551
|
Elkotamy MS, Abdelrahman MA, Giovannuzzi S, Alkabbani MA, Nocentini A, Supuran CT, Eldehna WM, Abdel-Aziz HA, Abou-Seri SM. Development of pyrazolo[1,5-a]pyrimidine-grafted coumarins as selective carbonic anhydrase inhibitors and tubulin polymerization inhibitors with potent anticancer activity. Int J Biol Macromol 2025; 303:140462. [PMID: 39884639 DOI: 10.1016/j.ijbiomac.2025.140462] [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: 11/19/2024] [Revised: 12/30/2024] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
This study presents the design, synthesis, and evaluation of a novel series of coumarin-based compounds (9a-t) as potential anticancer agents. The compounds were strategically designed to inhibit cancer-related carbonic anhydrase (CA) isoforms IX and XII and tubulin polymerization. Two approaches were employed for CA inhibition: utilizing the coumarin motif to occlude the CA active site entrance and incorporating zinc-binding groups (sulfonamide, carboxylic acid, and thiol) to interact with the catalytic zinc ion. The target compounds were also designed to inhibit tubulin polymerization by combining the privileged coumarin and pyrazolo[1,5-a]pyrimidine scaffolds. Biological evaluation of the target compounds (9a-t) revealed that sulfonamide-containing derivatives 9h and 9r exhibited potent inhibitory activity in the low nanomolar range against CA IX (Ki = 23 and 14 nM, respectively) and CA XII (Ki = 6 and 17 nM, respectively). In NCI-60 human tumor cell line screening, compounds 9k, 9m, and 9q demonstrated broad-spectrum anti-proliferative activity in the five-dose assay with MG-MID values of 7.31 μM, 10.68 μM, and 5.92 μM, respectively. Compound 9m showed significant tubulin polymerization inhibition with an IC50 = 5.28 μM, surpassing the efficacy of colchicine. Cell cycle analysis in MDA-MB-231 breast cancer cells revealed G2/M phase arrest for 9m, which induced significant apoptosis and modulated apoptotic markers. Molecular docking studies provided insights into the binding modes of the compounds with CA IX, CA XII, and tubulin. ADMET and toxicity predictions were performed to assess the drug-like properties of the compounds. These findings pave the way for further optimization of the coumarin scaffold to develop dual inhibitors of carbonic anhydrase IX/XII and tubulin polymerization.
Collapse
Affiliation(s)
- Mahmoud S Elkotamy
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829, Egypt
| | - Mohamed A Abdelrahman
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829, Egypt; Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Simone Giovannuzzi
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Firenze, Italy
| | - Mahmoud Abdelrahman Alkabbani
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Egyptian-Russian University, Badr City, Cairo 11829, Egypt
| | - Alessio Nocentini
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Firenze, Italy
| | - Claudiu T Supuran
- Department of NEUROFARBA, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Polo Scientifico, Via U. Schiff 6, 50019 Firenze, Italy
| | - Wagdy M Eldehna
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Kafrelsheikh University, Kafrelsheikh, P.O. Box 33516, Egypt.
| | - Hatem A Abdel-Aziz
- Department of Applied Organic Chemistry, National Research Center, Dokki, Cairo 12622, Egypt; Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Pharos University in Alexandria, Canal El Mahmoudia Street, Alexandria 21648, Egypt.
| | - Sahar M Abou-Seri
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo 11562, Egypt.
