry, Banasthali Vidyapith, Banasthali-304022, India
| |
Collapse
|
7
|
Dubey P, Pathak DP, Ali F, Chauhan G, Kalaiselvan V. In-vitro Evaluation of Triazine Scaffold for Anticancer Drug Development: A Review. Curr Drug Discov Technol 2024; 21:e170723218813. [PMID: 37461340 DOI: 10.2174/1570163820666230717161610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 03/27/2023] [Accepted: 05/12/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION The widespread importance of the synthesis and modification of anticancer agents has given rise to many numbers of medicinal chemistry programs. In this regard, triazine derivatives have attracted attention due to their remarkable activity against a wide range of cancer cells. This evaluation covers work reports to define the anticancer activity, the most active synthesized compound for the target, the SAR and, when described, the probable MOA besides similarly considered to deliver complete and target-pointed data for the development of types of anti-tumour medicines of triazine derivatives. Triazine scaffold for the development of anticancer analogues. Triazine can also relate to numerous beneficial targets, and their analogues have auspicious in-vitro and in-vivo anti-tumour activity. Fused molecules can improve efficacy, and drug resistance and diminish side effects, and numerous hybrid molecules are beneath diverse stages of clinical trials, so hybrid derivatives of triazine may offer valuable therapeutic involvement for the dealing of tumours. OBJECTIVE The objective of the recent review was to summarize the recent reports on triazine as well as its analogues with respect to its anticancer therapeutic potential. CONCLUSION The content of the review would be helpful to update the researchers working towards the synthesis and designing of new molecules for the treatment of various types of cancer disease with the recent molecules that have been produced from the triazine scaffold. Triazine scaffolds based on 1,3,5-triazine considerably boost molecular diversity levels and enable covering chemical space in key medicinal chemistry fields.
Collapse
Affiliation(s)
- Pragya Dubey
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Mehrauli- Badarpur Road, Sector 3, Pushp Vihar, New Delhi, 110017, India
| | - Dharam Pal Pathak
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Mehrauli- Badarpur Road, Sector 3, Pushp Vihar, New Delhi, 110017, India
| | - Faraat Ali
- Department of Analytical Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University, Akademika Heyrovského 1203, Hradec Králové 500 05, Czech Republic
- Department of Licensing and Enforcement, Laboratory Services, Botswana Medicines Regulatory Authority, Gaborone, Botswana
| | - Garima Chauhan
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Mehrauli- Badarpur Road, Sector 3, Pushp Vihar, New Delhi, 110017, India
| | - Vivekanandan Kalaiselvan
- Indian Pharmacopoeia Commission, Ministry of Health and Family Welfare, Government of India, Sector-23, Raj Nagar, Ghaziabad 201002, India
| |
Collapse
|
8
|
Gupta S, Paul K. Membrane-active substituted triazines as antibacterial agents against Staphylococcus aureus with potential for low drug resistance and broad activity. Eur J Med Chem 2023; 258:115551. [PMID: 37348297 DOI: 10.1016/j.ejmech.2023.115551] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/29/2023] [Accepted: 06/06/2023] [Indexed: 06/24/2023]
Abstract
A library of new naphthalimide-triazine analogues was synthesized as broad-spectrum antibacterial agents to overcome drug resistance. Bioactivity assay reveals that derivative 8e, with benzylamine in its structure, exhibits strong antibacterial properties against multi-drug resistance Staphylococcus aureus at a concentration of 1.56 μg/ml. It was also found to be better than chloromycin and amoxicillin. The active compound 8e efficiently inhibits the development of drug resistance within 11 passages. In addition, compound 8e inhibits the formation of biofilms in S. aureus and acts rapidly in bactericidal efficacy. Furthermore, mechanistic studies reveal that compound 8e effectively destroys the cytoplasmic membrane of bacteria, leading to leakage of intercellular protein content and loss in metabolic activity. Compound 8e binds to HSA readily with a binding constant of 1.32 × 105 M-1, indicating that the compound could be delivered to the target site effectively. Compound 8e can also form a supramolecular complex with DNA to obstruct DNA replications. These results suggest that analogue 8e could be further developed as a potential antibacterial agent. Furthermore, the cytotoxicity of all the synthesized compounds was evaluated against 60 human cancer cell lines to test their potential for anticancer agents.
Collapse
Affiliation(s)
- Saurabh Gupta
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, 147001, India
| | - Kamaldeep Paul
- School of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala, 147001, India.
| |
Collapse
|
9
|
A Review on Recent Approaches on Molecular Docking Studies of Novel Compounds Targeting Acetylcholinesterase in Alzheimer Disease. Molecules 2023; 28:molecules28031084. [PMID: 36770750 PMCID: PMC9921523 DOI: 10.3390/molecules28031084] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/09/2023] [Accepted: 01/18/2023] [Indexed: 01/24/2023] Open
Abstract
Alzheimer's disease (AD), a neurodegenerative brain disorder that affects millions of people worldwide, is characterized by memory loss and cognitive decline. Low levels of acetylcholine and abnormal levels of beta-amyloid, T protein aggregation, inflammation, and oxidative stress, have been associated with AD, and therefore, research has been oriented towards the cholinergic system and primarily on acetylcholinesterase (AChE) inhibitors. In this review, we are focusing on the discovery of AChE inhibitors using computer-based modeling and simulation techniques, covering the recent literature from 2018-2022. More specifically, the review discusses the structures of novel, potent acetylcholinesterase inhibitors and their binding mode to AChE, as well as the physicochemical requirements for the design of potential AChE inhibitors.
Collapse
|
10
|
Gandha Gogoi N, Dutta P, Saikia J, Handique JG. Antioxidant, Antibacterial, and BSA Binding Properties of Curcumin Caffeate Capped Silver Nanoparticles Prepared by Greener Method. ChemistrySelect 2022. [DOI: 10.1002/slct.202203989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Nishi Gandha Gogoi
- Department of Chemistry Dibrugarh University 786004 Dibrugarh Assam India
| | - Pankaj Dutta
- Department of Physics Dibrugarh University 786004 Dibrugarh Assam India
| | - Jiban Saikia
- Department of Chemistry Dibrugarh University 786004 Dibrugarh Assam India
| | | |
Collapse
|
11
|
Tawfik HO, Belal A, Abourehab MAS, Angeli A, Bonardi A, Supuran CT, El-Hamamsy MH. Dependence on linkers' flexibility designed for benzenesulfonamides targeting discovery of novel hCA IX inhibitors as potent anticancer agents. J Enzyme Inhib Med Chem 2022; 37:2765-2785. [PMID: 36210545 PMCID: PMC9559471 DOI: 10.1080/14756366.2022.2130285] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Herein we reported the design and synthesis of two series comprising twenty-two benzenesulfonamides that integrate the s-triazine moiety. Target compounds successfully suppressed the hCA IX, with IC50 ranging from 28.6 to 871 nM. Compounds 5d, 11b, 5b, and 7b were the most active analogues, which inhibited hCA IX isoform in the low nanomolar range (KI = 28.6, 31.9, 33.4, and 36.6 nM, respectively). Furthermore, they were assessed for their cytotoxic activity against a panel of 60 cancer cell lines following US-NCI protocol. According to five-dose assay, 13c showed significant anticancer activity than 5c with GI50-MID values of 25.08 and 189.01 µM, respectively. Additionally, 13c's effects on wound healing, cell cycle disruption, and apoptosis induction in NCI-H460 cancer cells were examined. Further, docking studies combined with molecular dynamic simulation showed a stable complex with high binding affinity of 5d to hCA IX, exploiting a favourable H-bond and lipophilic interactions.HIGHLIGHTSCarbonic anhydrase (CA) inhibitors comprising rigid and flexible linkers were developed.Compound 5d is the most potent CA IX inhibitor in the study (IC50: 28.6 nM).Compounds 5c and 13c displayed the greatest antiproliferative activity towards 60 cell lines.Compound 13c exposed constructive outcomes on normal cell lines, metastasis, and wound healing.Molecular docking and molecular dynamics (MDs) simulation was utilised to study binding mode.
