1
|
Xu H, Li T, Wang Q, Lv Y, Sun C, Yan R, Wu X, Jin Y, Wang Z. Small Molecular Oligopeptides Adorned with Tryptophan Residues as Potent Antitumor Agents: Design, Synthesis, Bioactivity Assay, Computational Prediction, and Experimental Validation. J Chem Inf Model 2025. [PMID: 39817413 DOI: 10.1021/acs.jcim.4c01759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Tryptophan participates in important life activities and is involved in various metabolic processes. The indole and aromatic binuclear ring structure in tryptophan can engage in diverse interactions, including π-π, π-alkyl, hydrogen bonding, cation-π, and CH-π interactions with other side chains and protein targets. These interactions offer extensive opportunities for drug development. In this letter, we have designed and synthesized a series of linear oligopeptides adorned with tryptophan residues and identified their potential targets through artificial intelligence-assisted technology and experimental verification. In vitro bioactivity assays revealed that the oligopeptides containing Gly-Pro-Trp residues exhibited promising antitumor activity by inducing autophagy and apoptosis. The PharmMapper pharmacophore mapping approach, molecular docking, and molecular dynamics simulations together identified poly(ADP-ribose) polymerase 1 (PARP1), an enzyme associated with chromatin regulation, as the potential target for the designed compounds. Experimental biolayer interferometry (BLI) and enzyme-linked immunosorbent assay (ELISA) have verified that the oligopeptides could bind with PARP1 and influence PARP1 expression levels. A quantitative structure-activity relationship has been established between the chemical structures of the prepared compounds and their IC50 values. In summary, the research presents a feasible approach for exploring oligopeptide-based antitumor agents.
Collapse
Affiliation(s)
- Hongyu Xu
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Tong Li
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Qi Wang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Yang Lv
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Changhong Sun
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Rui Yan
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Xiaodan Wu
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Yingxue Jin
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| | - Zhiqiang Wang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry and Chemical Engineering, Harbin Normal University, Harbin 150025, China
| |
Collapse
|
2
|
Panayides JL, Riley DL, Hasenmaile F, van Otterlo WAL. The role of silicon in drug discovery: a review. RSC Med Chem 2024; 15:3286-3344. [PMID: 39430101 PMCID: PMC11484438 DOI: 10.1039/d4md00169a] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/07/2024] [Indexed: 10/22/2024] Open
Abstract
This review aims to highlight the role of silicon in drug discovery. Silicon and carbon are often regarded as being similar with silicon located directly beneath carbon in the same group in the periodic table. That being noted, in many instances a clear dichotomy also exists between silicon and carbon, and these differences often lead to vastly different physiochemical and biological properties. As a result, the utility of silicon in drug discovery has attracted significant attention and has grown rapidly over the past decade. This review showcases some recent advances in synthetic organosilicon chemistry and examples of the ways in which silicon has been employed in the drug-discovery field.
Collapse
Affiliation(s)
- Jenny-Lee Panayides
- Pharmaceutical Technologies, Future Production: Chemicals, Council for Scientific and Industrial Research (CSIR) Meiring Naude Road, Brummeria Pretoria South Africa
| | - Darren Lyall Riley
- Department of Chemistry, Faculty of Natural and Agricultural Sciences, University of Pretoria Lynnwood Road Pretoria South Africa
| | - Felix Hasenmaile
- Department of Chemistry and Polymer Science, Stellenbosch University Matieland Stellenbosch 7600 South Africa
| | - Willem A L van Otterlo
- Department of Chemistry and Polymer Science, Stellenbosch University Matieland Stellenbosch 7600 South Africa
| |
Collapse
|
3
|
Bartoszewska E, Molik K, Woźniak M, Choromańska A. Telomerase Inhibition in the Treatment of Leukemia: A Comprehensive Review. Antioxidants (Basel) 2024; 13:427. [PMID: 38671875 PMCID: PMC11047729 DOI: 10.3390/antiox13040427] [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: 02/28/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Leukemia, characterized by the uncontrolled proliferation and differentiation blockage of myeloid or lymphoid precursor cells, presents significant therapeutic challenges despite current treatment modalities like chemotherapy and stem cell transplantation. Pursuing novel therapeutic strategies that selectively target leukemic cells is critical for improving patient outcomes. Natural products offer a promising avenue for developing effective chemotherapy and preventive measures against leukemia, providing a rich source of biologically active compounds. Telomerase, a key enzyme involved in chromosome stabilization and mainly active in cancer cells, presents an attractive target for intervention. In this review article, we focus on the anti-leukemic potential of natural substances, emphasizing vitamins (such as A, D, and E) and polyphenols (including curcumin and indole-3-carbinol), which, in combination with telomerase inhibition, demonstrate reduced cytotoxicity compared to conventional chemotherapies. We discuss the role of human telomerase reverse transcriptase (hTERT), particularly its mRNA expression, as a potential therapeutic target, highlighting the promise of natural compounds in leukemia treatment and prevention.