| |
Collapse
|
552
|
Shlyk NP, Yurchenko EA, Leshchenko EV, Chingizova EA, Chingizov AR, Chausova VE, Kirichuk NN, Khudyakova YV, Pivkin MV, Antonov AS, Popov RS, Isaeva MP, Yurchenko AN. The secondary metabolites of the alga-derived fungus Aspergillus niveoglaucus КММ 4176 and their antimicrobial and antibiofilm activities. J Antibiot (Tokyo) 2025; 78:314-329. [PMID: 39984736 DOI: 10.1038/s41429-025-00811-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 02/23/2025]
Abstract
Marine alga-derived fungal strain КММ 4176 was identified as Aspergillus niveoglaucus based on ITS region BenA, CaM and RPB2 gene sequence analysis. The anthraquinone derivatives emodin anthrone (1) and 4-hydroxyemodin anthrone (2), chromone derivative aloesone (3), and indole diketopiperazine alkaloid neoechinulin B (4) were isolated from the ethyl acetate extract of this fungus. In addition, UPLC MS data analysis of the KMM 4176 extract showed the presence of 17 echinulin-family alkaloids, as well as their biogenetic precursor cyclo(L-alanyl-L-tryptophyl) and a number of polyketide compounds. Emodin anthrone and 4-hydroxyemodin anthrone were found as inhibitors of biofilm formation by Staphylococcus aureus with half-maximal inhibitory concentrations (IC50) of 5.5 µM and 23.7 µM, respectively. Moreover, emodin anthrone (1) and 4-hydroxyemodin anthrone (2) inhibited staphylococcal sortase A activity with IC50 of 9.2 µM and 37.6 µM, respectively. Aloesone (3) also inhibited S. aureus biofilm formation but was less active. The first data on neoechinulin B (4) antibiofilm activity and sortase A inhibition were obtained. The positive effects of the isolated compounds on the growth of HaCaT keratinocytes infected with S. aureus were also observed.
Collapse
Affiliation(s)
- Nadezhda P Shlyk
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
- Far Eastern Federal University, Vladivostok, 690922, Russian Federation
| | - Ekaterina A Yurchenko
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Elena V Leshchenko
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Ekaterina A Chingizova
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Artur R Chingizov
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Viktoria E Chausova
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Natalya N Kirichuk
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Yuliya V Khudyakova
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Mikhail V Pivkin
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Alexandr S Antonov
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Roman S Popov
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Marina P Isaeva
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation
| | - Anton N Yurchenko
- G.B. Elyakov Paсific Institute of Bioorganic Chemistry, Far Eastern Branch of the Russian Academy of Sciences, 159 Prospect 100-letiya Vladivostoka, Vladivostok, 690022, Russian Federation.
| |
Collapse
|
553
|
Wang J, Guo Y, Hu J, Peng J. STING Activation in Various Cell Types in Metabolic Dysfunction-Associated Steatotic Liver Disease. Liver Int 2025; 45:e70063. [PMID: 40116753 DOI: 10.1111/liv.70063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/07/2025] [Accepted: 02/28/2025] [Indexed: 03/23/2025]
Abstract
BACKGROUND During the hepatic histological progression in metabolic dysfunction-associated steatotic liver disease (MASLD), the immunological mechanisms play a the pivotal role, especially when progressing to metabolic dysfunction-associated steatohepatitis (MASH). The discovery of the stimulator of interferon genes (STING) marked a significant advancement in understanding the immune system. METHODS We searched literature on STING involved in MASLD in PubMed to summarise the role of intrahepatic or extrahepatic STING signal pathways and the potential agonists or inhibitors of STING in MASLD. RESULTS Besides inflammation and type I interferon response induced by STING activation in the intrahepatic or extrahepatic immune cells, STING activation in hepatocytes leads to protein aggregates and lipid deposition. STING activation in hepatic macrophages inhibits autophagy in hepatocytes and promotes hepatic stellate cells (HSCs) activation. STING activation in HSCs promotes HSC activation and exacerbates liver sinusoidal endothelial cells (LSECs) impairment. However, it was also reported that STING activation in hepatic macrophages promotes lipophagy in hepatocytes and STING activation in HSCs leads to HSC senescence. STING activation in LSEC, inhibits angiogenesis. For extrahepatic tissue, STING signalling participates in the regulation of the intestinal permeability, intestinal microecology and insulin action in adipocytes, which were all involved in the pathogenesis of MASLD. CONCLUSION There're plenty of STING ligands in MASLD. How STING activation affects the intercellular conversation in MASLD deserves thorough investigation.
