1
|
Ibrahim H, Bala MD, Ntola P. The impact of wingtip N-substitution on the bioactivity of azolium salts. Eur J Med Chem 2025; 295:117797. [PMID: 40449118 DOI: 10.1016/j.ejmech.2025.117797] [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: 03/25/2025] [Revised: 05/19/2025] [Accepted: 05/21/2025] [Indexed: 06/02/2025]
Abstract
Studies have shown that the physicochemical properties of five-membered heterocyclic azolium compounds directly affect their biological activity as therapeutic drugs (spectrum of activity and potency) and the associated pharmacokinetic, pharmacologic, and toxicological profiles of the compounds. Hence, this review focused on the influence of N-functionalisation at the wingtip of such compounds, mainly the diazolium and the triazolium-based salts. The contribution of the N-donor groups to the overall biological efficacy of the azolium compounds and the ensuing structure-activity mechanisms in their pharmacological applications are comprehensively discussed.
Collapse
Affiliation(s)
- Halliru Ibrahim
- Department of Chemistry, Durban University of Technology, P.O Box 1334, Durban, 4000, South Africa
| | - Muhammad D Bala
- School of Chemistry and Physics, University of KwaZulu Natal, Private Bag X54001, Durban, 4000, South Africa.
| | - Pinkie Ntola
- Department of Chemistry, Durban University of Technology, P.O Box 1334, Durban, 4000, South Africa
| |
Collapse
|
2
|
Liu WB, Yang WG, Wu J, Chen BB, Du YF, Niu JB, Song J, Zhang SY. Discovery of novel thienopyridine indole derivatives as inhibitors of tubulin polymerization targeting the colchicine-binding site with potent anticancer activities. Eur J Med Chem 2025; 286:117314. [PMID: 39874633 DOI: 10.1016/j.ejmech.2025.117314] [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: 11/29/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Based on the molecular hybridization strategy, novel thienopyridine indole derivatives were designed and synthesized as tubulin polymerization inhibitors, and the in vitro antiproliferative potency on MGC-803, KYSE450 and HCT-116 cells was evaluated. Among them, compound 20b showed a broad-spectrum antiproliferative activity against 11 cancer cell lines, with IC50 values below 4 nmol/L. Notably, it demonstrated exceptional efficacy against MGC-803 (IC50 = 1.61 nmol/L) and HGC-27 (IC50 = 1.82 nmol/L) cells. Further mechanism explorations suggested that compound 20b could inhibit tubulin polymerization (IC50 = 2.505 μmol/L) by acting on the colchicine binding site, thereby disrupting intracellular microtubule networks and interfering with cell mitosis. In addition, compound 20b effectively inhibited the colony formation and cell migration activities, and induced G2/M phase cycle arrest and apoptosis in MGC-803 and HGC-27 cells. Besides, compound 20b also displayed potent anti-angiogenesis effects on HUVECs. Importantly, compound 20b demonstrated oral efficacy in inhibiting tumor growth with a TGI of 45.8 % (5 mg/kg/qod) in the mouse xenograft model bearing MGC-803 cells, surpassing that of CA-4 (TGI of 27.1 % at 20 mg/kg/qod), as well as also exhibited a good safety profile. Therefore, these results suggested that the thienopyridine indole derivative 20b represents a novel tubulin inhibitor with potent anticancer efficacy that inhibits gastric cancers.
Collapse
Affiliation(s)
- Wen-Bo Liu
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Wei-Guang Yang
- Children's Hospital Affiliated of Zhengzhou University, Henan Children's Hospital, Zhengzhou Children's Hospital, Zhengzhou, 450000, China
| | - Ji Wu
- School of Pharmaceutical Sciences, Institute of Drug Discovery & Development, Key Laboratory of Advanced Drug Preparation Technologies (Ministry of Education), Zhengzhou University, Zhengzhou, 450001, China
| | - Bing-Bing Chen
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yi-Fei Du
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Jin-Bo Niu
- The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jian Song
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| | - Sai-Yang Zhang
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
3
|
Manna T, Maji S, Maity M, Debnath B, Panda S, Khan SA, Nath R, Akhtar MJ. Anticancer potential and structure activity studies of purine and pyrimidine derivatives: an updated review. Mol Divers 2025; 29:817-848. [PMID: 38856835 DOI: 10.1007/s11030-024-10870-4] [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: 02/19/2024] [Accepted: 04/02/2024] [Indexed: 06/11/2024]
Abstract
Cancer is the world's leading cause of death impacting millions of lives globally. The increasing research over the past several decades has focused on the development of new anticancer drugs, but still cancer continues to be a global health challenge. Thus, several new alternative therapeutic strategies have been tried for the drug design and discovery. Purine and pyrimidine heterocyclic compounds have received attention recently due to their potential in targeting various cancers. It is evident from the recently published data over the last decade that incorporation of the purine and pyrimidine rings in the synthesized derivatives resulted in the development of potent anticancer molecules. This review presents synthetic strategies encompassing several examples of recently developed purine and pyrimidine-containing compounds as anticancer agents. In addition, their structure-activity relationships are represented in the schemes indicating the fragment or groups that are essential for the enhanced anticancer activities. Purine and pyrimidines combined with other heterocyclic compounds have resulted in many novel anticancer molecules that address the challenges of drug resistance. The purine and pyrimidine derivatives showed significantly enhanced anticancer activities against targeted receptor proteins with numerous compounds with an IC50 value in the nanomolar range. The review will support medicinal chemists and contribute in progression and development of synthesis of more potent chemotherapeutic drug candidates to mitigate the burden of this dreadful disease.
Collapse
Affiliation(s)
- Tanushree Manna
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Sumit Maji
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Mousumi Maity
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Biplab Debnath
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Shambo Panda
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India
| | - Shah Alam Khan
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PC 130, Azaiba, Bousher, PO 620, Muscat, Sultanate of Oman
| | - Rajarshi Nath
- Department of Pharmacy, Bharat Technology, Uluberia, 711316, Howrah, West Bengal, India.