Collapse
Affiliation(s)
- Elżbieta Bartoszewska
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland; (E.B.); (K.M.)
| | - Klaudia Molik
- Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 5, 50-345 Wroclaw, Poland; (E.B.); (K.M.)
| | - Marta Woźniak
- Department of Clinical and Experimental Pathology, Division of General and Experimental Pathology, Wroclaw Medical University, Marcinkowskiego 1, 50-368 Wroclaw, Poland;
| | - Anna Choromańska
- Department of Molecular and Cellular Biology, Wroclaw Medical University, Borowska 211A, 50-556 Wroclaw, Poland
| |
Collapse
|
4
|
Chen Z, Liu M, Wang N, Xiao W, Shi J. Unleashing the Potential of Camptothecin: Exploring Innovative Strategies for Structural Modification and Therapeutic Advancements. J Med Chem 2024; 67:3244-3273. [PMID: 38421819 DOI: 10.1021/acs.jmedchem.3c02115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
Camptothecin (CPT) is a potent anti-cancer agent targeting topoisomerase I (TOP1). However, CPT has poor pharmacokinetic properties, causes toxicities, and leads to drug resistance, which limit its clinical use. In this paper, to review the current state of CPT research. We first briefly explain CPT's TOP1 inhibition mechanism and the key hurdles in CPT drug development. Then we examine strategies to overcome CPT's limitations through structural modifications and advanced delivery systems. Though modifications alone seem insufficient to fully enhance CPT's therapeutic potential, structure-activity relationship analysis provides insights to guide optimization of CPT analogs. In comparison, advanced delivery systems integrating controlled release, imaging capabilities, and combination therapies via stimulus-responsive linkers and targeting moieties show great promise for improving CPT's pharmacological profile. Looking forward, multifaceted approaches combining selective CPT derivatives with advanced delivery systems, informed by emerging biological insights, hold promise for fully unleashing CPT's anti-cancer potential.
Collapse
Affiliation(s)
- Zheng Chen
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Maoyu Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
| | - Ningyu Wang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Wenjing Xiao
- Department of Pharmacy, The General Hospital of Western Theater Command of PLA, Chengdu 610083, China
| | - Jianyou Shi
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610072, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
5
|
Chen H, Wang Y, Shao C, Guo K, Liu G, Wang Z, Duan H, Pan M, Ding P, Zhang Y, Han J, Yan X. Molecular subgroup establishment and signature creation of lncRNAs associated with acetylation in lung adenocarcinoma. Aging (Albany NY) 2024; 16:1276-1297. [PMID: 38240708 PMCID: PMC10866443 DOI: 10.18632/aging.205407] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/13/2023] [Indexed: 02/06/2024]
Abstract
BACKGROUND The significance of long non-coding RNAs (lncRNAs) as pivotal mediators of histone acetylation and their influential role in predicting the prognosis of lung adenocarcinoma (LUAD) has been increasingly recognized. However, there remains uncertainty regarding the potential utility of acetylation-related lncRNAs (ARLs) in prognosticating the overall survival (OS) of LUAD specimens. METHODS The RNA-Seq and clinical information were downloaded from The Cancer Genome Atlas (TCGA). Through the differential analysis, weighted correlation network analysis (WGCNA), Pearson correlation test and univariate Cox regression, we found out the prognosis associated ARLs and divided LUAD specimens into two molecular subclasses. The ARLs were employed to construct a unique signature through the implementation of the Least Absolute Shrinkage and Selection Operator (LASSO) algorithm. Subsequently, the predictive performance was evaluated using ROC analysis and Kaplan-Meier survival curve analysis. Finally, ARL expression in LUAD was confirmed by quantitative real-time PCR (qRT-PCR). RESULTS We triumphantly built a ARLs prognostic model with excellent predictive accuracy for LUAD. Univariate and multivariate Cox analysis illustrated that risk model served as an independent predictor for influencing the overall survival OS of LUAD. Furthermore, a nomogram exhibited strong prognostic validity. Additionally, variations were observed among subgroups in the field of immunity, biological functions, drug sensitivity and gene mutations within the field. CONCLUSIONS Nine ARLs were identified as promising indicators of personalized prognosis and drug selection for people suffering with LUAD.