Collapse
Affiliation(s)
- JingJing Wang
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yue Guo
- Department of Nephropathy, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Hu
- Department of Nephropathy, The Seventh People's Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinghua Peng
- Institute of Liver Diseases, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Key Laboratory of Liver and Kidney Diseases (Shanghai University of Traditional Chinese Medicine), Ministry of Education, Shanghai, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| |
Collapse
|
554
|
Lee SH, Yoo S, Kim SH, Kim YM, Han SI, Lee H. Nature-inspired surface modification strategies for implantable devices. Mater Today Bio 2025; 31:101615. [PMID: 40115053 PMCID: PMC11925587 DOI: 10.1016/j.mtbio.2025.101615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/17/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Medical and implantable devices are essential instruments in contemporary healthcare, improving patient quality of life and meeting diverse clinical requirements. However, ongoing problems such as bacterial colonization, biofilm development, foreign body responses, and insufficient device-tissue adhesion hinder the long-term effectiveness and stability of these devices. Traditional methods to alleviate these issues frequently prove inadequate, necessitating the investigation of nature-inspired alternatives. Biomimetic surfaces, inspired by the chemical and physical principles found in biological systems, present potential opportunities to address these challenges. Recent breakthroughs in manufacturing techniques, including lithography, vapor deposition, self-assembly, and three-dimensional printing, now permit precise control of surface properties at the micro- and nanoscale. Biomimetic coatings can diminish inflammation, prevent bacterial adherence, and enhance stable tissue integration by replicating the antifouling, antibacterial, and adhesive properties observed in creatures such as geckos, mussels, and biological membranes. This review emphasizes the cutting-edge advancements in biomimetic surfaces for medical and implantable devices, outlining their design methodologies, functional results, and prospective clinical applications. Biomimetic coatings, by integrating biological inspiration with advanced surface engineering, have the potential to revolutionize implantable medical devices, providing safer, more lasting, and more effective interfaces for prolonged patient benefit.
Collapse
Affiliation(s)
- Soo-Hwan Lee
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sungjae Yoo
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sung Hoon Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Young-Min Kim
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Sang Ihn Han
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyojin Lee
- Biomaterials Research Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Division of Biomedical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
- SKKU-KIST, Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon, Gyeonggi, 16419, Republic of Korea
| |
Collapse
|
555
|
Tripathi N, Roy A, Agnivesh PK, Bhardwaj N, Kumari S, Kalia NP, Jain SK. Pharmacological Evaluation of Bakuchiol From Psoralea corylifolia L. as Potent Antimicrobial Agent Against Staphylococcus aureus. Chem Biodivers 2025; 22:e202401272. [PMID: 39585311 DOI: 10.1002/cbdv.202401272] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 11/24/2024] [Accepted: 11/25/2024] [Indexed: 11/26/2024]
Abstract
The surge in multidrug resistance in Staphylococcus aureus is the pressing need to identify novel alternatives to combat antimicrobial resistance effectively. Bakuchiol is a bioactive prenylated phenolic meroterpene largely abundant in the seeds of Psoralea corylifolia. In this study, we present the biological assessment of bakuchiol derived from P. corylifolia as an antimicrobial agent. S. aureus, a significant opportunistic pathogen, attracts global concern for its biofilm formation and resilience against numerous antibiotics, escaping antibiotic pressure. The primary screening of bakuchiol as an antimicrobial agent against S. aureus delineated its potential as a strong bactericidal agent with a minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of 2 and 8 µg/mL, respectively. Importantly, bakuchiol also exhibited low toxicity against HepG2 cells, showing a favorable selectivity index (SI) of 14.4. Furthermore, bakuchiol demonstrated comparable activity (MIC = 2 µg/mL) against a laboratory-generated ciprofloxacin-resistant mutant of S. aureus. Bakuchiol could significantly inhibit the biofilm formation of S. aureus in a dose-dependent manner with minimum biofilm inhibitory concentration (MBIC)50 of 0.956 µg/mL. Bakuchiol effectively inhibited DNA gyrase supercoiling activity at a concentration eight times the MIC, establishing DNA gyrase inhibition as the mechanism of action for bakuchiol. Our findings suggest bakuchiol as a potential therapeutic agent for S. aureus-mediated nosocomial infections.