Collapse
Affiliation(s)
- Haytham O. Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt,CONTACT H. O. Tawfik Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amany Belal
- Medicinal Chemistry Department, Faculty of Pharmacy, Beni-Suef University, Beni-Suef, Egypt,Department of Pharmaceutical Chemistry, College of Pharmacy, Taif University, Taif, Saudi Arabia
| | - Mohammed A. S. Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia,Department of Pharmaceutics and Industrial Pharmacy, College of Pharmacy, Minia University, Minia, Egypt
| | - Andrea Angeli
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Alessandro Bonardi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Claudiu T. Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy,C. T. Supuran Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Sesto Fiorentino, Italy
| | - Mervat H. El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
12
|
Gogoi NG, Rahman A, Saikia J, Dutta P, Baruah A, Handique JG. Enhanced biological activity of Curcumin Cinnamates: an experimental and computational analysis. Med Chem Res 2022. [DOI: 10.1007/s00044-022-02977-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
13
|
Seleno-vs. thioether triazine derivatives in search for new anticancer agents overcoming multidrug resistance in lymphoma. Eur J Med Chem 2022; 243:114761. [PMID: 36179403 DOI: 10.1016/j.ejmech.2022.114761] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/29/2022] [Accepted: 09/07/2022] [Indexed: 11/20/2022]
Abstract
Lymphomas are still difficult to treat even with modern therapies as, among others, multidrug resistance (MDR) is often counteracting a successful cancer therapy. P-gp/ABC-transporters are well-known for their crucial role in the main tumour MDR mechanism, eliminating drugs and cytotoxic substances from the cancer cell by efflux, and their modulators are promising for innovative therapy, but none has been approved in the pharmaceutical market yet. Herein, we have designed, synthesised and analysed 30 novel seleno- and thioether 1,3,5-triazine derivatives conducting comprehensive studies to evaluate their potential application in human JURKAT lymphoma cells. Among the new compounds, four (11, 12, 13 and 23) were much more effective than the reference inhibitor verapamil, being potent ABCB1 inhibitors already at 2 μM, while 5 and 15 showed very potent ABCB1 inhibitory activity only at 20 μM. Results of P-gp ATPase assays, supported with docking studies, indicated the competitive substrate mode of modulating action for 15, while ABCB1, ABCC1 and ABCG2 genes expression induction by 15 with q-PCR was confirmed. All compounds were evaluated for their cytotoxic and antiproliferative properties in both sensitive (PAR) and resistant (MDR) mouse T-lymphoma cell lines, and compound 15, also considering its promising ABCB1 inhibition properties, was revealed to be the best compound in terms of its cytotoxic effect (IC50: 16.73 μM) as well as concerning the antiproliferative effect (IC50: 5.35 μM) in MDR cells. Regarding the mechanistic studies looking at the cell cycle, the thioether 15 and selenium derivatives 26 and 29 were significantly effective in the regulation of cell cycle-related genes alone or in co-treatment with doxorubicin counteracting Cyclin D1 and E1 expression and increasing p53 and p21 levels, shedding first light on their mechanism of action. In summary, we explored the chemical space of seleno- and thioether 1,3,5-triazine derivatives with interesting activity against lymphoma. Especially compound 15 is worthy of being studied deeper to evaluate its precise mode of action further as well it can be improved regarding its potency and drug-likeness.
Collapse
|
14
|
Maliszewski D, Drozdowska D. Recent Advances in the Biological Activity of s-Triazine Core Compounds. Pharmaceuticals (Basel) 2022; 15:221. [PMID: 35215333 PMCID: PMC8875733 DOI: 10.3390/ph15020221] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/18/2022] Open
Abstract
An effective strategy for successful chemotherapy relies on creating compounds with high selectivity against cancer cells compared to normal cells and relatively low cytotoxicity. One such approach is the discovery of critical points in cancer cells, i.e., where specific enzymes that are potential therapeutic targets are generated. Triazine is a six-membered heterocyclic ring compound with three nitrogen replacing carbon-hydrogen units in the benzene ring structure. The subject of this review is the symmetrical 1,3,5-triazine, known as s-triazine. 1,3,5-triazine is one of the oldest heterocyclic compounds available. Because of its low cost and high availability, it has attracted researcher attention for novel synthesis. s-Triazine has a weak base, it has much weaker resonance energy than benzene, therefore, nucleophilic substitution is preferred to electrophilic substitution. Heterocyclic bearing a symmetrical s-triazine core represents an interesting class of compounds possessing a wide spectrum of biological properties such as anti-cancer, antiviral, fungicidal, insecticidal, bactericidal, herbicidal and antimicrobial, antimalarial agents. They also have applications as dyes, lubricants, and analytical reagents. Hence, the group of 1,3,5-triazine derivatives has developed over the years. Triazine is not only the core amongst them, but is also a factor increasing the kinetic potential of the entire derivatives. Modifying the structure and introducing new substituents makes it possible to obtain compounds with broad inhibitory activity on processes such as proliferation. In some cases, s-triazine derivatives induce cell apoptosis. In this review we will present currently investigated 1,3,5-triazine derivatives with anti-cancer activities, with particular emphasis on their inhibition of enzymes involved in the process of tumorigenesis.
Collapse
Affiliation(s)
- Dawid Maliszewski
- Department of Organic Chemistry, Medical University of Bialystok, 15-222 Białystok, Poland
| | - Danuta Drozdowska
- Department of Organic Chemistry, Medical University of Bialystok, 15-222 Białystok, Poland
| |
Collapse
|
15
|
Tawfik HO, Petreni A, Supuran CT, El-Hamamsy MH. Discovery of new carbonic anhydrase IX inhibitors as anticancer agents by toning the hydrophobic and hydrophilic rims of the active site to encounter the dual-tail approach. Eur J Med Chem 2022; 232:114190. [PMID: 35182815 DOI: 10.1016/j.ejmech.2022.114190] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/19/2022] [Accepted: 02/06/2022] [Indexed: 12/13/2022]
Abstract
The hydrophobic and the hydrophilic rims in the active site of human carbonic anhydrase IX (hCA IX) which as well contains a zinc ion as part of the catalytic core, were simultaneously matched to design and synthesize potent and selective inhibitors using a dual-tail approach. Seventeen new compounds, 5a-q, were designed to have the benzenesulfonamide moiety as a zinc binding group. In addition, N-substituted hydrazone and N-phenyl fragments were chosen as the hydrophilic and hydrophobic parts, respectively to achieve favorable interactions with the corresponding halves of the active site. All synthesized compounds successfully suppressed the CA IX, with IC50 values in nanomolar range from 13.3 to 259 nM. Compounds, 5h, 5c, 5m, 5e, and 5k were the top-five compounds efficiently inhibited the tumor-related CA IX isoform in the low nanomolar range (KI = 13.3, 22.6, 25.8, 26.9 and 27.2 nM, respectively). The target compounds 5a-q developed remarkable selectivity toward the tumor-associated isoforms (hCA IX and XII) over the off-target isoforms (hCA I and II). Furthermore, they were assessed for their anti-proliferative activity, according to US-NCI protocol, against a panel of fifty-nine cancer cell lines. Compounds 5d, 5k and 5o were passed the criteria for activity and scheduled automatically for evaluation at five concentrations with 10-fold dilutions. Compound 5k exhibited significant in vitro anticancer activity with GI50-MID; 8.68 μM compared to compounds 5d and 5o with GI50-MID; 25.76 μM and 34.97 μM respectively. The most selective compounds 5h and 5k were further screened for their in vitro cytotoxic activity against SK-MEL-5, HCC-2998 and RXF 393 cancer cell lines under hypoxic conditions. Furthermore, 5k was screened for cell cycle disturbance, apoptosis induction and intracellular reactive oxygen species (ROS) production in SK-MEL-5 cancer cells. Finally, molecular docking studies were performed to gain insights for the plausible binding interactions and affinities for selected compounds within hCA IX active site.
Collapse
Affiliation(s)
- Haytham O Tawfik
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Andrea Petreni
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019, Sesto Fiorentino, Firenze, Italy.
| | - Mervat H El-Hamamsy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
16
|
Shahedi A, Bolorizadeh MA, Karimi-Maleh H. A europium (III) complex tested for deoxyribonucleic acid-binding, bovine serum albumin binding, and antibacterial activity. J Mol Liq 2021. [DOI: 10.1016/j.molliq.2021.116323] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
17
|
Singh S, Mandal MK, Masih A, Saha A, Ghosh SK, Bhat HR, Singh UP. 1,3,5-Triazine: A versatile pharmacophore with diverse biological activities. Arch Pharm (Weinheim) 2021; 354:e2000363. [PMID: 33760298 DOI: 10.1002/ardp.202000363] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/05/2021] [Accepted: 02/12/2021] [Indexed: 12/14/2022]
Abstract
1,3,5-Triazine and its derivatives have been the epicenter of chemotherapeutic molecules due to their effective biological activities, such as antibacterial, fungicidal, antimalarial, anticancer, antiviral, antimicrobial, anti-inflammatory, antiamoebic, and antitubercular activities. The present review represents a summarized report of the crucial biological activities possessed by substituted 1,3,5-triazine derivatives, with special attention to the most potent compounds.