Collapse
Affiliation(s)
- Hao Chen
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Yuanyong Wang
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Changjian Shao
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Kai Guo
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Guanglin Liu
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Zhaoyang Wang
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Hongtao Duan
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Minghong Pan
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Peng Ding
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Yimeng Zhang
- Department of Ophthalmology, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Jing Han
- Department of Ophthalmology, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| | - Xiaolong Yan
- Department of Thoracic Surgery, Tangdu Hospital of Air Force Military Medical University, Xi’an 71003, China
| |
Collapse
|
6
|
Liao YT, Du XY, Wang M, Zheng CX, Li D, Chen CH, Li RT, Shao LD. A silicon-containing aryl/penta-1,4-dien-3-one/amine hybrid exhibits antiproliferative effects on breast cancer cells by targeting the HSP90 C-terminus without inducing heat-shock response. RSC Med Chem 2023; 14:2625-2639. [PMID: 38107168 PMCID: PMC10718586 DOI: 10.1039/d3md00431g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/11/2023] [Indexed: 12/19/2023] Open
Abstract
A pharmacophore-hybridized strategy based on previously reported HSP90 C-terminal inhibitors was utilized to prepare 32 aryl/penta-1,4-dien-3-one/amine hybrids. Among them, a silicon-containing compound 1z exhibited remarkable broad-spectrum antiproliferative effects on various human breast cancer cell lines. Through fluorescence polarization and AlphaScreen-based assays, we demonstrated that 1z specifically inhibited the HSP90 C-terminus without affecting HSP90 N-terminus. Furthermore, 1z effectively inhibited the HSP90 C-terminus without inducing heat-shock response (HSR), leading to the degradation of its client proteins HER2, pAKT, AKT, and CDK4, causing G1 arrest of MCF-7 and SKBr3 cells, and ultimately contributing to apoptosis of these cells through caspase-3, caspase-8, and caspase-9 activation. Additionally, the penta-1,4-dien-3-one linker in the hybrid, a large bulky lipophilic substitution in the aryl fragment at the 3'-site, and the presence of N-methylpiperazine as the amine fragment were identified as crucial factors that significantly contributed to the observed antiproliferative activity through structure-activity relationship (SAR) analysis. Lastly, we found that 1z exhibited superior thermostability compared to vibsanin B derivatives and good in vitro metabolic stability in simulated intestinal fluid, representing one of the few reported silicon-containing HSP90 C-terminal inhibitors.
Collapse
Affiliation(s)
- Yu-Ting Liao
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
| | - Xin-Ye Du
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
- Faculty of Life Science and Technology, Kunming University of Science and Technology Kunming 650500 China
| | - Mei Wang
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
| | - Chun-Xia Zheng
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
| | - Dashan Li
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
| | - Chuan-Huizi Chen
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
| | - Rong-Tao Li
- Faculty of Life Science and Technology, Kunming University of Science and Technology Kunming 650500 China
| | - Li-Dong Shao
- Yunnan Key Laboratory of Southern Medicinal Resources, School of Chinese Materia Medica, Yunnan University of Chinese Medicine Kunming 650500 China
| |
Collapse
|
7
|
Han J, Zhang S, He J, Li T. Piperine: Chemistry and Biology. Toxins (Basel) 2023; 15:696. [PMID: 38133200 PMCID: PMC10747706 DOI: 10.3390/toxins15120696] [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: 09/26/2023] [Revised: 11/17/2023] [Accepted: 11/17/2023] [Indexed: 12/23/2023] Open
Abstract
Piperine is a plant-derived promising piperamide candidate isolated from the black pepper (Piper nigrum L.). In the last few years, this natural botanical product and its derivatives have aroused much attention for their comprehensive biological activities, including not only medical but also agricultural bioactivities. In order to achieve sustainable development and improve survival conditions, looking for environmentally friendly pesticides with low toxicity and residue is an extremely urgent challenge. Fortunately, plant-derived pesticides are rising like a shining star, guiding us in the direction of development in pesticidal research. In the present review, the recent progress in the biological activities, mechanisms of action, and structural modifications of piperine and its derivatives from 2020 to 2023 are summarized. The structure-activity relationships were analyzed in order to pave the way for future development and utilization of piperine and its derivatives as potent drugs and pesticides for improving the local economic development.