- JIS University, Agarpara Campus, Kolkata-81, Nilgunj Road, Agarpara, Kolkata, 700109, India.
| | - Md Jawaid Akhtar
- Department of Pharmaceutical Chemistry, National University of Science and Technology, PC 130, Azaiba, Bousher, PO 620, Muscat, Sultanate of Oman.
| |
Collapse
|
4
|
Xu Q, Tu Y, Zhang Y, Xiu Y, Yu Z, Jiang H, Wang C. Discovery and biological evaluation of 6-aryl-4-(3,4,5-trimethoxyphenyl)quinoline derivatives with promising antitumor activities as novel colchicine-binding site inhibitors. Eur J Med Chem 2024; 279:116869. [PMID: 39316845 DOI: 10.1016/j.ejmech.2024.116869] [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: 08/05/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/26/2024]
Abstract
Tubulin, as the fundamental unit of microtubules, is a crucial target in the investigation of anticarcinogens. The synthesis and assessment of small-molecule tubulin polymerization inhibitors remains a promising avenue for the development of novel cancer therapeutics. Through an analysis of reported colchicine-binding site inhibitors (CBSIs) and tubulin binding models, a set of 6-aryl-4-(3,4,5-trimethoxyphenyl)quinoline derivatives were meticulously crafted as potential CBSIs. Notably, compound 14u exhibited potent anti-proliferative efficacy, displaying IC50 values ranging from 0.03 to 0.18 μM against three human cancer cell lines (Huh7, MCF-7, and SGC-7901). Mechanistic investigations revealed that compound 14u could disrupt tubulin polymerization, dismantle the microtubule architecture, arrest the cell cycle at G2/M phase, and induce apoptosis in cancer cells. Furthermore, compound 14u demonstrated significant inhibition of tumor proliferation in vivo with no discernible toxicity in the Huh7 orthotopic tumor model mice. Additionally, physicochemical property predictions indicated that compound 14u adhered well to Lipinski's rule of five. These findings collectively suggest that compound 14u holds promise as an antitumor agent targeting the colchicine-binding site on tubulin and warrants further investigation.
Collapse
Affiliation(s)
- Qianqian Xu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yuxuan Tu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yujing Zhang
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China
| | - Yutao Xiu
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, Shandong, China
| | - Zongjiang Yu
- CAS Key Laboratory of Biobased Materials, Qingdao Institute of Bioenergy and Bioprocess Technology, Chinese Academy of Sciences, Qingdao 26610, Shandong, China.
| | - Hongfei Jiang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, Shandong, China.
| | - Chao Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266071, Shandong, China; Qingdao Cancer Institute, Qingdao University, Qingdao, 266071, Shandong, China.
| |
Collapse
|
5
|
Fuentes-Martín R, Ayuda-Durán P, Hanes R, Gallego-Yerga L, Wolterinck L, Enserink JM, Álvarez R, Peláez R. Promising anti-proliferative indolic benzenesulfonamides alter mechanisms with sulfonamide nitrogen substituents. Eur J Med Chem 2024; 275:116617. [PMID: 38959729 DOI: 10.1016/j.ejmech.2024.116617] [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: 05/14/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Agents that cause apoptotic cell death by interfering with tubulin dynamics, such as vinblastine and paclitaxel, are an important class of chemotherapeutics. Unfortunately, these compounds are substrates for multidrug resistance (MDR) pumps, allowing cancer cells to gain resistance to these chemotherapeutics. The indolesulfonamide family of tubulin inhibitors are not excluded by MDR pumps and have a promising activity profile, although their high lipophilicity is a pharmacokinetic limitation for their clinical use. Here we present a new family of N-indolyl-3,4,5-trimethoxybenzenesulfonamide derivatives with modifications on the indole system at positions 1 and 3 and on the sulfonamide nitrogen. We synthesized and screened against HeLa cells 34 novel indolic benzenesulfonamides. The most potent derivatives (1.7-109 nM) were tested against a broad panel of cancer cell lines, which revealed that substituted benzenesulfonamides analogs had highest potency. Importantly, these compounds were only moderately toxic to non-tumorigenic cells, suggesting the presence of a therapeutic index. Consistent with known clinical anti-tubulin agents, these compounds arrested the cell cycle at G2/M phase. Mechanistically, they induced apoptosis via caspase 3/7 activation, which occurred during M arrest. The substituents on the sulfonamide nitrogen appeared to determine different mechanistic results and cell fates. These results suggest that the compounds act differently depending on the bridge substituents, thus making them very interesting as mechanistic probes as well as potential drugs for further development.
Collapse
Affiliation(s)
- Raúl Fuentes-Martín
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain
| | - Pilar Ayuda-Durán
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Robert Hanes
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Laura Gallego-Yerga
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain
| | - Lisanne Wolterinck
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; HAN University of Applied Sciences, Nijmegen, the Netherlands
| | - Jorrit M Enserink
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway; Section for Biochemistry and Molecular Biology, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Raquel Álvarez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain
| | - Rafael Peláez
- Laboratorio de Química Orgánica y Farmacéutica, Departamento de Ciencias Farmacéuticas, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca (IBSAL), Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain; Centro de Investigación de Enfermedades Tropicales de la Universidad de Salamanca (CIETUS). Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno, E-37007, Salamanca, Spain.
| |
Collapse
|
6
|
Majirská M, Pilátová MB, Kudličková Z, Vojtek M, Diniz C. Targeting hematological malignancies with isoxazole derivatives. Drug Discov Today 2024; 29:104059. [PMID: 38871112 DOI: 10.1016/j.drudis.2024.104059] [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: 02/20/2024] [Revised: 05/18/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Compounds with a heterocyclic isoxazole ring are well known for their diverse biologic activities encompassing antimicrobial, antipsychotic, immunosuppressive, antidiabetic and anticancer effects. Recent studies on hematological malignancies have also shown that some of the isoxazole-derived compounds feature encouraging cancer selectivity, low toxicity to normal cells and ability to overcome cancer drug resistance of conventional treatments. These characteristics are particularly promising because patients with hematological malignancies face poor clinical outcomes caused by cancer drug resistance or relapse of the disease. This review summarizes the knowledge on isoxazole-derived compounds toward hematological malignancies and provides clues on their mechanism(s) of action (apoptosis, cell cycle arrest, ROS production) and putative pharmacological targets (c-Myc, BET, ATR, FLT3, HSP90, CARM1, tubulin, PD-1/PD-L1, HDACs) wherever known.