Collapse
Affiliation(s)
- Saumya Singh
- Drug Design and Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology, and Sciences, Allahabad, Uttar Pradesh, India
| | - Milan K Mandal
- Drug Design and Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology, and Sciences, Allahabad, Uttar Pradesh, India
| | - Anup Masih
- Drug Design and Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology, and Sciences, Allahabad, Uttar Pradesh, India
| | - Ashmita Saha
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Surajit K Ghosh
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Hans R Bhat
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Udaya P Singh
- Drug Design and Discovery Laboratory, Department of Pharmaceutical Sciences, Sam Higginbottom University of Agriculture, Technology, and Sciences, Allahabad, Uttar Pradesh, India
| |
Collapse
|
18
|
Choudhary S, Arora M, Verma H, Kumar M, Silakari O. Benzimidazole based hybrids against complex diseases: A catalogue of the SAR profile. Eur J Pharmacol 2021; 899:174027. [PMID: 33731294 DOI: 10.1016/j.ejphar.2021.174027] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 02/21/2021] [Accepted: 03/10/2021] [Indexed: 12/12/2022]
Abstract
The fused heterocyclic ring system has been recognized as a privileged structure that is used as a template in medicinal chemistry for drug discovery. Benzimidazole is one of the common scaffolds found in several natural products such as histidine, purines, and an integral part of vitamin B12. This hetero-aromatic bicyclic ring system acts as a pharmacophore in various drugs of therapeutic interest and has a broad spectrum of activity. Literature reports suggest that diversely substituted benzimidazoles possess distinct pharmacological profiles with multi-targeting potential, thereby, an indispensable anchor for the development of novel therapeutic agents against complex diseases such as cancer, malaria, inflammatory disorders, microbial diseases, hypertension, etc. Thus, lots of efforts have been diverted towards exploring the therapeutic potential of benzimidazoles. Despite great efforts made by the research community, still, some multi-factorial diseases continue to progress due to their complex pathophysiology. Under these sets of circumstances, there is a need to explore this nucleus for hybrid designing with multi-targeting potential against complex diseases. Benzimidazole-based hybrids have been reported to treat multifactorial diseases, making it a scaffold of interest for various pharmaceutical companies and research groups. In this write-up, we shed light on the recent pharmacological profiles, various designing strategies, and structure-activity relationships (SAR) of different benzimidazole-based hybrids.
Collapse
Affiliation(s)
- Shalki Choudhary
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Mohit Arora
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Himanshu Verma
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Manoj Kumar
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India
| | - Om Silakari
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab, 147002, India.
| |
Collapse
|
19
|
Chalermnon M, Cherdchom S, Sereemaspun A, Rojanathanes R, Khotavivattana T. Biguanide-Based Synthesis of 1,3,5-Triazine Derivatives with Anticancer Activity and 1,3,5-Triazine Incorporated Calcium Citrate Nanoparticles. Molecules 2021; 26:1028. [PMID: 33672071 PMCID: PMC7919653 DOI: 10.3390/molecules26041028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 02/11/2021] [Accepted: 02/12/2021] [Indexed: 11/24/2022] Open
Abstract
Twelve derivatives of biguanide-derived 1,3,5-triazines, a promising class of anticancer agent, were synthesised and evaluated for their anticancer activity against two colorectal cancer cell lines-HCT116 and SW620. 2c and 3c which are the derivatives containing o-hydroxyphenyl substituents exhibited the highest activity with IC50 against both cell lines in the range of 20-27 µM, which is comparable to the IC50 of cisplatin reference. Moreover, the potential use of the calcium citrate nanoparticles (CaCit NPs) as a platform for drug delivery system was studied on a selected 1,3,5-triazine derivative 2a. Condition optimisation revealed that the source of citrate ions and reaction time significantly influence the morphology, size and %drug loading of the particles. With the optimised conditions, "CaCit-2a NPs" were successfully synthesised with the size of 148 ± 23 nm and %drug loading of up to 16.3%. Furthermore, it was found that the release of 2a from the synthesised CaCit-2a NPs is pH-responsive, and 2a could be control released under the acidic cancer environment. The knowledge from this study is perceptive for further development of the 1,3,5-triazine-based anticancer drugs and provide the platform for the incorporation of other drugs in the CaCit NPs in the future.
Collapse
Affiliation(s)
- Monnaya Chalermnon
- Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Sarocha Cherdchom
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Phayathai Road, Wangmai, Patumwan, Bangkok 10330, Thailand;
- NanoMedicine Research Unit, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Rama 4 Road, Patumwan, Bangkok 10330, Thailand
| | - Amornpun Sereemaspun
- Chula Medical Innovation Centre (CMIC), Nanomedicine Research Unit, Department of Anatomy, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Rojrit Rojanathanes
- Centre of Excellence in Materials and Bio-Interfaces, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Tanatorn Khotavivattana
- Centre of Excellence in Natural Products Chemistry, Department of Chemistry, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
20
|
Majeed Ganai A, Khan Pathan T, Hampannavar GA, Pawar C, Obakachi VA, Kushwaha B, Deshwar Kushwaha N, Karpoormath R. Recent Advances on the s‐Triazine Scaffold with Emphasis on Synthesis, Structure‐Activity and Pharmacological Aspects: A Concise Review. ChemistrySelect 2021. [DOI: 10.1002/slct.202004591] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Ab Majeed Ganai
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Tabasum Khan Pathan
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Girish A. Hampannavar
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
- Department of Pharmaceutical Chemistry K.L.E.U's College of Pharmacy Vidyanagar, Hubli 580031, Karnataka India
| | - Chandrakant Pawar
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Vincent A. Obakachi
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Babita Kushwaha
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Narva Deshwar Kushwaha
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| | - Rajshekhar Karpoormath
- Department of Pharmaceutical Chemistry, Discipline of Pharmaceutical Sciences, College of Health Sciences University of KwaZulu-Natal (Westville) Durban 4000 South Africa
| |
Collapse
|
21
|
Singh K, Turk P, Dhanda A. Synthesis, spectral characterization, and antimicrobial evaluation of new imine derived from 3‐methylthiophene‐2‐carboxaldehyde and its Co(II), Ni(II), Cu(II), and Zn(II) metal complexes. Appl Organomet Chem 2021. [DOI: 10.1002/aoc.6088] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Kiran Singh
- Department of Chemistry Kurukshetra University Kurukshetra 136119 India
| | - Prerna Turk
- Department of Chemistry Kurukshetra University Kurukshetra 136119 India
- Government College Bherian Kurukshetra 136128 India
| | - Anita Dhanda
- Department of Microbiology Kurukshetra University Kurukshetra 136119 India
| |
Collapse
|
22
|
DNA interaction analysis with automated biosensor of paraben derivative s-triazines. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
23
|
Lang DK, Kaur R, Arora R, Saini B, Arora S. Nitrogen-Containing Heterocycles as Anticancer Agents: An Overview. Anticancer Agents Med Chem 2020; 20:2150-2168. [DOI: 10.2174/1871520620666200705214917] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/31/2020] [Accepted: 04/26/2020] [Indexed: 01/14/2023]
Abstract
Background:
Cancer is spreading all over the world, and it is becoming the leading cause of major
deaths. Today’s most difficult task for every researcher is to invent a new drug that can treat cancer with minimal
side effects. Many factors, including pollution, modern lifestyle and food habits, exposure to oncogenic
agents or radiations, enhanced industrialization, etc. can cause cancer. Treatment of cancer is done by various
methods that include chemotherapy, radiotherapy, surgery and immunotherapy in combination or singly along
with kinase inhibitors. Most of the anti-cancer drugs use the concept of kinase inhibition.
Objective:
The number of drugs being used in chemotherapy has heterocycles as their basic structure in spite of
various side effects. Medicinal chemists are focusing on nitrogen-containing heterocyclic compounds like pyrrole,
pyrrolidine, pyridine, imidazole, pyrimidines, pyrazole, indole, quinoline, oxadiazole, azole, benzimidazole,
etc. as the key building blocks to develop active biological compounds. The aim of this study is to attempt
to compile a dataset of nitrogen-containing heterocyclic anti-cancer drugs.
Methods:
We adopted a structural search on notorious journal publication websites and electronic databases
such as Bentham Science, Science Direct, PubMed, Scopus, USFDA, etc. for the collection of peer-reviewed
research and review articles for the present review. The quality papers were retrieved, studied, categorized into
different sections, analyzed and used for article writing.
Conclusion:
As per FDA databases, nitrogen-based heterocycles in the drug design are almost 60% of unique
small-molecule drugs. Some of the nitrogen-containing heterocyclic anti-cancer drugs are Axitinib, Bosutinib,
Cediranib, Dasatanib (Sprycel®), Erlotinib (Tarceva®), Gefitinib (Iressa®), Imatinib (Gleevec®), Lapatinib (Tykerb
®), Linifanib, Sorafenib (Nexavar®), Sunitinib (Sutent®), Tivozanib, etc. In the present review, we shall focus
on the overview of nitrogen-containing heterocyclic active compounds as anti-cancer agents.