Collapse
Affiliation(s)
- Jin Han
- School of Public Administration, Xi’an University of Finance and Economics, Xi’an 710061, China;
| | - Shaoyong Zhang
- Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, College of Life Science, Huzhou University, Huzhou 313000, China;
| | - Jun He
- College of Plant Protection, Northwest A&F University, Xianyang 712100, China;
| | - Tianze Li
- College of Plant Protection, Northwest A&F University, Xianyang 712100, China;
| |
Collapse
|
8
|
Wang X, Zhuang Y, Wang Y, Jiang M, Yao L. The recent developments of camptothecin and its derivatives as potential anti-tumor agents. Eur J Med Chem 2023; 260:115710. [PMID: 37595544 DOI: 10.1016/j.ejmech.2023.115710] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 07/07/2023] [Accepted: 08/03/2023] [Indexed: 08/20/2023]
Abstract
This review article focuses on the research progress made in the structural modifications of camptothecin (CPT), a potent cytotoxic natural alkaloid. CPT possesses a unique 5-fused ring structure and exhibits various beneficial activities such as anti-proliferative, anti-fungal, insecticidal, and anti-SARS-CoV-2 properties. CPT and its analogs, including Topotecan and Irinotecan, have been successfully developed and marketed as topoisomerase I inhibitors. To enhance the therapeutic potential of CPT, researchers have undertaken structural modifications primarily on the A, B, and E rings of the CPT core structure. These modifications aim to improve the efficacy, selectivity, and pharmacokinetic properties of CPT derivatives. The article reviews the advancements in hybridizing CPT with other bioactive compounds, the synthesis of novel CPT analogs, and their associated biological activities. Moreover, the structure-activity relationship (SAR) of these modified CPT derivatives is summarized to gain insights into their structure-function correlations. In addition to discussing the modifications and biological activities of CPT derivatives, the article also touches upon the mechanism of parent drug release. Many CPT derivatives are prodrugs, meaning they require metabolic activation to generate the active form of the drug. It is a resource for researchers interested in developing novel anti-tumor agents based on CPT, addressing the limitations associated with the parent drug, and exploring various aspects of CPT modifications.
Collapse
Affiliation(s)
- Xianzhang Wang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Yumeng Zhuang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Yuankun Wang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Maokai Jiang
- School of Pharmacy, Yantai University, Yantai, 264005, China
| | - Lei Yao
- School of Pharmacy, Yantai University, Yantai, 264005, China.
| |
Collapse
|
9
|
Yakkala PA, Penumallu NR, Shafi S, Kamal A. Prospects of Topoisomerase Inhibitors as Promising Anti-Cancer Agents. Pharmaceuticals (Basel) 2023; 16:1456. [PMID: 37895927 PMCID: PMC10609717 DOI: 10.3390/ph16101456] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/08/2023] [Accepted: 10/11/2023] [Indexed: 10/29/2023] Open
Abstract
Topoisomerases are very important enzymes that regulate DNA topology and are vital for biological actions like DNA replication, transcription, and repair. The emergence and spread of cancer has been intimately associated with topoisomerase dysregulation. Topoisomerase inhibitors have consequently become potential anti-cancer medications because of their ability to obstruct the normal function of these enzymes, which leads to DNA damage and subsequently causes cell death. This review emphasizes the importance of topoisomerase inhibitors as marketed, clinical and preclinical anti-cancer medications. In the present review, various types of topoisomerase inhibitors and their mechanisms of action have been discussed. Topoisomerase I inhibitors, which include irinotecan and topotecan, are agents that interact with the DNA-topoisomerase I complex and avert resealing of the DNA. The accretion of DNA breaks leads to the inhibition of DNA replication and cell death. On the other hand, topoisomerase II inhibitors like etoposide and teniposide, function by cleaving the DNA-topoisomerase II complex thereby effectively impeding the release of double-strand DNA breaks. Moreover, the recent advances in exploring the therapeutic efficacy, toxicity, and MDR (multidrug resistance) issues of new topoisomerase inhibitors have been reviewed in the present review.