Collapse
Affiliation(s)
- Monika Majirská
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovakia
| | - Martina Bago Pilátová
- Department of Pharmacology, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Slovakia.
| | - Zuzana Kudličková
- NMR Laboratory, Institute of Chemistry, Faculty of Science, Pavol Jozef Šafárik University in Košice, Slovakia
| | - Martin Vojtek
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal.
| | - Carmen Diniz
- LAQV/REQUIMTE, Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, Porto, Portugal
| |
Collapse
|
7
|
Said AM, Mansour YE, Soliman RR, Islam R, Fatahala SS. Design, synthesis, molecular modeling, in vitro and in vivo biological evaluation of potent anthranilamide derivatives as dual P-glycoprotein and CYP3A4 inhibitors. Eur J Med Chem 2024; 273:116492. [PMID: 38762918 DOI: 10.1016/j.ejmech.2024.116492] [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: 03/09/2024] [Revised: 05/06/2024] [Accepted: 05/08/2024] [Indexed: 05/21/2024]
Abstract
Paclitaxel (PTX) is considered the blockbuster chemotherapy treatment for cancer. Paclitaxel's (PTX) oral administration has proven to be extremely difficult, mostly because of its susceptibility to intestinal P-glycoprotein (P-gp) and cytochrome P450 (CYP3A4). The concurrent local inhibition of intestinal P-gp and CYP3A4 is a promising approach to improve the oral bioavailability of paclitaxel while avoiding potential unfavorable side effects of their systemic inhibition. Herein, we report the rational design and evaluation of novel dual potent inhibitors of P-gp and CYP3A4 using an anthranilamide derivative tariquidar as a starting point for their structural optimizations. Compound 14f, bearing N-imidazolylbenzyl side chain, was found to have potent and selective P-gp (EC50 = 28 nM) and CYP3A4 (IC50 = 223 nM) inhibitory activities with low absorption potential (Papp (A-to-B) <0.06). In vivo, inhibitor 14f improved the oral absorption of paclitaxel by 6 times in mice and by 30 times in rats as compared to vehicle, while 14f itself remained poorly absorbed. Compound 14f, possessing dual P-gp and CYP3A4 inhibitory activities, offered additional enhancement in paclitaxel oral absorption compared to tariquidar in mice. Evaluating the CYP effect of 14f on oral absorption of paclitaxel requires considering the variations in CYP expression between animal species. This study provides further medicinal chemistry advice on strategies for resolving concerns with the oral administration of chemotherapeutic agents.
Collapse
Affiliation(s)
- Ahmed M Said
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE, 68198, USA; Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo, 11795, Egypt; Department of Chemistry, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA.
| | - Yara E Mansour
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo, 11795, Egypt
| | - Radwa R Soliman
- Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, NY, 14260, USA
| | - Ridwan Islam
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, BCC, Omaha, NE, 68198, USA
| | - Samar S Fatahala
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Helwan University, Ein-Helwan, Helwan, Cairo, 11795, Egypt.
| |
Collapse
|
8
|
Wang S, Wang J, Lu X, Liu M, Liu Y, Li M, Kong X, Wu L, Guan Q, Zhang W. Design, synthesis and biological evaluation of novel tubulin-targeting agents with a dual-mechanism for polymerization inhibition and protein degradation. Eur J Med Chem 2024; 272:116458. [PMID: 38703557 DOI: 10.1016/j.ejmech.2024.116458] [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: 03/03/2024] [Revised: 04/12/2024] [Accepted: 04/25/2024] [Indexed: 05/06/2024]
Abstract
Microtubules are recognized as one of the most vital and attractive targets in anticancer therapy. The development of novel tubulin-targeting agents with a new action mechanism is imperative. Based on the hydrophobic tagging strategy, the molecular scaffold of tirbanibulin was selected as tubulin target-binding moiety, subsequent to which a series of target compounds were rationally designed by selecting various combinations of linkers and hydrophobic tags. A set of novel molecules were synthesized and most of them exhibited potent antiproliferative activity against tumor cells in vitro. The most active compound 14b inhibited polymerization of purified recombinant tubulin and induced degradation of α- and β-tubulin in MCF-7 cells. Notably, following treatment with compound 14b, an unexpected phenomenon of "microtubules fragmentation" was observed via immunofluorescence staining. Furthermore, compound 14b possessed antitumor activity in the 4T1 allograft models with TGI of 74.27 % without significant toxicity. In this work, we report the discovery of novel dual-mechanism tubulin-targeting agents.
Collapse
Affiliation(s)
- Sibo Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Jiahao Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xiankun Lu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Meitong Liu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Yue Liu
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mi Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Xuejie Kong
- Department of Geratology, The First Affiliated Hospital, Chinese Medical University, Shenyang, 110001, China
| | - Lan Wu
- Department of Geratology, The First Affiliated Hospital, Chinese Medical University, Shenyang, 110001, China.
| | - Qi Guan
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
9
|
Zoroddu S, Sanna L, Bordoni V, Weidong L, Gadau SD, Carta A, Kelvin DJ, Bagella L. Identification of 3-Aryl-1-benzotriazole-1-yl-acrylonitrile as a Microtubule-Targeting Agent (MTA) in Solid Tumors. Int J Mol Sci 2024; 25:5704. [PMID: 38891892 PMCID: PMC11172098 DOI: 10.3390/ijms25115704] [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: 03/03/2024] [Revised: 05/06/2024] [Accepted: 05/21/2024] [Indexed: 06/21/2024] Open
Abstract
Recently, a compound derived from recent scientific advances named 34 has emerged as the focus of this research, the aim of which is to explore its potential impact on solid tumor cell lines. Using a combination of bioinformatics and biological assays, this study conducted an in-depth investigation of the effects of 34. The results of this study have substantial implications for cancer research and treatment. 34 has shown remarkable efficacy in inhibiting the growth of several cancer cell lines, including those representing prostate carcinoma (PC3) and cervical carcinoma (HeLa). The high sensitivity of these cells, indicated by low IC50 values, underscores its potential as a promising chemotherapeutic agent. In addition, 34 has revealed the ability to induce cell cycle arrest, particularly in the G2/M phase, a phenomenon with critical implications for tumor initiation and growth. By interfering with DNA replication in cancer cells, 34 has shown the capacity to trigger cell death, offering a new avenue for cancer treatment. In addition, computational analyses have identified key genes affected by 34 treatment, suggesting potential therapeutic targets. These genes are involved in critical biological processes, including cell cycle regulation, DNA replication and microtubule dynamics, all of which are central to cancer development and progression. In conclusion, this study highlights the different mechanisms of 34 that inhibit cancer cell growth and alter the cell cycle. These promising results suggest the potential for more effective and less toxic anticancer therapies. Further in vivo validation and exploration of combination therapies are critical to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Stefano Zoroddu
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
| | - Luca Sanna
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
| | - Valentina Bordoni
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
| | - Lyu Weidong
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou 515031, China; (L.W.); (D.J.K.)
| | | | - Antonio Carta
- Department of Medicine, Surgery and Pharmacy, University of Sassari, 07100 Sassari, Italy;
| | - David J. Kelvin
- Division of Immunology, International Institute of Infection and Immunity, Shantou University Medical College, Shantou 515031, China; (L.W.); (D.J.K.)
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS B3H 4R2, Canada
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, 07100 Sassari, Italy; (S.Z.); (L.S.); (V.B.)
