1
|
Batheja S, Gupta S, Tejavath KK, Gupta U. TPP-based conjugates: potential targeting ligands. Drug Discov Today 2024; 29:103983. [PMID: 38641237 DOI: 10.1016/j.drudis.2024.103983] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/21/2024]
Abstract
Mitochondria are one of the major sources of energy as well as regulators of cancer cell metabolism. Thus, they are potential targets for the effective treatment and management of cancer. Research has explored triphenylphosphonium (TPP) derivatives as potent cancer-targeting ligands because of their lipophilic nature and mitochondrial affinity. In this review, we summarize the utility of TPP-based conjugates targeting mitochondria in different types of cancer and other diseases, such as neurodegenerative and cardiovascular disorders. Such conjugates offer versatile therapeutic potential by modulating membrane potential, influencing reactive oxygen species (ROS) production, and coupling of molecular modifications (such as ATP metabolism and energy metabolism). Thus, we highlight TPP conjugates as promising mitochondria-targeting agents for use in targeted drug delivery systems.
Collapse
Affiliation(s)
- Sanya Batheja
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer 305817, India
| | - Shruti Gupta
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer 305817, India
| | - Kiran Kumar Tejavath
- Department of Biochemistry, School of Life Sciences, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer 305817, India; Department of Biochemistry, All India Institute of Medical Sciences, BIBINAGAR, Hyderabad Metropolitan Region (HMR), Telangana 508126, India.
| | - Umesh Gupta
- Nanopolymeric Drug Delivery Lab, Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, NH-8, Bandarsindri, Kishangarh, Ajmer 305817, India.
| |
Collapse
|
2
|
Rashid F, Saeed A, Iqbal J. In Vitro Anticancer Effects of Stilbene Derivatives: Mechanistic Studies on HeLa and MCF-7 Cells. Anticancer Agents Med Chem 2021; 21:793-802. [PMID: 32781966 DOI: 10.2174/1871520620666200811123230] [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: 02/13/2020] [Revised: 04/14/2020] [Accepted: 05/10/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND OBJECTIVE The growing prevalence of cancer and the resulting chemoresistance exert a huge burden on healthcare systems and impose a great challenge to public health around the world. In efforts to develop new chemotherapeutic agents for cancer treatment, a class of heterocyclic compounds i.e. triazine-based molecules were investigated as anticancer agents. MATERIALS AND METHODS New triazine hybrids of stilbene were synthesized and evaluated as anticancer agents for cervical (HeLa) and breast (MCF-7) carcinoma cells. The compound (7e), sodium (E)-6,6'-(ethene-1,2- diyl)bis(3-((4-chloro-6-((3-luorophenyl)amino)-1,3,5-triazin-2-yl)amino)benzenesulfonate) was found to be most potent among synthesized derivatives and was explored further for detailed mechanistic studies. RESULTS In a set comprised of twelve derivatives, compound 7e, sodium (E)-6,6'-(ethene-1,2-diyl)bis(3-((4- chloro-6-((3-luorophenyl)amino)-1,3,5-triazin-2-yl)amino)benzenesulfonate) was found most potent inhibitor for HeLa and MCF-7 cells. DISCUSSION The present study has revealed that compound 7e may activate mitochondrial pathway of apoptosis in HeLa and MCF-7 cells which was assessed by DNA binding studies, estimation of the release of Lactate Dehydrogenase (LDH), fluorescence imaging, production of Reactive Oxygen Species (ROS) in cancer cells, analysis of cell cycle by flow cytometry, change in Mitochondrial Membrane Potential (MMP) and activation of caspase-9 and caspase-3. CONCLUSION Compound 7e may serve as a lead in designing new anticancer compounds based on stilbene scaffold.