Collapse
Affiliation(s)
- Prasanna Anjaneyulu Yakkala
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Naveen Reddy Penumallu
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
| | - Syed Shafi
- Department of Chemistry, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi 110062, India;
| | - Ahmed Kamal
- Department of Pharmaceutical Chemistry, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India;
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Hyderabad Campus, Dist. Medchal, Hyderabad 500078, India
- Telangana State Council of Science & Technology, Environment, Forests, Science & Technology Department, Hyderabad 500004, India
| |
Collapse
|
10
|
Fotie J, Matherne CM, Wroblewski JE. Silicon switch: Carbon-silicon Bioisosteric replacement as a strategy to modulate the selectivity, physicochemical, and drug-like properties in anticancer pharmacophores. Chem Biol Drug Des 2023; 102:235-254. [PMID: 37029092 DOI: 10.1111/cbdd.14239] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/19/2023] [Accepted: 03/21/2023] [Indexed: 04/09/2023]
Abstract
Bioisosterism is one of the leading strategies in medicinal chemistry for the design and modification of drugs, consisting in replacing an atom or a substituent with a different atom or a group with similar chemical properties and an inherent biocompatibility. The objective of such an exercise is to produce a diversity of molecules with similar behavior while enhancing the desire biological and pharmacological properties, without inducing significant changes to the chemical framework. In drug discovery and development, the optimization of the absorption, distribution, metabolism, elimination, and toxicity (ADMETox) profile is of paramount importance. Silicon appears to be the right choice as a carbon isostere because they possess very similar intrinsic properties. However, the replacement of a carbon by a silicon atom in pharmaceuticals has proven to result in improved efficacy and selectivity, while enhancing physicochemical properties and bioavailability. The current review discusses how silicon has been strategically introduced to modulate drug-like properties of anticancer agents, from a molecular design strategy, biological activity, computational modeling, and structure-activity relationships perspectives.
Collapse
Affiliation(s)
- Jean Fotie
- Department of Chemistry and Physics, Southeastern Louisiana University, Hammond, Louisiana, USA
| | - Caitlyn M Matherne
- Department of Chemistry and Physics, Southeastern Louisiana University, Hammond, Louisiana, USA
| | - Jordan E Wroblewski
- Department of Chemistry and Physics, Southeastern Louisiana University, Hammond, Louisiana, USA
| |
Collapse
|
11
|
Belova LO, Golub NA, Pletneva MV, Kirilina NI, Kirilin AD. Reactions of Isonicotinic Acid Hydrazide and Its Trimethylsilyl Derivatives with Isocyanates. RUSS J GEN CHEM+ 2022. [DOI: 10.1134/s1070363222110032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
12
|
Flick AC, Leverett CA, Ding HX, McInturff EL, Fink SJ, Mahapatra S, Carney DW, Lindsey EA, DeForest JC, France SP, Berritt S, Bigi-Botterill SV, Gibson TS, Watson RB, Liu Y, O'Donnell CJ. Synthetic Approaches to the New Drugs Approved During 2020. J Med Chem 2022; 65:9607-9661. [PMID: 35833579 DOI: 10.1021/acs.jmedchem.2c00710] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
New drugs introduced to the market are privileged structures that have affinities for biological targets implicated in human diseases and conditions. These new chemical entities (NCEs), particularly small molecules and antibody-drug conjugates (ADCs), provide insight into molecular recognition and simultaneously function as leads for the design of future medicines. This Review is part of a continuing series presenting the most likely process-scale synthetic approaches to 44 new chemical entities approved for the first time anywhere in the world during 2020.