- Sbarro Institute for Cancer Research and Molecular Medicine, Centre for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
10
|
Zhuang S, Liu Z, Wu J, Yao Y, Li Z, Shen Y, Yu B, Wu D. Can O-GIcNAc Transferase (OGT) Complex Be Used as a Target for the Treatment of Hematological Malignancies? Pharmaceuticals (Basel) 2024; 17:664. [PMID: 38931332 PMCID: PMC11206344 DOI: 10.3390/ph17060664] [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: 01/27/2024] [Revised: 03/03/2024] [Accepted: 03/14/2024] [Indexed: 06/28/2024] Open
Abstract
The circulatory system is a closed conduit system throughout the body and consists of two parts as follows: the cardiovascular system and the lymphatic system. Hematological malignancies usually grow and multiply in the circulatory system, directly or indirectly affecting its function. These malignancies include multiple myeloma, leukemia, and lymphoma. O-linked β-N-acetylglucosamine (O-GlcNAc) transferase (OGT) regulates the function and stability of substrate proteins through O-GlcNAc modification. Abnormally expressed OGT is strongly associated with tumorigenesis, including hematological malignancies, colorectal cancer, liver cancer, breast cancer, and prostate cancer. In cells, OGT can assemble with a variety of proteins to form complexes to exercise related biological functions, such as OGT/HCF-1, OGT/TET, NSL, and then regulate glucose metabolism, gene transcription, cell proliferation, and other biological processes, thus affecting the development of hematological malignancies. This review summarizes the complexes involved in the assembly of OGT in cells and the role of related OGT complexes in hematological malignancies. Unraveling the complex network regulated by the OGT complex will facilitate a better understanding of hematologic malignancy development and progression.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Donglu Wu
- College of Traditional Chinese Medicine, Changchun University of Traditional Chinese Medicine, Changchun 130117, China; (S.Z.); (Z.L.); (J.W.); (Y.Y.); (Z.L.); (Y.S.); (B.Y.)
| |
Collapse
|
11
|
Barreca M, Bertoni F, Barraja P. New strategies to hit hematological cancers. Eur J Med Chem 2024; 270:116350. [PMID: 38582688 DOI: 10.1016/j.ejmech.2024.116350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2024]
Affiliation(s)
- Marilia Barreca
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy.
| | - Francesco Bertoni
- Institute of Oncology Research, Faculty of Biomedical Sciences, USI, Bellinzona, Switzerland; Oncology Institute of Southern Switzerland, EOC, Bellinzona, Switzerland
| | - Paola Barraja
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Palermo, Italy
| |
Collapse
|
12
|
Huang M, Han H, Liu H, Liu R, Li J, Li M, Guan Q, Zhang W, Wang D. Structure-based approaches for the design of 6-aryl-1-(3,4,5-trimethoxyphenyl)-1H-benzo[d][1,2,3]triazoles as tubulin polymerization inhibitors. Eur J Med Chem 2024; 269:116309. [PMID: 38471357 DOI: 10.1016/j.ejmech.2024.116309] [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: 01/07/2024] [Revised: 02/23/2024] [Accepted: 03/04/2024] [Indexed: 03/14/2024]
Abstract
The colchicine binding site on tubulin has been widely acknowledged as an attractive target for anticancer drug exploitation. Here, we reported the structural optimization of the lead compound 4, which was proved in our previous work as a colchicine binding site inhibitor (CBSI). Based on docking researches for the active binding conformation of compound 4, a series of novel 6-aryl-1-(3,4,5-trimethoxyphenyl)-1H-benzo[d][1,2,3]triazole derivatives (9a-9x) were developed by replacing a CH group in the 1H-benzo[d]imidazole skeleton of compound 4 with a nitrogen atom as a hydrogen bond acceptor. Among them, compound 9a showed the strongest antiproliferative activity with IC50 values ranging from 14 to 45 nM against three human cancer cell lines (MCF-7, SGC-7901 and A549), lower than that of compound 4. Mechanistic studies indicated that compound 9a could inhibit tubulin polymerization, destroy the microtubule skeleton, block the cell cycle in G2/M phase, induce cancer cell apoptosis, prevent cancer cell migration and colony formation. Moreover, compound 9a significantly inhibited tumor growth in vivo without observable toxicity in the mice 4T1 xenograft tumor model. In conclusion, this report shows a successful case of the structure-based design approach of a potent tubulin polymerization inhibitor for cancer treatment.
Collapse
Affiliation(s)
- Mingxin Huang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Hongyao Han
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Haoyuan Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Runlai Liu
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Jiwei Li
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Mi Li
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China
| | - Qi Guan
- School of Pharmaceutical Engineering, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Weige Zhang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| | - Dun Wang
- Key Laboratory of Structure-Based Drug Design and Discovery, Ministry of Education, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenhe District, Shenyang, 110016, China.
| |
Collapse
|
13
|
Zhang C, Yan W, Liu Y, Tang M, Teng Y, Wang F, Hu X, Zhao M, Yang J, Li Y. Structure-based design and synthesis of BML284 derivatives: A novel class of colchicine-site noncovalent tubulin degradation agents. Eur J Med Chem 2024; 268:116265. [PMID: 38430854 DOI: 10.1016/j.ejmech.2024.116265] [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: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/18/2024] [Indexed: 03/05/2024]
Abstract
Our previous studies have demonstrated that BML284 is a colchicine-site tubulin degradation agent. To improve its antiproliferative properties, 45 derivatives or analogs of BML284 were designed and synthesized based on the cocrystal structure of BML284 and tubulin. Among them, 5i was the most potent derivative, with IC50 values ranging from 0.02 to 0.05 μM against the five tested tumor cell lines. Structure-activity relationship studies verified that the N1 atom of the pyrimidine ring was the key functional group for its tubulin degradation ability. The 5i-tubulin cocrystal complex revealed that the binding pattern of 5i to tubulin is similar to that of BML284. However, replacing the benzodioxole ring with an indole ring strengthened the hydrogen bond formed by the 2-amino group with E198, which improved the antiproliferative activity of 5i. Compound 5i effectively suppressed tumor growth at an intravenous dose of 40 mg/kg (every 2 days) in paclitaxel sensitive A2780S and paclitaxel resistant A2780T ovarian xenograft models, with tumor growth inhibition values of 79.4% and 82.0%, respectively, without apparent side effects, showing its potential to overcome multidrug resistance. This study provided a successful example of crystal structure-guided discovery of 5i as a colchicine-targeted tubulin degradation agent, expanding the scope of targeted protein degradation.