Collapse
Affiliation(s)
- Faisal Rashid
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| | - Aamer Saeed
- Department of Chemistry, Quaid-i-Azam University, Islamabad, Pakistan
| | - Jamshed Iqbal
- Centre for Advanced Drug Research, COMSATS University Islamabad, Abbottabad Campus, Abbottabad-22060, Pakistan
| |
Collapse
|
3
|
Chen L, Tan Y, Xu H, Wang K, Chen ZH, Zheng N, Li YQ, Lin LR. Enhanced E/ Z-photoisomerization and luminescence of stilbene derivative co-coordinated in di-β-diketonate lanthanide complexes. Dalton Trans 2020; 49:16745-16761. [PMID: 33146650 DOI: 10.1039/d0dt03383a] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
A new tetradentate chelating ligand appending a stilbene derivative, E-N',N'-bis(pyridin-2-ylmethyl)-4-styrylbenzohydrazide (HL) was synthesized, together with two β-diketonates (4,4,4-trifluoro-1-phenylbutane-1,3-dionate, tfd), with or without the trifluoroacetate anion present as a ligand for coordination with lanthanide(iii) ions to form [Ln(tfd)2(HL)(CF3CO2)] (LnC49H36F9N4O7, Ln = La (1), Nd (2), Eu (3), Gd (4)) and [Yb(tfd)2(L)] (YbC47H35F6N4O5 (5), L = deprotonated HL). All five complexes were structurally characterized, and five crystals were obtained and analyzed by single-crystal X-ray diffraction. The quantum yield of trans-to-cis photoisomerization of the stilbene group in gadolinium complex 4 was enhanced about five-fold compared with that of HL itself. Other complexes showed slightly enhanced or depressed photoisomerization. The total luminescence quantum yield/sensitization efficiency of europium complex 3 in the solid state and acetonitrile solution were 22.1%/96.7% and 19.3%/97.9%, respectively. The transfer of ligand energy to the Eu3+ ion was highly efficient. This enhanced photoisomerization and luminescence of the stilbene group within complexes was found to be related to the energy level of lanthanide ions and whether a ligand-to-metal center or ligand-to-ligand charge transfer process was present. The interpretation of experimental results is rationally supported by time-dependent density-functional theory calculations. In complex 4, except for the intramolecular absorption transition of HL ligand itself (IL, πHL-π*HL), the presence of the ligand-to-ligand charge transfer transition from tfd to HL (LLCT, πtfd-π*HL) and the triplet state energy of HL being unable to transfer to the higher 6P7/2 excited energy level of the Gd3+ ion would facilitate HL photoisomerization. For complex 3, this was due to reversed ligand-to-ligand charge transfer transition from HL to tfd (LLCT, πHL-π*tfd) and its energy transfer to the metal center. Although the observed radiative lifetimes of NIR luminescent complexes 2 and 5 were around 10 μs, these systems contained only two diketone ligands, indicating that HL still had a certain promoting effect compared with tris(diketonate) lanthanide complexes. These results offer an important route for the design of new lanthanide-based molecular switching materials.
Collapse
Affiliation(s)
- Lu Chen
- Department of Chemistry, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen, 361005, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Dhiman N, Kaur K, Jaitak V. Tetrazoles as anticancer agents: A review on synthetic strategies, mechanism of action and SAR studies. Bioorg Med Chem 2020; 28:115599. [PMID: 32631569 DOI: 10.1016/j.bmc.2020.115599] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/22/2022]
Abstract
Cancer is a leading cause of death worldwide. Even after the availability of numerous drugs and treatments in the market, scientists and researchers are focusing on new therapies because of their resistance and toxicity issues. The newly synthesized drug candidates are able to demonstrate in vitro activity but are unable to reach clinical trials due to their rapid metabolism and low bioavailability. Therefore there is an imperative requisite to expand novel anticancer negotiators with tremendous activity as well as in vivo efficacy. Tetrazole is a promising pharmacophore which is metabolically more stable and acts as a bioisosteric analogue for many functional groups. Tetrazole fragment is often castoff with other pharmacophores in the expansion of novel anticancer drugs. This is the first systematic review that emphasizes on contemporary strategies used for the inclusion of tetrazole moiety, mechanistic targets along with comprehensive structural activity relationship studies to provide perspective into the rational design of high-efficiency tetrazole-based anticancer drug candidates.