Collapse
Affiliation(s)
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Rashmi Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Balraj Saini
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sandeep Arora
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
24
|
Patil V, Noonikara-Poyil A, Joshi SD, Patil SA, Patil SA, Lewis AM, Bugarin A. Synthesis, molecular docking studies, and in vitro evaluation of 1,3,5-triazine derivatives as promising antimicrobial agents. J Mol Struct 2020. [DOI: 10.1016/j.molstruc.2020.128687] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
25
|
Ali W, Spengler G, Kincses A, Nové M, Battistelli C, Latacz G, Starek M, Dąbrowska M, Honkisz-Orzechowska E, Romanelli A, Rasile MM, Szymańska E, Jacob C, Zwergel C, Handzlik J. Discovery of phenylselenoether-hydantoin hybrids as ABCB1 efflux pump modulating agents with cytotoxic and antiproliferative actions in resistant T-lymphoma. Eur J Med Chem 2020; 200:112435. [DOI: 10.1016/j.ejmech.2020.112435] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/23/2020] [Accepted: 05/06/2020] [Indexed: 02/08/2023]
|
26
|
Guo H, Diao QP. 1,3,5-Triazine-azole Hybrids and their Anticancer Activity. Curr Top Med Chem 2020; 20:1481-1492. [DOI: 10.2174/1568026620666200310122741] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 02/12/2020] [Accepted: 02/17/2020] [Indexed: 12/24/2022]
Abstract
1,3,5-Triazine and azole can interact with various therapeutic targets, and their derivatives
possess promising in vitro and in vivo anticancer activity. Hybrid molecules have the potential to enhance
efficiency, overcome drug resistance and reduce side effects, and many hybrid molecules are under
different phases of clinical trials, so hybridization of 1,3,5-triazine with azole may provide valuable
therapeutic intervention for the treatment of cancer. Substantial efforts have been made to develop
azole-containing 1,3,5-triazine hybrids as novel anticancer agents, and some of them exhibited excellent
activity. This review emphasizes azole-containing 1,3,5-triazine hybrids with potential anticancer activity,
and the structure-activity relationships as well as the mechanisms of action are also discussed to
provide comprehensive and target-oriented information for the development of this kind of anticancer
drugs.
Collapse
Affiliation(s)
- Hua Guo
- School of Chemistry and Life Science, Anshan Normal University, Anshan, Liaoning, China
| | - Quan-Ping Diao
- School of Chemistry and Life Science, Anshan Normal University, Anshan, Liaoning, China
| |
Collapse
|
27
|
Lolak N, Akocak S, Türkeş C, Taslimi P, Işık M, Beydemir Ş, Gülçin İ, Durgun M. Synthesis, characterization, inhibition effects, and molecular docking studies as acetylcholinesterase, α-glycosidase, and carbonic anhydrase inhibitors of novel benzenesulfonamides incorporating 1,3,5-triazine structural motifs. Bioorg Chem 2020; 100:103897. [PMID: 32413628 DOI: 10.1016/j.bioorg.2020.103897] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 12/12/2022]
Abstract
Some metabolic enzyme inhibitors can be used in the treatment of many diseases. Therefore, synthesis and determination of alternative inhibitors are essential. In this study, the inhibition effect of newly synthesized compounds on carbonic anhydrase (cytosolic isoforms, hCA I and hCA II), α-glycosidase (α-GLY), and acetylcholinesterase (AChE) were investigated. The possible binding mechanism of the compounds with a high inhibitory effect on the active site of the enzyme was demonstrated by molecular docking method. We investigated the inhibition effects of novel synthesized compounds (MZ1-MZ11) on metabolic enzymes such as α-GLY, AChE, and hCA I and II. The compound MZ6 for AChE, MZ8 for CA I and CA II and MZ7 for α-GLY showed a very active inhibition profile (KIs 51.67 ± 4.76 for hCA I, 40.35 ± 5.74 nM for hCA II, 41.74 ± 8.08 nM for α-GLY and 335.76 ± 46.91 nM for AChE). The novel synthesized compounds (MZ1-MZ11) have a higher enzyme (α-GLY, AChE, hCA I, and II) inhibitory potential than ACR, TAC, and AZA, respectively. The compounds may have the potential to be used as alternative medicines after further research in the treatment of many diseases such as diabetes, Alzheimer's disease, heart failure, ulcer, and epilepsy.
Collapse
Affiliation(s)
- Nebih Lolak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman 02040, Turkey
| | - Süleyman Akocak
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Adıyaman University, Adıyaman 02040, Turkey.
| | - Cüneyt Türkeş
- Department of Biochemistry, Faculty of Pharmacy, Erzincan Binali Yıldırım University, Erzincan 24100, Turkey
| | - Parham Taslimi
- Department of Biotechnology, Faculty of Science, Bartın University, Bartın 74100, Turkey
| | - Mesut Işık
- Department of Pharmacy Services, Vocational School of Health Services, Harran University, Şanlıurfa 63300, Turkey
| | - Şükrü Beydemir
- Department of Biochemistry, Faculty of Pharmacy, Anadolu University, Eskişehir 26470, Turkey
| | - İlhami Gülçin
- Department of Chemistry, Faculty of Sciences, Atatürk University, Erzurum 25240, Turkey
| | - Mustafa Durgun
- Department of Chemistry, Faculty of Arts and Sciences, Harran University, Şanlıurfa 63290, Turkey
| |
Collapse
|
28
|
Prasher P, Sharma M, Aljabali AAA, Gupta G, Negi P, Kapoor DN, Singh I, Zacconi FC, Jesus Andreoli Pinto T, Silva MW, Bakshi HA, Chellappan DK, Tambuwala MM, Dua K. Hybrid molecules based on 1,3,5‐triazine as potential therapeutics: A focused review. Drug Dev Res 2020; 81:837-858. [DOI: 10.1002/ddr.21704] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 04/25/2020] [Accepted: 05/29/2020] [Indexed: 12/30/2022]
Affiliation(s)
- Parteek Prasher
- UGC‐Sponsored Centre for Advanced Studies, Department of Chemistry Guru Nanak Dev University Amritsar India
- Department of Chemistry University of Petroleum & Energy Studies Dehradun India
| | - Mousmee Sharma
- UGC‐Sponsored Centre for Advanced Studies, Department of Chemistry Guru Nanak Dev University Amritsar India
- Department of Chemistry Uttaranchal University Dehradun India
| | - Alaa A. A. Aljabali
- Department of Pharmaceutics and Pharmaceutical Technology Faculty of Pharmacy, Yarmouk University Irbid Jordan
| | - Gaurav Gupta
- School of Pharmacy Suresh Gyan Vihar University Jaipur India
| | - Poonam Negi
- School of Pharmaceutical Sciences Shoolini University of Biotechnology and Management Sciences Solan India
| | - Deepak N. Kapoor
- School of Pharmaceutical Sciences Shoolini University of Biotechnology and Management Sciences Solan India
| | - Inderbir Singh
- Chitkara College of Pharmacy Chitkara University Punjab India
| | - Flavia C. Zacconi
- Departamento de Organica, faculdad de Quimica y de Farmacia, Pontificia Universidad Catolica de Chile Santiago Chile
| | | | - Mateus Webba Silva
- School of Pharmacy and Pharmaceutical Science Ulster University Coleraine United Kingdom
| | - Hamid A. Bakshi
- School of Pharmacy and Pharmaceutical Science Ulster University Coleraine United Kingdom
| | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy International Medical University Kuala Lumpur Malaysia
| | - Murtaza M. Tambuwala
- School of Pharmacy and Pharmaceutical Science Ulster University Coleraine United Kingdom
| | - Kamal Dua
- School of Pharmaceutical Sciences Shoolini University of Biotechnology and Management Sciences Solan India
- Discipline of Pharmacy, Graduate School of Health University of Technology Sydney Sydney New South Wales Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute (HMRI) & School of Biomedical Sciences and Pharmacy University of Newcastle Callaghan New South Wales Australia
- Centre for Inflammation, Centenary Institute Royal Prince Alfred Hospital Sydney New South Wales Australia
| |
Collapse
|
29
|
Yinhua D, Foroughi MM, Aramesh-Boroujeni Z, Jahani S, Peydayesh M, Borhani F, Khatami M, Rohani M, Dusek M, Eigner V. The synthesis, characterization, DNA/BSA/HSA interactions, molecular modeling, antibacterial properties, and in vitro cytotoxic activities of novel parent and niosome nano-encapsulated Ho(iii) complexes. RSC Adv 2020; 10:22891-22908. [PMID: 35520355 PMCID: PMC9054688 DOI: 10.1039/d0ra03436c] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/25/2020] [Indexed: 12/22/2022] Open
Abstract
Based on the importance of metal-centered complexes that can interact with DNA, this research focused on the synthesis of a new Ho(iii) complex. This complex was isolated and characterized via elemental analysis, and FT-IR, fluorescence, and UV-vis spectroscopy. Additional confirmation of the Ho(iii) complex structure was obtained via single-crystal X-ray diffraction. DNA interaction studies were carried out via circular dichroism (CD) spectroscopy, UV-vis absorption spectroscopy, viscosity measurements and emission spectroscopy; it was proposed that the metal complex acts as an effective DNA binder based on studies in the presence of fish DNA (FS-DNA), showing high binding affinity to DNA in the presence of hydrophobic and electron donating substituents. Also, the interactions of this complex with human (HSA) and bovine serum albumin (BSA) proteins were studied via fluorescence spectroscopy techniques and the obtained results reveal an excellent propensity for binding in both cases. Furthermore, the interactions of the Ho(iii) complex with DNA, BSA and HSA were confirmed via molecular docking analysis. The antimicrobial activities of the Ho(iii) complex were tested against Gram-negative bacteria and Gram-positive bacteria. In addition, a niosome nano-encapsulated Ho(iii) complex was synthesized, and the parent and encapsulated complexes were evaluated as potential antitumor candidates. The main structure of the Ho(iii) complex is maintained after encapsulation using niosome nanoparticles. The MTT method was used to assess the anticancer properties of the Ho(iii) complex and its encapsulated form toward human lung carcinoma and breast cancer cell lines. The anticancer activity in the encapsulated form was more than that of the parent Ho(iii) complex. In conclusion, these compounds could be considered as new antitumor candidates.