Collapse
Affiliation(s)
- Chufeng Zhang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Wei Yan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yan Liu
- National Facility for Translational Medicine (Sichuan), West China Hospital, Sichuan University, 610041, Sichuan, China
| | - Minghai Tang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Yaxin Teng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Fang Wang
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Xiuying Hu
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China
| | - Min Zhao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu, 610041, China.
| | - Jianhong Yang
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China.
| | - Yong Li
- Innovation Center of Nursing Research, Nursing Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and Collaborative Innovation Center of Biotherapy, Sichuan University, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
14
|
Yang H, Zhang D, Yuan Z, Qiao H, Xia Z, Cao F, Lu Y, Jiang F. Novel 4-Aryl-4H-chromene derivative displayed excellent in vivo anti-glioblastoma efficacy as the microtubule-targeting agent. Eur J Med Chem 2024; 267:116205. [PMID: 38350361 DOI: 10.1016/j.ejmech.2024.116205] [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: 12/14/2023] [Revised: 01/23/2024] [Accepted: 01/31/2024] [Indexed: 02/15/2024]
Abstract
In this study, a series of novel 4-Aryl-4H-chromene derivatives (D1-D31) were designed and synthesized by integrating quinoline heterocycle to crolibulin template molecule based on the strategy of molecular hybridization. One of these compounds D19 displayed positive antiproliferative activity against U87 cancer cell line (IC50 = 0.90 ± 0.03 μM). Compound D19 was verified as the microtubule-targeting agent through downregulating tubulin related genes of U87 cells, destroying the cytoskeleton of tubulins and interacting with the colchicine-binding site to inhibit the polymerization of tubulins by transcriptome analysis, immune-fluorescence staining, microtubule dynamics and EBI competition assays as well as molecular docking simulations. Moreover, compound D19 induced G2/M phase arrest, resulted in cell apoptosis and inhibited the migration of U87 cells by flow cytometry analysis and wound healing assays. Significantly, compound D19 dose-dependently inhibited the tumor growth of orthotopic glioma xenografts model (GL261-Luc) and effectively prolonged the survival time of mice, which were extremely better than those of positive drug temozolomide (TMZ). Compound D19 exhibited potent in vivo antivascular activity as well as no observable toxicity. Furthermore, the results of in silico simulation studies and P-gp transwell assays verified the positive correlation between compound D19's Blood-Brain Barrier (BBB) permeability and its in vivo anti-GBM activity. Overall, compound D19 can be used as a promising anti-GBM lead compound for the treatment of glioblastoma.
Collapse
Affiliation(s)
- Haoyi Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China
| | - Dongyu Zhang
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Ziyang Yuan
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Haishi Qiao
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhuolu Xia
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China
| | - Feng Cao
- School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, China.
| | - Feng Jiang
- School of Engineering, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
15
|
Barbosa F, Araújo J, Gonçalves VMF, Palmeira A, Cunha A, Silva PMA, Fernandes C, Pinto M, Bousbaa H, Queirós O, Tiritan ME. Evaluation of Antitumor Activity of Xanthones Conjugated with Amino Acids. Int J Mol Sci 2024; 25:2121. [PMID: 38396802 PMCID: PMC10889492 DOI: 10.3390/ijms25042121] [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: 12/24/2023] [Revised: 02/04/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Cancer is a complex disease characterized by several alterations, which confer, to the cells, the capacity to proliferate uncontrollably and to resist cellular death. Multiresistance to conventional chemotherapy drugs is often the cause of treatment failure; thus, the search for natural products or their derivatives with therapeutic action is essential. Chiral derivatives of xanthones (CDXs) have shown potential inhibitory activity against the growth of some human tumor cell lines. This work reports the screening of a library of CDXs, through viability assays, in different cancer cell lines: A375-C5, MCF-7, NCI-H460, and HCT-15. CDXs' effect was analyzed based on several parameters of cancer cells, and it was also verified if these compounds were substrates of glycoprotein-P (Pgp), one of the main mechanisms of resistance in cancer therapy. Pgp expression was evaluated in all cell lines, but no expression was observed, except for HCT-15. Also, when a humanized yeast expressing the human gene MDR1 was used, no conclusions could be drawn about CDXs as Pgp substrates. The selected CDXs did not induce significant differences in the metabolic parameters analyzed. These results show that some CDXs present promising antitumor activity, but other mechanisms should be triggered by these compounds.
Collapse
Affiliation(s)
- Flávia Barbosa
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
| | - Joana Araújo
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (J.A.); (A.P.); (C.F.); (M.P.)
| | - Virgínia M. F. Gonçalves
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), University Institute of Health Sciences-CESPU (IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Andreia Palmeira
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (J.A.); (A.P.); (C.F.); (M.P.)
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Andrea Cunha
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
| | - Patrícia M. A. Silva
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), University Institute of Health Sciences-CESPU (IUCS-CESPU), 4585-116 Gandra, Portugal
| | - Carla Fernandes
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (J.A.); (A.P.); (C.F.); (M.P.)
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Madalena Pinto
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (J.A.); (A.P.); (C.F.); (M.P.)
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| | - Hassan Bousbaa
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
| | - Odília Queirós
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
| | - Maria Elizabeth Tiritan
- UNIPRO—Oral Pathology and Rehabilitation Research Unit, University Institute of Health Sciences (IUCS-CESPU), 4585-116 Gandra, Portugal; (F.B.); (V.M.F.G.); (A.C.); (P.M.A.S.); (H.B.); (O.Q.)
- Laboratory of Organic and Pharmaceutical Chemistry, Department of Chemical Sciences, Faculty of Pharmacy, University of Porto, Rua Jorge Viterbo Ferreira, 228, 4050-313 Porto, Portugal; (J.A.); (A.P.); (C.F.); (M.P.)
- 1H-TOXRUN—One Health Toxicology Research Unit, University Institute of Health Sciences (IUCS), University Institute of Health Sciences-CESPU (IUCS-CESPU), 4585-116 Gandra, Portugal
- CIIMAR-Interdisciplinary Center for Marine and Environmental Research, University of Porto, Avenida General Norton de Matos, 4450-208 Matosinhos, Portugal
| |
Collapse
|
16
|
Kairytė K, Vaickelionienė R, Grybaitė B, Anusevičius K, Mickevičius V, Petrikaitė V. The Effect of 4-(Dimethylamino)phenyl-5-oxopyrrolidines on Breast and Pancreatic Cancer Cell Colony Formation, Migration, and Growth of Tumor Spheroids. Int J Mol Sci 2024; 25:1834. [PMID: 38339112 PMCID: PMC10855844 DOI: 10.3390/ijms25031834] [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: 01/04/2024] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
A series of hydrazones, azoles, and azines bearing a 4-dimethylaminophenyl-5-oxopyrrolidine scaffold was synthesized. Their cytotoxic effect against human pancreatic carcinoma Panc-1 and triple-negative breast cancer MDA-MB-231 cell lines was established by MTT assay. Pyrrolidinone derivatives 3c and 3d, with incorporated 5-chloro and 5-methylbenzimidazole fragments; hydrazone 5k bearing a 5-nitrothien-2-yl substitution; and hydrazone 5l with a naphth-1-yl fragment in the structure significantly decreased the viability of both cancer cell lines. Compounds 3c and 5k showed the highest selectivity, especially against the MDA-MB-231 cancer cell line. The EC50 values of the most active compound 5k against the MDA-MB231 cell line was 7.3 ± 0.4 μM, which were slightly higher against the Panc-1 cell line (10.2 ± 2.6 μM). Four selected pyrrolidone derivatives showed relatively high activity in a clonogenic assay. Compound 5k was the most active in both cell cultures, and it completely disturbed MDA-MB-231 cell colony growth at 1 and 2 μM and showed a strong effect on Panc-1 cell colony formation, especially at 2 μM. The compounds did not show an inhibitory effect on cell line migration by the 'wound-healing' assay. Compound 3d most efficiently inhibited the growth of Panc-1 spheroids and reduced cell viability in MDA-MB-231 spheroids. Considering these different activities in biological assays, the selected pyrrolidinone derivatives could be further tested to better understand the structure-activity relationship and their mechanism of action.