Collapse
Affiliation(s)
- Nancy Tripathi
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Arnab Roy
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Puja Kumari Agnivesh
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Nivedita Bhardwaj
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Sanju Kumari
- School of Biomedical Engineering, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| | - Nitin Pal Kalia
- Department of Biological Sciences, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shreyans K Jain
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (BHU), Varanasi, Uttar Pradesh, India
| |
Collapse
|
556
|
Yu X, Jin J, Si Y, Zhang H, Song Z. A peptide-based fluorescent bioprobe for EphA2-overexpressing tumor targeting and image-guided surgical resection. Bioorg Med Chem 2025; 120:118090. [PMID: 39904197 DOI: 10.1016/j.bmc.2025.118090] [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: 11/12/2024] [Revised: 01/22/2025] [Accepted: 01/25/2025] [Indexed: 02/06/2025]
Abstract
Fluorescence-guided surgery (FGS) is an emerging and highly promising surgical technique in clinic. Owing to its real-time and visual characteristics, it assists in achieving clear pictures on lesion site, tumor boundary and degree of metastasis, which will definitely improve surgery accuracy and minimize cancer recurrence as much as possible. Herein, we report a near-infrared fluorescent bioprobe, YK80, which utilizes a modified heptamethine cyanine dye as the fluorophore and a self-assembling peptide targeting Ephrin receptor A2 (EphA2) proteins as the ligand. The design strategy and the synthetic route to YK80 are described, and then optical properties, pharmacokinetics, binding affinity between YK80 and the protein are further investigated. YK80 shows high affinity (KD ≈ 100 nM) with EphA2-expressing cancer cells and excellent targeting ability in mouse models bearing colorectal tumors. Meanwhile, indocyanine green (ICG), the commonly used non-targeted fluorescent contrast agent is employed as the comparison for in vivo experiments. However, ICG owns no such capability towards cancer cells or solid tumors. Thus, YK80 could potentially serve as a targeted contrast agent for image-guided surgery and this successful example will boost the development of medical imaging, surgical methods as well as translational medicine.
Collapse
Affiliation(s)
- Xudong Yu
- Department of Gastroenterology, Yiwu Central Hospital, the Affiliated Yiwu Hospital of Wenzhou Medical University, Yiwu, Zhejiang Province, China.
| | - Jianfei Jin
- Zhejiang Yike Biotech. Co., Ltd, Yiwu, Zhejiang Province, China
| | - Yun Si
- Zhejiang Yike Biotech. Co., Ltd, Yiwu, Zhejiang Province, China
| | - Huanmin Zhang
- Zhejiang Yike Biotech. Co., Ltd, Yiwu, Zhejiang Province, China
| | - Zhegang Song
- Zhejiang Yike Biotech. Co., Ltd, Yiwu, Zhejiang Province, China.
| |
Collapse
|
557
|
M S, Joga R, Gandhi K, Yerram S, Raghuvanshi RS, Srivastava S. Exploring the clinical trials, regulatory insights, and challenges of PROTACs in oncology. Semin Oncol 2025; 52:152339. [PMID: 40253775 DOI: 10.1016/j.seminoncol.2025.152339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/27/2025] [Accepted: 03/05/2025] [Indexed: 04/22/2025]
Abstract
While various targeted therapies exist for cancer, resistance mechanisms remain a significant challenge. Recent advancements in cancer treatment have led to the emergence of proteolysis-targeting chimeras (PROTACs), a promising technology utilizing hetero-bifunctional molecules to target and degrade proteins implicated in cancer progression through the ubiquitin-proteasome system (UPS). PROTACs offer a novel approach, with recent studies and clinical trials demonstrating promising outcomes in degrading endogenous proteins linked to cancer. This work explores classification, regulatory approvals, and ongoing clinical trials of PROTAC technology in cancer management. It emphasizes the importance of regulatory compliance to expedite approvals from relevant authorities. It also highlights challenges and opportunities associated with their implementation. Despite these preliminary efforts, PROTACs show immense potential in effectively addressing cancer. Their ability to target specific proteins for degradation represents a significant advancement in cancer therapeutics, offering new hope for improved outcomes in patient care.