Collapse
Affiliation(s)
- Deng Yinhua
- Department of Pharmacy, Hunan Provincial People's Hospital Changsha 410005 P. R. China
- Department of Pharmacy, The First Hospital Affiliated to Hunan Normal University Changsha 410005 P. R. China
| | | | - Zahra Aramesh-Boroujeni
- Department of Clinical Laboratory, AlZahra Hospital, Isfahan University of Medical Sciences Isfahan Iran
| | - Shohreh Jahani
- Nanobioelectrochemistry Research Center, Bam University of Medical Sciences Bam Iran +98 35331321750
| | - Mohadesh Peydayesh
- Clinical Research Center, Pastor Educational Hospital, Bam University of Medical Sciences Bam Iran
| | - Fariba Borhani
- Medical Ethics and Law Research Center, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mehrdad Khatami
- Nanobioelectrochemistry Research Center, Bam University of Medical Sciences Bam Iran +98 35331321750
- Cell Therapy and Regenerative Medicine Comprehensive Center, Kerman University of Medical Sciences Kerman Iran
| | - Meysam Rohani
- Department of Medicine, Medical School, Bam University of Medical Sciences Bam Iran
| | - Michal Dusek
- Institute of Physics of the Czech Academy of Sciences Na Slovance 2 18221 Prague 8 Czech Republic
| | - Vaclav Eigner
- Institute of Physics of the Czech Academy of Sciences Na Slovance 2 18221 Prague 8 Czech Republic
| |
Collapse
|
30
|
Investigations of antiproliferative and antioxidant activity of β-lactam morpholino-1,3,5-triazine hybrids. Bioorg Med Chem 2020; 28:115408. [PMID: 32165076 DOI: 10.1016/j.bmc.2020.115408] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 02/20/2020] [Accepted: 02/29/2020] [Indexed: 12/13/2022]
Abstract
This article reports for the first time the synthesis of some novel β-lactam morpholino-1,3,5-triazine hybrids by a [2+2]-cycloaddition reaction of imines 7a-c, 9a-c and 11 with ketenes derived from substituted acetic acids. The reaction was totally diastereoselective, leading exclusively to the formation of cis-β-lactams 8a-l, 10a-f and 12a-c. The synthesized compounds were tested for activity towards SW1116, MCF-7 and HepG2 cancer cell lines and non-cancerous HEK-293 cell line by MTT assay. None of the compounds exert an observable effect on HepG2, MCF-7 and HEK-293 cells, but compounds 7b, 8f, 8g, 8l, 10c, and 10e exhibited excellent growth inhibitory activity (IC50 < 5 µM) against SW 1116 cells, comparable to that of doxorubicin (IC50 = 6.9 µM). An evaluation of the antioxidant potential of each of the compounds, performed by diphenylpicrylhydrazyl (DPPH) assay, indicated that 7b, 9a, 9b and 9c have strong free radical scavenging activity. UV absorption titration studies reveal that 7b, 8l, 8g and 8f interact strongly with calf-thymus DNA (CT-DNA) in the order of 8l > 7b > 8f > 8g. Collectively, the in vitro capabilities of some of these morpholino-triazine imines and β-lactams suggest possible applications to development of new antioxidants and DNA binding therapeutics.
Collapse
|
31
|
Cascioferro S, Petri GL, Parrino B, Carbone D, Funel N, Bergonzini C, Mantini G, Dekker H, Geerke D, Peters GJ, Cirrincione G, Giovannetti E, Diana P. Imidazo[2,1-b] [1,3,4]thiadiazoles with antiproliferative activity against primary and gemcitabine-resistant pancreatic cancer cells. Eur J Med Chem 2020; 189:112088. [PMID: 32007666 DOI: 10.1016/j.ejmech.2020.112088] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/31/2022]
Abstract
A new series of eighteen imidazo [2,1-b] [1,3,4]thiadiazole derivatives was efficiently synthesized and screened for antiproliferative activity against the National Cancer Institute (NCI-60) cell lines panel. Two out of eighteen derivatives, compounds 12a and 12h, showed remarkably cytotoxic activity with the half maximal inhibitory concentration values (IC50) ranging from 0.23 to 11.4 μM, and 0.29-12.2 μM, respectively. However, two additional compounds, 12b and 13g, displayed remarkable in vitro antiproliferative activity against pancreatic ductal adenocarcinoma (PDAC) cell lines, including immortalized (SUIT-2, Capan-1, Panc-1), primary (PDAC-3) and gemcitabine-resistant (Panc-1R), eliciting IC50 values ranging from micromolar to sub-micromolar level, associated with significant reduction of cell-migration and spheroid shrinkage. These remarkable results might be explained by modulation of key regulators of epithelial-to-mesenchymal transition (EMT), including E-cadherin and vimentin, and inhibition of metalloproteinase-2/-9. High-throughput arrays revealed a significant inhibition of the phosphorylation of 45 tyrosine kinases substrates, whose visualization on Cytoscape highlighted PTK2/FAK as an important hub. Inhibition of phosphorylation of PTK2/FAK was validated as one of the possible mechanisms of action, using a specific ELISA. In conclusion, novel imidazothiadiazoles show potent antiproliferative activity, mediated by modulation of EMT and PTK2/FAK.
Collapse
Affiliation(s)
- Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Giovanna Li Petri
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy; Department of Medical Oncology, Amsterdam University Medical Center, VU University Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Daniela Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Niccola Funel
- Unit of Anatomic Pathology II, Azienda Ospedaliero-Universitaria Pisana, Via Roma 67, 56126, Pisa, Italy
| | - Cecilia Bergonzini
- Department of Medical Oncology, Amsterdam University Medical Center, VU University Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Giulia Mantini
- Department of Medical Oncology, Amsterdam University Medical Center, VU University Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Henk Dekker
- Department of Medical Oncology, Amsterdam University Medical Center, VU University Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Daan Geerke
- AIMMS Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, Faculty of Sciences, VU University Amsterdam, De Boelelaan 1108, 1081 HZ, Amsterdam, the Netherlands
| | - Godefridus J Peters
- Department of Medical Oncology, Amsterdam University Medical Center, VU University Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy
| | - Elisa Giovannetti
- Department of Medical Oncology, Amsterdam University Medical Center, VU University Cancer Center Amsterdam, De Boelelaan 1117, 1081HV, Amsterdam, the Netherlands; Fondazione Pisana per la Scienza, Via Ferruccio Giovannini 13, 56017, San Giuliano Terme, Pisa, Italy.
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123, Palermo, Italy.
| |
Collapse
|
32
|
Reddy MVK, Anusha G, Reddy PVG. Sterically enriched bulky 1,3-bis(N,N′-aralkyl)benzimidazolium based Pd-PEPPSI complexes for Buchwald–Hartwig amination reactions. NEW J CHEM 2020. [DOI: 10.1039/d0nj01294g] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
A simple and efficient synthesis of a series of unexisting Pd-PEPPSI complexes is summarized. These complexes are exploited for their high catalytic activity towards Buchwald–Hartwig amination.