Collapse
Affiliation(s)
- Karolina Kairytė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania; (K.K.); (R.V.); (B.G.); (K.A.); (V.M.)
| | - Rita Vaickelionienė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania; (K.K.); (R.V.); (B.G.); (K.A.); (V.M.)
| | - Birutė Grybaitė
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania; (K.K.); (R.V.); (B.G.); (K.A.); (V.M.)
| | - Kazimieras Anusevičius
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania; (K.K.); (R.V.); (B.G.); (K.A.); (V.M.)
| | - Vytautas Mickevičius
- Department of Organic Chemistry, Kaunas University of Technology, Radvilėnų Rd. 19, LT-50254 Kaunas, Lithuania; (K.K.); (R.V.); (B.G.); (K.A.); (V.M.)
| | - Vilma Petrikaitė
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio Al. 7, LT-10257 Vilnius, Lithuania
- Faculty of Medicine, Lithuanian University of Health Sciences, A. Mickevičiaus 9, LT-44307 Kaunas, Lithuania
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, Sukilėlių Pr. 13, LT-50162 Kaunas, Lithuania
| |
Collapse
|
17
|
Moskalik MY. Monofluoromethylation of N-Heterocyclic Compounds. Int J Mol Sci 2023; 24:17593. [PMID: 38139426 PMCID: PMC10744182 DOI: 10.3390/ijms242417593] [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: 11/30/2023] [Revised: 12/13/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
The review focuses on recent advances in the methodologies for the formation or introduction of the CH2F moiety in N-heterocyclic substrates over the past 5 years. The monofluoromethyl group is one of the most versatile fluorinated groups used to modify the properties of molecules in synthetic medical chemistry. The review summarizes two strategies for the monofluoromethylation of N-containing heterocycles: direct monofluoromethylation with simple XCH2F sources (for example, ICH2F) and the assembly of N-heterocyclic structures from CH2F-containing substrates. The review describes the monofluoromethylation of pharmaceutically important three-, five- and six-membered N-heterocycles: pyrrolidines, pyrroles, indoles, imidazoles, triazoles, benzothiazoles, carbazoles, indazoles, pyrazoles, oxazoles, piperidines, morpholines, pyridines, quinolines and pyridazines. Assembling of 6-fluoromethylphenanthridine, 5-fluoromethyl-2-oxazolines, C5-monofluorinated isoxazoline N-oxides, and α-fluoromethyl-α-trifluoromethylaziridines is also shown. Fluoriodo-, fluorchloro- and fluorbromomethane, FCH2SO2Cl, monofluoromethyl(aryl)sulfoniummethylides, monofluoromethyl sulfides, (fluoromethyl)triphenylphosphonium iodide and 2-fluoroacetic acid are the main fluoromethylating reagents in recent works. The replacement of atoms and entire functional groups with a fluorine atom(s) leads to a change and often improvement in activity, chemical or biostability, and pharmacokinetic properties. The monofluoromethyl group is a bioisoster of -CH3, -CH2OH, -CH2NH2, -CH2CH3, -CH2NO2 and -CH2SH moieties. Bioisosteric replacement with the CH2F group is both an interesting task for organic synthesis and a pathway to modify drugs, agrochemicals and useful intermediates.
Collapse
Affiliation(s)
- Mikhail Yu Moskalik
- A.E. Favorsky Irkutsk Institute of Chemistry, Siberian Division of the Russian Academy of Sciences, 1 Favorsky Street, 664033 Irkutsk, Russia
| |
Collapse
|
18
|
Guillén-Mancina E, García-Lozano MDR, Burgos-Morón E, Mazzotta S, Martínez-Aguado P, Calderón-Montaño JM, Vega-Pérez JM, López-Lázaro M, Iglesias-Guerra F, Vega-Holm M. Repurposing Study of 4-Acyl-1-phenylaminocarbonyl-2-substituted-piperazine Derivatives as Potential Anticancer Agents-In Vitro Evaluation against Breast Cancer Cells. Int J Mol Sci 2023; 24:17041. [PMID: 38069364 PMCID: PMC10706865 DOI: 10.3390/ijms242317041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/21/2023] [Accepted: 11/28/2023] [Indexed: 12/18/2023] Open
Abstract
Breast cancer is the most common type of cancer in women. Although current treatments can increase patient survival, they are rarely curative when the disease is advanced (metastasis). Therefore, there is an urgent need to develop new cytotoxic drugs with a high selectivity toward cancer cells. Since repurposing approved drugs for cancer therapy has been a successful strategy in recent years, in this study, we screened a library of antiviral piperazine-derived compounds as anticancer agents. The compounds included a piperazine ring and aryl urea functions, which are privileged structures present in several anti-breast cancer drugs. The selective cytotoxic activity of a set of thirty-four 4-acyl-2-substituted piperazine urea derivatives against MCF7 breast cancer cells and MCF 10A normal breast cells was determined. Compounds 31, 32, 35, and 37 showed high selective anticancer activity against breast cancer cells and were also tested against another common type of cancer, non-small cell lung cancer (A549 lung cancer cells versus MRC-5 lung normal cells). Compounds 35 and 37 also showed selectivity against lung cancer cells. These results suggest that compounds 35 and 37 may be promising hit compounds for the development of new anticancer agents.
Collapse
Affiliation(s)
- Emilio Guillén-Mancina
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (E.G.-M.); (E.B.-M.); (J.M.C.-M.); (M.L.-L.)
| | - María del Rosario García-Lozano
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (M.d.R.G.-L.); (S.M.); (P.M.-A.); (J.M.V.-P.)
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, 41013 Seville, Spain
| | - Estefanía Burgos-Morón
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (E.G.-M.); (E.B.-M.); (J.M.C.-M.); (M.L.-L.)
| | - Sarah Mazzotta
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (M.d.R.G.-L.); (S.M.); (P.M.-A.); (J.M.V.-P.)