Collapse
Affiliation(s)
- Andrew C Flick
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Carolyn A Leverett
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Hong X Ding
- Pharmacodia (Beijing) Co. Ltd., Beijing 100085, China
| | - Emma L McInturff
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Sarah J Fink
- Takeda Pharmaceuticals, 125 Binney Street, Cambridge, Massachusetts 02142, United States
| | - Subham Mahapatra
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Daniel W Carney
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Erick A Lindsey
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Jacob C DeForest
- Pfizer Worldwide Research and Development, La Jolla Laboratories, 10777 Science Center Drive, San Diego, California 92121, United States
| | - Scott P France
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Simon Berritt
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | | | - Tony S Gibson
- Takeda Pharmaceuticals, 9625 Towne Centre Drive, San Diego, California 92121, United States
| | - Rebecca B Watson
- Pfizer Worldwide Research and Development, La Jolla Laboratories, 10777 Science Center Drive, San Diego, California 92121, United States
| | - Yiyang Liu
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| | - Christopher J O'Donnell
- Pfizer Worldwide Research and Development, Groton Laboratories, 445 Eastern Point Road, Groton, Connecticut 06340, United States
| |
Collapse
|
13
|
Hao H, Chen S, Wu Z, Su P, Ke C, Feng D. The degradation and environmental risk of camptothecin, a promising marine antifoulant. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 821:153384. [PMID: 35085640 DOI: 10.1016/j.scitotenv.2022.153384] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 12/30/2021] [Accepted: 01/20/2022] [Indexed: 06/14/2023]
Abstract
Given the adverse environmental impacts of the antifoulants currently used in marine antifouling paints, such as copper and booster biocides, it is urgent to identify potential substitutes that are environmentally benign. Here, we examined the degradation of camptothecin (a natural product previously identified as an efficient antifoulant in the laboratory and in the field) under various conditions and evaluated the environmental risks associated with its use as a marine antifoulant. We found that camptothecin was rapidly photolyzed in seawater: the half-life of camptothecin was less than 1 d under a light intensity of 1000-20,000 lx and was approximately 0.17 d under sunlight irradiation. At pH 4 and pH 7, camptothecin had half-lives of 30.13 and 16.90 d, respectively; at 4 °C, 25 °C, and 35 °C, the half-lives of camptothecin were 23.90, 21.66, and 26.65 d, respectively. Camptothecin biodegradation in seawater was negligible. The predicted no-effect concentration (PNEC) of camptothecin was 2.19 × 10-1 μg L-1, while the average predicted environmental concentrations (PECs) in open seas, shipping lanes, commercial harbors, and marinas were 6.14 × 10-7, 9.39 × 10-7, 6.80 × 10-3, and 5.03 × 10-2 μg L-1, respectively. The PEC/PNEC ratio of camptothecin was much lower than 1 (i.e., 2.80 × 10-6, 4.29 × 10-6, 3.11 × 10-2, and 2.30 × 10-1 for open seas, shipping lanes, commercial harbors, and marinas, respectively), indicating that the use of camptothecin as a marine antifoulant posed little environmental risk.
Collapse
Affiliation(s)
- Huanhuan Hao
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Siyu Chen
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China
| | - Zhiwen Wu
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361102, China
| | - Pei Su
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361102, China
| | - Caihuan Ke
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China; State Key Laboratory of Marine Environmental Science, Xiamen University, Xiamen 361102, China
| | - Danqing Feng
- State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen 361102, China.
| |
Collapse
|
14
|
Tong L, Sun W, Wu S, Han Y. Characterization of Caerulomycin A as a dual-targeting anticancer agent. Eur J Pharmacol 2022; 922:174914. [PMID: 35337812 PMCID: PMC9077901 DOI: 10.1016/j.ejphar.2022.174914] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/18/2022] [Accepted: 03/19/2022] [Indexed: 11/19/2022]
Abstract
Caerulomycin A (CaeA), isolated from actinomycetes, has a featured 2,2'-bipyridine core structure. Based on the results of in silico drug-protein docking analysis, CaeA shows potential ligands for interacting with both tubulin and DNA topoisomerase I (Topo-1). The result was confirmed by cell-free tubulin polymerization assay and Topo-1 activity assay. In vitro assays also demonstrated that CaeA increases the polymerization of tubulin and increases cell size. In addition, CaeA inhibits cell viability and growth of various cancer cells, yet exhibits low cytotoxicity. CaeA also affects paclitaxel-resistant cancer cells and synergizes the effect with paclitaxel in reducing cancer cell colony formation rate. In vivo experiments confirm the effect of CaeA on reducing tumor size and weight in nude mouse inoculated with tumor cells with no noticeable side effects. Taken together, our data demonstrate that CaeA is a potential potent agent for cancer treatment through tubulin and Topo-1 dual-targeting with little side effects.