Collapse
|
33
|
Ribeiro AG, Almeida SMVD, de Oliveira JF, Souza TRCDL, Santos KLD, Albuquerque APDB, Nogueira MCDBL, Carvalho Junior LBD, Moura ROD, da Silva AC, Pereira VRA, Castro MCABD, Lima MDCAD. Novel 4-quinoline-thiosemicarbazone derivatives: Synthesis, antiproliferative activity, in vitro and in silico biomacromolecule interaction studies and topoisomerase inhibition. Eur J Med Chem 2019; 182:111592. [DOI: 10.1016/j.ejmech.2019.111592] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Revised: 07/07/2019] [Accepted: 08/05/2019] [Indexed: 12/16/2022]
|
34
|
Abstract
Background:
This review presents the exhaustive exploration of 1,3,5-triazine scaffold
for development of analogs of anticancer drugs, over the last century. In the recent years, striazine
moiety has been one of the most studied moiety, showing broad-spectrum pharmacological
activities such as antibacterial, antifungal, analgesic, anti-HIV, antileishmanial, antitrypanosomal,
antimalarial and antiviral. Nowadays, many boffins are have become interested in novel
synthesis of s-triazine derivatives because of low cost and ease of availability.
Methods:
This scaffold has been extensively investigated mainly in the past decade. Many products
have been synthesized from different starting materials and these synthetic products possess
anticancer potential against various cell lines.
Results:
Many 1,3,5-triazine analogs exhibited significant anticancer activity in various models
and cell lines exhibiting different mechanisms. Some analogs have also shown good pharmacokinetic
parameters with less IC50 values.
Conclusion:
Various 1,3,5-triazine analogs have shown potent activities and may be regarded as
clinical candidates for future anticancer formulations. This review may be helpful to those researchers
seeking required information with regard to the drug design and medicinal properties of
1,3,5-triazine derivatives for selected targets. This review may also offer help to find and improve
clinically viable anticancer molecules.
Collapse
Affiliation(s)
- Rajeev Kumar
- Devsthali Vidyapeeth College of Pharmacy, Lalpur, Rudrapur (U.S. Nagar)-263148, Uttarakhand, India
| | - Neeraj Kumar
- Devsthali Vidyapeeth College of Pharmacy, Lalpur, Rudrapur (U.S. Nagar)-263148, Uttarakhand, India
| | | | - Anita Singh
- Department of Pharmacy, Kumaun University, Bhimtal, Nainital-263136, Uttarakhand, India
| |
Collapse
|
35
|
Ghosh R, Bharathkar SK, Kishore N. Anticancer altretamine recognition by bovine serum albumin and its role as inhibitor of fibril formation: Biophysical insights. Int J Biol Macromol 2019; 138:359-369. [DOI: 10.1016/j.ijbiomac.2019.07.093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 07/08/2019] [Accepted: 07/13/2019] [Indexed: 01/15/2023]
|
36
|
da Silva Filho FA, de Freitas Souza T, Ribeiro AG, Alves JEF, de Oliveira JF, de Lima Souza TRC, de Moura RO, do Carmo Alves de Lima M, de Carvalho Junior LB, de Almeida SMV. Topoisomerase inhibition and albumin interaction studies of acridine-thiosemicarbazone derivatives. Int J Biol Macromol 2019; 138:582-589. [PMID: 31323270 DOI: 10.1016/j.ijbiomac.2019.07.097] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 06/24/2019] [Accepted: 07/15/2019] [Indexed: 10/26/2022]
Abstract
In the present study, acridine-thiosemicarbazones (ATD) derivatives were tested for their interaction properties with BSA through UV-Vis absorption and fluorescence spectroscopic studies. Both hyperchromic and hypochromic effects, as well as red or blue shifts were demonstrated after the derivatives were added to the BSA. Values for the binding constant (Kb) ranged from 1.62 × 104 to 8.71 × 105 M-1 and quenching constant (KSV) from 3.46 × 102 to 7.83 × 103 M-1 indicating a good affinity to BSA protein. Complementary, two compounds were selected to assess their inhibition activity against topoisomerase IIα enzyme, of which derivative 3a presented the best result. Moreover, to evaluate protein-ligand interactions, as well as the antitopoisomerase potential of these compounds, tests of molecular modeling were performed between all compounds using the albumin and Topoisomerase IIα/DNA complex. Finally, in silico studies showed that all derivatives used in this research displayed good oral bioavailability potential.
Collapse
Affiliation(s)
- Francivaldo Araújo da Silva Filho
- Universidade de Pernambuco (UPE), campus Garanhuns, Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Garanhuns, PE, Brazil
| | - Thais de Freitas Souza
- Universidade de Pernambuco (UPE), campus Garanhuns, Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Garanhuns, PE, Brazil
| | - Amélia Galdino Ribeiro
- Laboratório de Química e Inovação Terapêutica (LQIT), Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Jamerson Ferreira de Oliveira
- Laboratório de Química e Inovação Terapêutica (LQIT), Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Ricardo Olímpio de Moura
- Departamento de Ciências Farmacêuticas, Centro de Ciências Biológicas e da Saúde, Universidade Estadual da Paraíba - Bodocongo, Campina Grande, PB, Brazil
| | - Maria do Carmo Alves de Lima
- Laboratório de Química e Inovação Terapêutica (LQIT), Departamento de Antibióticos, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | | | - Sinara Mônica Vitalino de Almeida
- Universidade de Pernambuco (UPE), campus Garanhuns, Faculdade de Ciências, Educação e Tecnologia de Garanhuns (FACETEG), Garanhuns, PE, Brazil; Laboratório de Imunopatologia Keizo Asami (LIKA), Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
37
|
Pricopie AI, Ionuț I, Marc G, Arseniu AM, Vlase L, Grozav A, Găină LI, Vodnar DC, Pîrnău A, Tiperciuc B, Oniga O. Design and Synthesis of Novel 1,3-Thiazole and 2-Hydrazinyl-1,3-Thiazole Derivatives as Anti- Candida Agents: In Vitro Antifungal Screening, Molecular Docking Study, and Spectroscopic Investigation of their Binding Interaction with Bovine Serum Albumin. Molecules 2019; 24:E3435. [PMID: 31546673 PMCID: PMC6804233 DOI: 10.3390/molecules24193435] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 08/18/2019] [Accepted: 09/20/2019] [Indexed: 02/03/2023] Open
Abstract
In the context of there being a limited number of clinically approved drugs for the treatment of Candida sp.-based infections, along with the rapid development of resistance to the existing antifungals, two novel series of 4-phenyl-1,3-thiazole and 2-hydrazinyl-4-phenyl-1,3-thiazole derivatives were synthesized and tested in vitro for their anti-Candida potential. Two compounds (7a and 7e) showed promising inhibitory activity against the pathogenic C. albicans strain, exhibiting substantially lower MIC values (7.81 μg/mL and 3.9 μg/mL, respectively) as compared with the reference drug fluconazole (15.62 μg/mL). Their anti-Candida activity is also supported by molecular docking studies, using the fungal lanosterol C14α-demethylase as the target enzyme. The interaction of the most biologically active synthesized compound 7e with bovine serum albumin was investigated through fluorescence spectroscopy, and the obtained data suggested that this molecule might efficiently bind carrier proteins in vivo in order to reach the target site.
Collapse
Affiliation(s)
- Andreea-Iulia Pricopie
- Department of Pharmaceutical Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| | - Ioana Ionuț
- Department of Pharmaceutical Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| | - Gabriel Marc
- Department of Pharmaceutical Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| | - Anca-Maria Arseniu
- Department of Pharmaceutical Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
- Preclinic Department, Pharmacy Specialization, Faculty of Medicine, Lucian Blaga University of Sibiu, 2A Lucian Blaga Street, 550169 Sibiu, Romania.
| | - Laurian Vlase
- Department of Pharmaceutical Technology and Biopharmaceutics, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| | - Adriana Grozav
- Department of Organic Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| | - Luiza Ioana Găină
- Research Center on Fundamental and Applied Heterochemistry, Faculty of Chemistry and Chemical Engineering, "Babeş-Bolyai" University, 11 Arany Janos Street, 400028 Cluj-Napoca, Romania.
| | - Dan C Vodnar
- Department of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, 3-5 Mănăștur Street, 400372 Cluj-Napoca, Romania.
| | - Adrian Pîrnău
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donath Street, 400293 Cluj-Napoca, Romania.
| | - Brîndușa Tiperciuc
- Department of Pharmaceutical Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| | - Ovidiu Oniga
- Department of Pharmaceutical Chemistry, "Iuliu Hațieganu" University of Medicine and Pharmacy, 41 Victor Babeș Street, 400012 Cluj-Napoca, Romania.