- Department of Chemistry, University of Milan, 20133 Milan, Italy
| | - Pablo Martínez-Aguado
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (M.d.R.G.-L.); (S.M.); (P.M.-A.); (J.M.V.-P.)
- Institute of Biomedicine of Seville (IBiS), Virgen del Rocío University Hospital, CSIC, University of Seville, 41013 Seville, Spain
- Infectious Diseases and Microbiology Clinical Unit, University Hospital Virgen Macarena, 41009 Seville, Spain
- Departament of Medicine, School of Medicine, University of Seville, 41012 Seville, Spain
| | - José Manuel Calderón-Montaño
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (E.G.-M.); (E.B.-M.); (J.M.C.-M.); (M.L.-L.)
| | - José Manuel Vega-Pérez
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (M.d.R.G.-L.); (S.M.); (P.M.-A.); (J.M.V.-P.)
| | - Miguel López-Lázaro
- Department of Pharmacology, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (E.G.-M.); (E.B.-M.); (J.M.C.-M.); (M.L.-L.)
| | - Fernando Iglesias-Guerra
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (M.d.R.G.-L.); (S.M.); (P.M.-A.); (J.M.V.-P.)
| | - Margarita Vega-Holm
- Department of Organic and Medicinal Chemistry, Faculty of Pharmacy, University of Seville, 41012 Seville, Spain; (M.d.R.G.-L.); (S.M.); (P.M.-A.); (J.M.V.-P.)
| |
Collapse
|
19
|
Morales-Salazar I, Garduño-Albino CE, Montes-Enríquez FP, Nava-Tapia DA, Navarro-Tito N, Herrera-Zúñiga LD, González-Zamora E, Islas-Jácome A. Synthesis of Pyrrolo[3,4- b]pyridin-5-ones via Ugi-Zhu Reaction and In Vitro-In Silico Studies against Breast Carcinoma. Pharmaceuticals (Basel) 2023; 16:1562. [PMID: 38004428 PMCID: PMC10674953 DOI: 10.3390/ph16111562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/26/2023] Open
Abstract
An Ugi-Zhu three-component reaction (UZ-3CR) coupled in a one-pot manner to a cascade process (N-acylation/aza Diels-Alder cycloaddition/decarboxylation/dehydration) was performed to synthesize a series of pyrrolo[3,4-b]pyridin-5-ones in 20% to 92% overall yields using ytterbium triflate as a catalyst, toluene as a solvent, and microwaves as a heat source. The synthesized molecules were evaluated in vitro against breast cancer cell lines MDA-MB-231 and MCF-7, finding that compound 1f, at a concentration of 6.25 μM, exhibited a potential cytotoxic effect. Then, to understand the interactions between synthesized compounds and the main proteins related to the cancer cell lines, docking studies were performed on the serine/threonine kinase 1 (AKT1) and Orexetine type 2 receptor (Ox2R), finding moderate to strong binding energies, which matched accurately with the in vitro results. Additionally, molecular dynamics were performed between proteins related to the studied cell lines and the three best ligands.
Collapse
Affiliation(s)
- Ivette Morales-Salazar
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Carlos E. Garduño-Albino
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Flora P. Montes-Enríquez
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Dania A. Nava-Tapia
- Laboratorio de Biología Celular del Cáncer, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico;
| | - Napoleón Navarro-Tito
- Laboratorio de Biología Celular del Cáncer, Universidad Autónoma de Guerrero, Chilpancingo de los Bravo 39086, Mexico;
| | - Leonardo David Herrera-Zúñiga
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Eduardo González-Zamora
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| | - Alejandro Islas-Jácome
- Departamento de Química, Universidad Autónoma Metropolitana-Iztapalapa, San Rafael Atlixco 186, Col. Vicentina, Iztapalapa, Ciudad de México 09340, Mexico; (I.M.-S.); (C.E.G.-A.); (F.P.M.-E.); (E.G.-Z.)
| |
Collapse
|
20
|
Chen GQ, Guo HY, Quan ZS, Shen QK, Li X, Luan T. Natural Products-Pyrazine Hybrids: A Review of Developments in Medicinal Chemistry. Molecules 2023; 28:7440. [PMID: 37959859 PMCID: PMC10649211 DOI: 10.3390/molecules28217440] [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] [Revised: 11/02/2023] [Accepted: 11/03/2023] [Indexed: 11/15/2023] Open
Abstract
Pyrazine is a six-membered heterocyclic ring containing nitrogen, and many of its derivatives are biologically active compounds. References have been downloaded through Web of Science, PubMed, Science Direct, and SciFinder Scholar. The structure, biological activity, and mechanism of natural product derivatives containing pyrazine fragments reported from 2000 to September 2023 were reviewed. Publications reporting only the chemistry of pyrazine derivatives are beyond the scope of this review and have not been included. The results of research work show that pyrazine-modified natural product derivatives have a wide range of biological activities, including anti-inflammatory, anticancer, antibacterial, antiparasitic, and antioxidant activities. Many of these derivatives exhibit stronger pharmacodynamic activity and less toxicity than their parent compounds. This review has a certain reference value for the development of heterocyclic compounds, especially pyrazine natural product derivatives.
Collapse
Affiliation(s)
- Guo-Qing Chen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Hong-Yan Guo
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Zhe-Shan Quan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Qing-Kun Shen
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Xiaoting Li
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji 133002, China; (G.-Q.C.); (H.-Y.G.); (Z.-S.Q.); (Q.-K.S.)
| | - Tian Luan
- Department of Pharmacy, Shenyang Medical College, Shenyang 110034, China
| |
Collapse
|
21
|
Rusu A, Moga IM, Uncu L, Hancu G. The Role of Five-Membered Heterocycles in the Molecular Structure of Antibacterial Drugs Used in Therapy. Pharmaceutics 2023; 15:2554. [PMID: 38004534 PMCID: PMC10675556 DOI: 10.3390/pharmaceutics15112554] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2023] [Revised: 10/24/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
Five-membered heterocycles are essential structural components in various antibacterial drugs; the physicochemical properties of a five-membered heterocycle can play a crucial role in determining the biological activity of an antibacterial drug. These properties can affect the drug's activity spectrum, potency, and pharmacokinetic and toxicological properties. Using scientific databases, we identified and discussed the antibacterials used in therapy, containing five-membered heterocycles in their molecular structure. The identified five-membered heterocycles used in antibacterial design contain one to four heteroatoms (nitrogen, oxygen, and sulfur). Antibacterials containing five-membered heterocycles were discussed, highlighting the biological properties imprinted by the targeted heterocycle. In some antibacterials, heterocycles with five atoms are pharmacophores responsible for their specific antibacterial activity. As pharmacophores, these heterocycles help design new medicinal molecules, improving their potency and selectivity and comprehending the structure-activity relationship of antibiotics. Unfortunately, particular heterocycles can also affect the drug's potential toxicity. The review extensively presents the most successful five-atom heterocycles used to design antibacterial essential medicines. Understanding and optimizing the intrinsic characteristics of a five-membered heterocycle can help the development of antibacterial drugs with improved activity, pharmacokinetic profile, and safety.