Collapse
Affiliation(s)
- Lingying Tong
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA
| | - Weichao Sun
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA
| | - Shiyong Wu
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA; Department of Chemistry and Biochemistry, Ohio University, Athens, OH, 45701, USA; Molecular and Cellular Biology Program, Ohio University, Athens, OH, 45701, USA
| | - Yong Han
- Edison Biotechnology Institute, Ohio University, Athens, OH, 45701, USA.
| |
Collapse
|
15
|
Lv M, Zhuang X, Zhang Q, Cheng Y, Wu D, Wang X, Qiao T. Acetyl-11-keto-β-boswellic acid enhances the cisplatin sensitivity of non-small cell lung cancer cells through cell cycle arrest, apoptosis induction, and autophagy suppression via p21-dependent signaling pathway. Cell Biol Toxicol 2020; 37:209-228. [PMID: 32562082 PMCID: PMC8012341 DOI: 10.1007/s10565-020-09541-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 06/09/2020] [Indexed: 02/06/2023]
Abstract
Cisplatin-based therapy is a widely used chemotherapeutic regimen for non-small cell lung cancer (NSCLC); however, drug resistance limits its efficacy. Acetyl-11-keto-β-boswellic acid (AKBA), a bioactive compound from frankincense, has been shown to exert anti-cancer effects. The aim of this study is to explore the potential of AKBA in combination with cisplatin as a new regimen for NSCLC. CCK8 assay and clone formation assay were used to determine the effects of AKBA in combination with cisplatin on cell viability of NSCLC cell lines. A three-dimensional spherification assay was used to simulate in vivo tumor formation. Flow cytometry was performed to examine cell cycle distribution and the percentages of apoptotic cells. The associated proteins and mRNA of cell cycle, apoptosis, and autophagy were measured by western blotting and real-time fluorescence quantitative PCR. Immunofluorescence assay was used to test apoptotic nuclei and autolysosome. Small interfering RNA experiments were used to silence the expression of p21. Combination treatment of AKBA and cisplatin inhibited cell viability, clone formation, and three-dimensional spherification, enhanced G0/G1 phase arrest, increased the percentages of apoptotic cells, and decreased the ratio of positive autolysosomes, compared with cisplatin alone. AKBA in combination with cisplatin suppressed the protein expressions of cyclin A2, cyclin E1, p-cdc2, CDK4, Bcl-xl, Atg5, and LC3A/B, and upregulated p27 and p21 mRNA levels in A549 cells. Downregulation of p21 decreased G0/G1 phase arrest and the percentages of apoptotic cells, and promoted autophagy in NSCLC A549 cells. Our study demonstrates that AKBA enhances the cisplatin sensitivity of NSCLC cells and that the mechanisms involve G0/G1 phase arrest, apoptosis induction, and autophagy suppression via targeting p21-dependent signaling pathway.
Collapse
Affiliation(s)
- Minghe Lv
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China
| | - Xibing Zhuang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China
| | - Qi Zhang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China
| | - Yunfeng Cheng
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China
| | - Duojiao Wu
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China
| | - Xiangdong Wang
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China
| | - Tiankui Qiao
- Center for Tumor Diagnosis and Therapy, Jinshan Hospital, Fudan University, Jinshan District, Shanghai, 201508, China.
| |
Collapse
|
16
|
Hevener K, Verstak TA, Lutat KE, Riggsbee DL, Mooney JW. Recent developments in topoisomerase-targeted cancer chemotherapy. Acta Pharm Sin B 2018; 8:844-861. [PMID: 30505655 PMCID: PMC6251812 DOI: 10.1016/j.apsb.2018.07.008] [Citation(s) in RCA: 144] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2018] [Revised: 07/18/2018] [Accepted: 07/18/2018] [Indexed: 12/17/2022] Open
Abstract
The DNA topoisomerase enzymes are essential to cell function and are found ubiquitously in all domains of life. The various topoisomerase enzymes perform a wide range of functions related to the maintenance of DNA topology during DNA replication, and transcription are the targets of a wide range of antimicrobial and cancer chemotherapeutic agents. Natural product-derived agents, such as the camptothecin, anthracycline, and podophyllotoxin drugs, have seen broad use in the treatment of many types of cancer. Selective targeting of the topoisomerase enzymes for cancer treatment continues to be a highly active area of basic and clinical research. The focus of this review will be to summarize the current state of the art with respect to clinically used topoisomerase inhibitors for targeted cancer treatment and to discuss the pharmacology and chemistry of promising new topoisomerase inhibitors in clinical and pre-clinical development.
Collapse
Affiliation(s)
- KirkE. Hevener
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | | | | | | | | |
Collapse
|