| |
Collapse
|
38
|
Reddy SMK, Kothandapani J, Sengan M, Veerappan A, Selva Ganesan S. Exploring the influence of designer surfactant hydrophobicity in key C C/C N bond forming reactions. MOLECULAR CATALYSIS 2019. [DOI: 10.1016/j.mcat.2019.01.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
39
|
Tang X, Su S, Chen M, He J, Xia R, Guo T, Chen Y, Zhang C, Wang J, Xue W. Novel chalcone derivatives containing a 1,2,4-triazine moiety: design, synthesis, antibacterial and antiviral activities. RSC Adv 2019; 9:6011-6020. [PMID: 35517271 PMCID: PMC9060900 DOI: 10.1039/c9ra00618d] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/06/2019] [Indexed: 11/21/2022] Open
Abstract
A series of novel chalcone derivatives containing the 1,2,4-triazine moiety were synthesized and their structures were confirmed by 1H NMR, 13C NMR and elemental analyses. Antiviral bioassays revealed that most of the compounds exhibited good antiviral activity against tobacco mosaic virus (TMV) at a concentration of 500 μg mL-1. The designated compound 4l was 50% effective in terms of curative and protective activities against TMV with 50% effective concentrations (EC50) of 10.9 and 79.4 μg mL-1, which were better than those of ningnanmycin (81.4 and 82.2 μg mL-1). Microscale thermophoresis (MST) also showed that the binding of compound 4l to coat protein (TMV-CP) yielded a K d value of 0.275 ± 0.160 μmol L-1, which was better than that of ningnanmycin (0.523 ± 0.250 μmol L-1). At the same time, molecular docking studies for 4l with TMV-CP (PDB code:1EI7) showed that the compound was embedded well in the pocket between the two subunits of TMV-CP. Meanwhile, compound 4a demonstrated excellent antibacterial activities against Ralstonia solanacearum (R. solanacearum), with an EC50 value of 0.1 μg mL-1, which was better than that of thiodiazole-copper (36.1 μg mL-1) and bismerthiazol (49.5 μg mL-1). The compounds act by causing folding and deformation of the bacterial cell membrane as observed using scanning electron microscopy (SEM). The chalcone derivatives thus synthesized could become potential alternative templates for novel antiviral and antibacterial agents.
Collapse
Affiliation(s)
- Xu Tang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Shijun Su
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Mei Chen
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Jun He
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Rongjiao Xia
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Tao Guo
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Ying Chen
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Cheng Zhang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Jun Wang
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| | - Wei Xue
- State Key Laboratory Breeding Base of Green Pesticide and Agricultural Bioengineering, Key Laboratory of Green Pesticide and Agricultural Bioengineering, Ministry of Education, Center for Research and Development of Fine Chemicals, Guizhou University Guiyang 550025 China +86-851-88292090 +86-851-88292090
| |
Collapse
|
40
|
Singh I, Rani R, Luxami V, Paul K. Synthesis of 5-(4-(1H-phenanthro[9,10-d]imidazol-2-yl)benzylidene)thiazolidine-2,4-dione as promising DNA and serum albumin-binding agents and evaluation of antitumor activity. Eur J Med Chem 2019; 166:267-280. [PMID: 30721822 DOI: 10.1016/j.ejmech.2019.01.053] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 01/19/2019] [Accepted: 01/21/2019] [Indexed: 10/27/2022]
Abstract
A series of phenanthro[9,10-d]imidazole/oxazole and acenaphtho[1,2-d]imidazole with different aryl groups at C2-position has been synthesized. These compounds were in vitro evaluated for antitumor activity against a panel of 60 human cancer cell lines. Compound 8 exhibits higher cytotoxicity towards leukemia, colon, melanoma, renal, and breast cancer cell lines than the other evaluated cell panels and low toxicity against normal cell line Hek293. The binding properties of compound 8 with DNA have been investigated with absorption, emission and circular dichroism as well as thermal denaturation experiments which indicate intercalation with base pairs of human and calf thymus DNA. The molecular docking and site-selective binding studies also reveal the predominant intercalation of compound 8 in base pairs of DNA. The interaction between thiazolidine based phenanthrene 8 and serum albumins (HSA and BSA), transport proteins, has also been explored which shows quenching of fluorescence through static mechanism. The thermodynamic parameters, obtained from van't Hoff relationship indicate the prevalence of hydrogen-bonding/hydrophobic interactions for the binding phenomenon.
Collapse
Affiliation(s)
- Iqubal Singh
- School of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala 147 001, India
| | - Richa Rani
- School of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala 147 001, India
| | - Vijay Luxami
- School of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala 147 001, India
| | - Kamaldeep Paul
- School of Chemistry and Biochemistry, Thapar Institute of Engineering & Technology, Patiala 147 001, India.
| |
Collapse
|
41
|
Gouveia RG, Ribeiro AG, Segundo MÂSP, de Oliveira JF, de Lima MDCA, de Lima Souza TRC, de Almeida SMV, de Moura RO. Synthesis, DNA and protein interactions and human topoisomerase inhibition of novel Spiroacridine derivatives. Bioorg Med Chem 2018; 26:5911-5921. [DOI: 10.1016/j.bmc.2018.10.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 12/20/2022]
|
42
|
Kumar B, K S, Chandra, Kaur M, Jasinski JP, Revanasiddappa H. 4-Chloro-2-(5,6-dihydro-benzo[4,5]imidazo[1,2-c]quinazolin-6-yl)-phenol (HL); synthesis, characterization, crystal structure, Hirshfeld surface analysis and BSA binding studies. J Mol Struct 2018. [DOI: 10.1016/j.molstruc.2018.07.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
43
|
Synthesis and PI3 Kinase Inhibition Activity of Some Novel Trisubstituted Morpholinopyrimidines. Molecules 2018; 23:molecules23071675. [PMID: 29996482 PMCID: PMC6100461 DOI: 10.3390/molecules23071675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Revised: 07/05/2018] [Accepted: 07/07/2018] [Indexed: 11/17/2022] Open
Abstract
A number of new substituted morpholinopyrimidines were prepared utilizing sequential nucleophilic aromatic substitution and cross-coupling reactions. One of the disubstituted pyrimidines was converted into two trisubstituted compounds which were screened as PI3K inhibitors relative to the well-characterized PI3K inhibitor ZSTK474, and were found to be 1.5⁻3-times more potent. A leucine linker was attached to the most active inhibitor since it would remain on any peptide-containing prodrug after cleavage by prostate-specific antigen, and it did not prevent inhibition of AKT phosphorylation and hence the inhibition of PI3K by the modified inhibitor.
Collapse
|
44
|
Synthesis and PI 3-Kinase Inhibition Activity of Some Novel 2,4,6-Trisubstituted 1,3,5-Triazines. Molecules 2018; 23:molecules23071628. [PMID: 29973512 PMCID: PMC6100378 DOI: 10.3390/molecules23071628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 01/20/2023] Open
Abstract
A number of new trisubstituted triazine phosphatidylinositol 3-kinase (PI3K) inhibitors were prepared via a three-step procedure utilizing sequential nucleophilic aromatic substitution and cross-coupling reactions. All were screened as PI3K inhibitors relative to the well-characterized PI3K inhibitor, ZSTK474. The most active inhibitors prepared here were 2–4 times more potent than ZSTK474. A leucine linker was attached to the most active inhibitor since it would remain on any peptide-containing prodrug after cleavage by a prostate-specific antigen, and it did not prevent inhibition of protein kinase B (Akt) phosphorylation, and hence, the inhibition of PI3K by the modified inhibitor.