Collapse
Affiliation(s)
- Sowndharya M
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Ramesh Joga
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Kajal Gandhi
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sravani Yerram
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Rajeev Singh Raghuvanshi
- Central Drug Standard Control Organization (CDSCO), Ministry of Health & Family Welfare, Government of India, New Delhi, India
| | - Saurabh Srivastava
- Department of Regulatory Affairs, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India; Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India.
| |
Collapse
|
558
|
Anakök DA, Angeli A, D'Agostino I, Renzi G, Massardi ML, Tavani C, Çete S, Carradori S, Ronca R, Capasso C, Carta F, Supuran CT. A Journey Around Boronic Acids: Sulfonyl Hydrazone-Containing Derivatives as Carbonic Anhydrase Inhibitors. Chem Biol Drug Des 2025; 105:e70108. [PMID: 40205824 DOI: 10.1111/cbdd.70108] [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: 01/23/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Recently, a rising interest in boronic acids and their derivatives was recorded in the Medicinal Chemistry field due to their high versatility and broad applicability as bioactive compounds in several diseases, including cancer and microbial infections. The ability of boronic acid moieties to bind zinc ions was first hypothesized by the inhibitory activity of bortezomib, a boron-containing protease inhibitor, on different isoforms of the Carbonic Anhydrase (CA, EC: 4.2.1.1) enzyme family and then assessed through X-ray crystallographic studies on benzoxaboroles in complex with hCA II. These findings, along with the overexpression of isoforms IX and XII in hypoxic cancer and, in particular, breast cancer, drove us to explore the chemical space around the phenylboronic acids by generating a focused library of 16 derivatives (1-4a-d) decorated with alkyl sulfonyl hydrazones. The compounds were then subjected to stopped flow-based inhibition assays on a panel of hCAs, including the tumor-associated isoforms, revealing low micromolar inhibition constants (KIs) in some cases. However, antiproliferative assays conducted on a human triple-negative breast cancer cell line showed a lack of activity at the tested concentrations.
Collapse
Affiliation(s)
- Deniz A Anakök
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Turkey
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - Andrea Angeli
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - Ilaria D'Agostino
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Department of Pharmacy, "G. D'annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Gioele Renzi
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - Maria Luisa Massardi
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Camilla Tavani
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Servet Çete
- Department of Chemistry, Faculty of Science, Gazi University, Ankara, Turkey
| | - Simone Carradori
- Department of Pharmacy, "G. D'annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Roberto Ronca
- Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Clemente Capasso
- Department of Biology, Agriculture and Food Sciences, National Research Council (CNR), Institute of Biosciences and Bioresources, Naples, Italy
| | - Fabrizio Carta
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| | - Claudiu T Supuran
- NEUROFARBA Department, Pharmaceutical and Nutraceutical Section, University of Florence, Florence, Italy
| |
Collapse
|
559
|
Haroon M, Sultana S, Najibi SA, Wang ET, Michaelson A, Al Muied PSM, Nielsen AE, Mancini RJ. Efflux-Enhanced Imidazoquinolines To Exploit Chemoresistance. ACS OMEGA 2025; 10:12319-12333. [PMID: 40191321 PMCID: PMC11966297 DOI: 10.1021/acsomega.4c11297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 02/19/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025]
Abstract