Collapse
Affiliation(s)
- Neha Dhiman
- Department of Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences, Central University of Punjab, Bathinda 151 001, India
| | - Kamalpreet Kaur
- Department of Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences, Central University of Punjab, Bathinda 151 001, India
| | - Vikas Jaitak
- Department of Pharmaceutical Sciences and Natural Products, School of Basic and Applied Sciences, Central University of Punjab, Bathinda 151 001, India.
| |
Collapse
|
5
|
Ahmadi R, Ebrahimzadeh MA. Resveratrol - A comprehensive review of recent advances in anticancer drug design and development. Eur J Med Chem 2020; 200:112356. [PMID: 32485531 DOI: 10.1016/j.ejmech.2020.112356] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 04/12/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Resveratrol is a natural polyphenolic stilbene isolated from various plants, foods and beverages with a broad spectrum of biological and pharmacological properties through modulating diverse targets and signaling pathways. Particularly, it has attracted a great deal of attention as a promising and multitarget anticancer agent due to its potential use in chemoprevention and chemotherapy of various tumors. However, unfavorable pharmacokinetics/pharmacodynamics profile such as poor bioavailability restricted its applications. Therefore, medicinal chemists have synthesized a lot of novel derivatives and analogues of resveratrol using different modification strategies to overcome these limitations and improve anticancer efficacy. Herein, we reviewed the design, synthesis, structure-activity relationship and mechanism of the most potent and privileged resveratrol-based compounds that showed promising anticancer activities in the last five years. We classified these compounds into the ten different categories based on their chemical structure similarities.
Collapse
Affiliation(s)
- Reza Ahmadi
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Department of Medicinal Chemistry, School of Pharmacy and Pharmaceutical Sciences Research Center, Hemoglobinopathy Institute, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
6
|
Bommagani S, Penthala NR, Balasubramaniam M, Kuravi S, Caldas-Lopes E, Guzman ML, Balusu R, Crooks PA. A novel tetrazole analogue of resveratrol is a potent anticancer agent. Bioorg Med Chem Lett 2018; 29:172-178. [PMID: 30528695 DOI: 10.1016/j.bmcl.2018.12.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2018] [Revised: 11/26/2018] [Accepted: 12/04/2018] [Indexed: 12/27/2022]
Abstract
A series of novel tetrazole analogues of resveratrol were synthesized and evaluated for their anti-leukemic activity against an extensive panel of human cancer cell lines and against the MV4-11 AML cell line. These molecules were designed as drug-like derivatives of the resveratrol analogue DMU-212 and its cyano derivatives. Four compounds 8g, 8h, 10a and 10b exhibited LD50 values of 4.60 µM, 0.02 µM, 1.46 µM, and 1.08 µM, respectively, against MV4-11 leukemia cells. The most potent compounds, 8h and 10b, were also found to be active against an extensive panel of human hematological and solid tumor cell lines; compound 8h was the most potent compound with GI50 values <10 nM against more than 90% of the human cancer cell lines in the 60-cell panel. Analogues 8g, 8h, 10a and 10b were also tested for their ability to inhibit the polymerization of tubulin, and compound 8h was found to be the most potent analogue. Molecular modeling studies demonstrated that 8h binds to the colchicine binding site on tubulin. Thus, compound 8h is considered to be a lead druglike molecule from this tetrazole series of compounds.
Collapse
Affiliation(s)
- Shobanbabu Bommagani
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Narsimha Reddy Penthala
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Meenakshisundaram Balasubramaniam
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; Department of Geriatrics, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Sudhakiranmayi Kuravi
- Department of Hematology and Oncology, University of Kansas Medical Center, KS 66160, USA
| | - Eloisi Caldas-Lopes
- Division of Hematology & Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Monica L Guzman
- Division of Hematology & Medical Oncology, Department of Medicine, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ramesh Balusu
- Department of Hematology and Oncology, University of Kansas Medical Center, KS 66160, USA
| | - Peter A Crooks
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|