Collapse
|
45
|
Unravelling the binding mechanism of benproperine with human serum albumin: A docking, fluorometric, and thermodynamic approach. Eur J Med Chem 2018; 146:245-250. [DOI: 10.1016/j.ejmech.2018.01.064] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 01/01/2023]
|
46
|
Basu A, Bhayye S, Kundu S, Das A, Mukherjee A. Andrographolide inhibits human serum albumin fibril formations through site-specific molecular interactions. RSC Adv 2018; 8:30717-30724. [PMID: 35548768 PMCID: PMC9085492 DOI: 10.1039/c8ra04637a] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 08/03/2018] [Indexed: 01/13/2023] Open
Abstract
Protein misfolding and fibrillation are the fundamental traits in degenerative diseases like Alzheimer's, Parkinsonism, and diabetes mellitus. Bioactives such as flavonoids and terpenoids from plant sources are known to express protective effects against an array of diseases including diabetes, Alzheimer's and obesity. Andrographolide (AG), a labdane diterpenoid is prescribed widely in the Indian and Chinese health care systems for classical efficacy against a number of degenerative diseases. This work presents an in depth study on the effects of AG on protein fibrillating pathophysiology. Thioflavin T fluorescence spectroscopy and DLS results indicated concentration dependent inhibition of human serum albumin (HSA) fibrillation. The results were confirmed by electron microscopy studies. HSA fibril formations were markedly reduced in the presence of AG. Fluorescence studies and UV-Vis experiments confirmed further that AG molecularly interacts with HSA at site. In silico molecular docking studies revealed hydrogen bonding and hydrophobic interactions with HSA in the native state. Thus AG interacts with HSA, stabilizes the native protein structure and inhibits fibrillation. The results demonstrated that the compound possesses anti-amyloidogenic properties and can be promising against some human degenerative diseases. Andrographolide inhibited HSA protein fibrillation through site specific interactions.![]()
Collapse
Affiliation(s)
- Aalok Basu
- Division of Pharmaceutical and Fine Chemical Technology
- Department of Chemical Technology
- University of Calcutta
- Kolkata 700009
- India
| | - Sagar Bhayye
- Division of Pharmaceutical and Fine Chemical Technology
- Department of Chemical Technology
- University of Calcutta
- Kolkata 700009
- India
| | - Sonia Kundu
- Division of Pharmaceutical and Fine Chemical Technology
- Department of Chemical Technology
- University of Calcutta
- Kolkata 700009
- India
| | - Aatryee Das
- Division of Pharmaceutical and Fine Chemical Technology
- Department of Chemical Technology
- University of Calcutta
- Kolkata 700009
- India
| | - Arup Mukherjee
- Division of Pharmaceutical and Fine Chemical Technology
- Department of Chemical Technology
- University of Calcutta
- Kolkata 700009
- India
| |
Collapse
|
47
|
Cascioferro S, Parrino B, Spanò V, Carbone A, Montalbano A, Barraja P, Diana P, Cirrincione G. 1,3,5-Triazines: A promising scaffold for anticancer drugs development. Eur J Med Chem 2017; 142:523-549. [PMID: 29046238 DOI: 10.1016/j.ejmech.2017.09.035] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/16/2017] [Accepted: 09/18/2017] [Indexed: 12/18/2022]
Abstract
This review covering literature reports from the beginning of this century to 2016 describes the synthetic pathways, the antitumor activity, the structure-activity relationship and, whenever reported, the possible mechanism of action of 1,3,5-triazine derivatives as well as of their hetero-fused compounds. Many 1,3,5-triazine derivatives, both uncondensed and hetero-fused, have shown remarkable antitumor activities and some of them reached clinical development.
Collapse
Affiliation(s)
- Stella Cascioferro
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Barbara Parrino
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Virginia Spanò
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Carbone
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Alessandra Montalbano
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Paola Barraja
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Patrizia Diana
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Girolamo Cirrincione
- Dipartimento di Scienze e Tecnologie Biologiche Chimiche e Farmaceutiche (STEBICEF), Università degli Studi di Palermo, Via Archirafi 32, 90123 Palermo, Italy.
| |
Collapse
|
48
|
Recent advances (2015-2016) in anticancer hybrids. Eur J Med Chem 2017; 142:179-212. [PMID: 28760313 DOI: 10.1016/j.ejmech.2017.07.033] [Citation(s) in RCA: 189] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 07/13/2017] [Accepted: 07/18/2017] [Indexed: 12/26/2022]
Abstract
In spite of the development of a large number of novel anticancer drugs over the years, Cancer remains as a prominent cause of death, worldwide. Numerous drugs that are currently in clinical practice have developed multidrug resistance along with fatal side effects. Therefore, the utilization of single-target therapy is incapable of providing an effective control on the malignant process. Molecular hybridization, involving a combination of two or more pharmacophores of bioactive scaffolds to generate a single molecular architecture with improved affinity and activity, in comparison to their parent molecules, has emerged as a promising strategy in recent drug discovery research. Hybrid anticancer drugs are of great therapeutic interests since they can potentially overcome most of the pharmacokinetic drawbacks encountered with conventional anticancer drugs. Strategically, the design of anticancer drugs involved the blending or linking of an anticancer drug with another anticancer drug or a carrier molecule which can efficiently target cancer cells with improved biological potential. Major advantages of hybrid anticancer drugs involved increased specificity, better patient compliance, and lower side effects along with reduction in chemo-resistance. The successful utilization of this technique in design and synthesis of novel anticancer hybrids has been well illustrated and documented in the literature. The purpose of the present review article will be to provide an emphasis on the recent developments (2015-16) in anticancer hybrids with insights into their structure-activity relationship (SAR) and mechanism of action.
Collapse
|
49
|
Fu DJ, Song J, Hou YH, Zhao RH, Li JH, Mao RW, Yang JJ, Li P, Zi XL, Li ZH, Zhang QQ, Wang FY, Zhang SY, Zhang YB, Liu HM. Discovery of 5,6-diaryl-1,2,4-triazines hybrids as potential apoptosis inducers. Eur J Med Chem 2017; 138:1076-1088. [PMID: 28763643 DOI: 10.1016/j.ejmech.2017.07.011] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 07/04/2017] [Accepted: 07/07/2017] [Indexed: 12/18/2022]
Abstract
A series of 5,6-diaryl-1,2,4-triazines hybrids bearing a 1,2,3-triazole linker were synthesized by molecular hybridization strategy and evaluated for antiproliferative activity against three selected cancer cell lines (MGC-803, EC-109 and PC-3). The first structure-activity relationship (SAR) for these 5,6-diaryl-1,2,4-triazines is explored in this report with evaluation of 15 variants of the structural class. Among these chemical derivatives, 3-(((1-(4-fluorobenzyl)-1H-1,2,3-triazol-4-yl)methyl)thio)-5,6-diphenyl-1,2,4-triazine (11E) showed the more potent inhibitory effect against three cell lines than 5-Fu. Cellular mechanism studies in MGC-803 cells elucidated 11E inhibited colony formation and arrested cell cycle at G2/M phase. Furthermore, compound 11E caused morphological changes, decreased mitochondrial membrane potential, and induced apoptosis through the apoptosis-related proteins in MGC-803 cells. It was the first time, to our knowledge, that 5,6-diaryl-1,2,4-triazines bearing a 1,2,3-triazole linker were used as potential apoptosis inducers.
Collapse
Affiliation(s)
- Dong-Jun Fu
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Jian Song
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Yu-Hui Hou
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Ruo-Han Zhao
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Jia-Huan Li
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Ruo-Wang Mao
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Jia-Jia Yang
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Ping Li
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Xiao-Lin Zi
- Pathology and Laboratory Medicine, University of California, Irvine, Orange, CA 92868, USA
| | - Zhong-Hua Li
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Qing-Qing Zhang
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Fei-Yan Wang
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China.
| | - Yan-Bing Zhang
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China.
| | - Hong-Min Liu
- New Drug Research & Development Center, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, China; Key Laboratory of Technology of Drug Preparation, Zhengzhou University, Ministry of Education, China; Key Laboratory of Henan Province for Drug Quality and Evaluation, China.
| |
Collapse
|
50
|
Shrivastava N, Naim MJ, Alam MJ, Nawaz F, Ahmed S, Alam O. Benzimidazole Scaffold as Anticancer Agent: Synthetic Approaches and Structure-Activity Relationship. Arch Pharm (Weinheim) 2017; 350. [PMID: 28544162 DOI: 10.1002/ardp.201700040] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Revised: 04/22/2017] [Accepted: 04/25/2017] [Indexed: 11/07/2022]
Abstract
Cancer, also known as malignant neoplasm, is a dreadful disease which involves abnormal cell growth having the potential to invade or spread to other parts of the body. Benzimidazole is an organic compound that is heterocyclic and aromatic in nature. It is a bicyclic compound formed by the fusion of the benzene and imidazole ring systems. It is an important pharmacophore and a privileged structure in medicinal chemistry. According to the World Health Organisation (2015 survey), one in six deaths is due to cancer around the globe, accounting for 8.8 million deaths of which 70% of the cases were from low- and middle-income countries. In the efforts to develop suitable anticancer drugs, medicinal chemists have focussed on benzimidazole derivatives. This review article covers the current development of benzimidazole-based anticancer agents along with the synthetic approaches and structure-activity relationships (SAR).
Collapse
Affiliation(s)
- Neelima Shrivastava
- Faculty of Pharmacy, , Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| | - Mohd Javed Naim
- Faculty of Pharmacy, , Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| | - Md Jahangir Alam
- Faculty of Pharmacy, , Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| | - Farah Nawaz
- Faculty of Pharmacy, , Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| | - Shujauddin Ahmed
- Faculty of Pharmacy, , Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| | - Ozair Alam
- Faculty of Pharmacy, , Department of Pharmaceutical Chemistry, Jamia Hamdard, New Delhi, India
| |
Collapse
|