Collapse
Affiliation(s)
- Aura Rusu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (I.-M.M.); (G.H.)
| | - Ioana-Maria Moga
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (I.-M.M.); (G.H.)
| | - Livia Uncu
- Scientific Center for Drug Research, “Nicolae Testemitanu” State University of Medicine and Pharmacy, 8 Bd. Stefan Cel Mare si Sfant 165, MD-2004 Chisinau, Moldova;
| | - Gabriel Hancu
- Pharmaceutical and Therapeutic Chemistry Department, Faculty of Pharmacy, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Targu Mures, 540142 Targu Mures, Romania; (I.-M.M.); (G.H.)
| |
Collapse
|
22
|
Anichina K, Mavrova A, Vuchev D, Popova-Daskalova G, Bassi G, Rossi A, Montesi M, Panseri S, Fratev F, Naydenova E. Benzimidazoles Containing Piperazine Skeleton at C-2 Position as Promising Tubulin Modulators with Anthelmintic and Antineoplastic Activity. Pharmaceuticals (Basel) 2023; 16:1518. [PMID: 38004384 PMCID: PMC10675210 DOI: 10.3390/ph16111518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/17/2023] [Accepted: 10/20/2023] [Indexed: 11/26/2023] Open
Abstract
Benzimidazole anthelmintic drugs hold promise for repurposing as cancer treatments due to their interference with tubulin polymerization and depolymerization, manifesting anticancer properties. We explored the potential of benzimidazole compounds with a piperazine fragment at C-2 as tubulin-targeting agents. In particular, we assessed their anthelmintic activity against isolated Trichinella spiralis muscle larvae and their effects on glioblastoma (U-87 MG) and breast cancer (MDA-MB-231) cell lines. Compound 7c demonstrated exceptional anthelmintic efficacy, achieving a 92.7% reduction in parasite activity at 100 μg/mL after 48 hours. In vitro cytotoxicity analysis of MDA-MB 231 and U87 MG cell lines showed that derivatives 7b, 7d, and 7c displayed lower IC50 values compared to albendazole (ABZ), the control. These piperazine benzimidazoles effectively reduced cell migration in both cell lines, with compound 7c exhibiting the most significant reduction, making it a promising candidate for further study. The binding mode of the most promising compound 7c, was determined using the induced fit docking-molecular dynamics (IFD-MD) approach. Regular docking and IFD were also employed for comparison. The IFD-MD analysis revealed that 7c binds to tubulin in a unique binding cavity near that of ABZ, but the benzimidazole ring was fitted much deeper into the binding pocket. Finally, the absolute free energy of perturbation technique was applied to evaluate the 7c binding affinity, further confirming the observed binding mode.
Collapse
Affiliation(s)
- Kameliya Anichina
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| | - Anelia Mavrova
- Department of Organic Synthesis, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| | - Dimitar Vuchev
- Department of Infectious Diseases, Parasitology and Tropical Medicine, Medical University, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria; (D.V.); (G.P.-D.)
| | - Galya Popova-Daskalova
- Department of Infectious Diseases, Parasitology and Tropical Medicine, Medical University, 15A Vasil Aprilov Blvd., 4002 Plovdiv, Bulgaria; (D.V.); (G.P.-D.)
| | - Giada Bassi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
- Department of Neurosciences, Imaging and Clinical Sciences, University of G. D’Annunzio, Via Luigi Polacchi, 11, 66100 Chieti, Italy
| | - Arianna Rossi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres, 31, 98166 Messina, Italy
| | - Monica Montesi
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
| | - Silvia Panseri
- Institute of Science, Technology and Sustainability for Ceramics, National Research Council of Italy, Via Granarolo 64, 48018 Faenza, Italy; (G.B.); (A.R.); (M.M.); (S.P.)
| | - Filip Fratev
- Micar Innovation (Micar 21) Ltd., 34B Persenk Str., 1407 Sofia, Bulgaria;
- Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, 1101 N Campbell St., El Paso, TX 79968, USA
| | - Emilia Naydenova
- Department of Organic Chemistry, University of Chemical Technology and Metallurgy, 8 Kliment Ohridski Blvd., 1756 Sofia, Bulgaria;
| |
Collapse
|
23
|
Ji T, Jian X, Chen L, Zeng W, Huo X, Li M, Chen P, Zhang Y, You W, Zhao P. Discovery of novel 6-p-tolyl-3-(3,4,5-trimethoxybenzyl)-7H-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazine derivative as a potent tubulin inhibitor with promising in vivo antitumor activity. Eur J Med Chem 2023; 256:115437. [PMID: 37172475 DOI: 10.1016/j.ejmech.2023.115437] [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: 03/02/2023] [Revised: 04/27/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023]
Abstract
Building on our prior research, a novel series of trimethoxyphenoxymethyl- and trimethoxybenzyl-substituted triazolothiadiazine compounds has been designed and achieved successfully via a direct ring-closing strategy. Initial biological evaluation illustrated that the most active derivative B5 exhibited significant cell growth inhibitory activity toward HeLa, HT-29, and A549 giving the IC50 values of 0.046, 0.57, and 0.96 μM, respectively, which are greater or similar with CA-4. The mechanism study revealed that B5 caused the G2/M phase arrest, induced cell apoptosis in HeLa cells in a concentration-dependent manner, and also showed potent tubulin polymerization inhibitory effect. Meanwhile, B5 exerted significant antivascular activity in the wound-healing and tube formation assays. Most importantly, B5 remarkably inhibited tumor growth without obvious signs of toxicity in A549-xenograft mice model. These observations indicate that 6-p-tolyl-3-(3,4,5-trimethoxybenzyl)-7H-[1,2,4]triazolo[3,4-b][1,3,4]thiadiazine might be considered as the potential lead compound to develop highly efficient anticancer agents with potent selectivity over normal human cells.
Collapse
Affiliation(s)
- Tangyang Ji
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Xieer Jian
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Lin Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Wenbin Zeng
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Xiansen Huo
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Mingxia Li
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Peng Chen
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Yuqi Zhang
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Wenwei You
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China
| | - Peiliang Zhao
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Science, Southern Medical University, Guangzhou, 510515, PR China.
| |
Collapse
|