The imidazoquinoline family of toll-like receptor (TLR) immune cell agonists has long demonstrated moderate anticancer immunogenic effects by activating tumoricidal immune cells and depleting immunosuppressive cells within the tumor microenvironment. At a molecular level, we have also established that several imidazoquinolines traffic from within cancer cells to the extracellular space via P-glycoprotein (P-gp)-mediated efflux, a process commonly upregulated as multidrug-resistant (MDR) cancers acquire chemoresistance. However, imidazoquinoline P-gp efflux has never been deliberately enhanced to exploit this process. This study pioneers efforts to optimize imidazoquinoline efflux, ultimately balancing immunogenic potency alongside functional efflux susceptibility. Starting from an established imidazoquinoline scaffold previously optimized for potency, efflux was significantly enhanced by elaborating the N1 benzylic position with amide- and sulfonamide-linked P-gp affinity fragments consisting of empirically established P-gp substrates as well as computationally predicted P-gp binders. Lead compounds were identified from this series that exhibited enhanced P-gp efflux with functional retention of TLR agonism. Similar to the parent imidazoquinoline scaffold, leads had limited direct cytotoxicity in both treatment-naive and MDR B16 melanoma models and did not significantly affect the efficacy or trafficking of the chemotherapeutic doxorubicin. Efflux-enhanced imidazoquinolines were preferentially expelled from MDR-B16 cells relative to treatment-naive cells, resulting in immunogenicity that was enhanced as a consequence of the acquired MDR phenotype. Because enhanced P-gp-mediated efflux is common to most MDR cancer types, we envision that these results could inspire the design of immunotherapeutic drugs with mechanisms of action that are broadly enhanced in MDR cancers that have failed treatment or acquired resistance to chemotherapeutics.
Collapse
Affiliation(s)
- Muhammad Haroon
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
| | - Sharmin Sultana
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
| | - Seyedeh A. Najibi
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
| | - Emily T. Wang
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
| | - Abbey Michaelson
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
| | - Pranto S. M. Al Muied
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
| | - Amy E. Nielsen
- Astante
Therapeutics Inc., 201
E. Fifth Street, Cincinnati, Ohio 45202, United States
| | - Rock J. Mancini
- Department
of Chemistry and Biochemistry, Miami University, 651 E. High Street, Oxford, Ohio 45056, United States
- Astante
Therapeutics Inc., 201
E. Fifth Street, Cincinnati, Ohio 45202, United States
| |
Collapse
|
560
|
Patil B, Surana S, Shirkhedkar A. Co-crystallization in Antiepileptic Drugs: A Path Toward Better Therapeutic Outcomes. Cureus 2025; 17:e82230. [PMID: 40370920 PMCID: PMC12077019 DOI: 10.7759/cureus.82230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2025] [Indexed: 05/16/2025] Open
Abstract
Pharmaceutical cocrystals are highly valued in drug development as they can enhance active pharmaceutical ingredients (APIs)' physicochemical characteristics without changing their chemical structure. This is especially important for biopharmaceutical classification system (BCS) class II and IV drugs, which have poor water solubility, resulting in low bioavailability. Co-crystallization is the process of forming a crystalline solid where the API and a co-former are combined in a specified stoichiometric proportion within a crystal lattice, mainly stabilized by non-covalent interactions. This method can enhance properties like stability, dissolution rate, solubility, compressibility, and powder flowability and pharmacokinetics, resulting in an improved drug delivery system and therapeutic effectiveness. Pharmaceutical antiepileptic drugs (AEDs) are the main focus of this review. Pharmaceutical characteristics, conventional and advanced cocrystal generation, and assessment techniques, as well as recent developments related to cocrystals, may suggest perception for cocrystal potential, design approaches, and regulatory aspects. The study's overall finding emphasizes the growth of co-crystallization as a significant technique to enhance the drug's performance and also demonstrates its potential as a significant technique in the AED category and its future application.
Collapse
Affiliation(s)
- Bhagyashree Patil
- Pharmaceutics, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, IND
| | - Sanjay Surana
- Pharmacognosy, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, IND
| | - Atul Shirkhedkar
- Pharmaceutical Chemistry, R. C. Patel Institute of Pharmaceutical Education and Research, Shirpur, IND
| |
Collapse
|
561
|
Heng Y, Wang F, Zhang Z, Lin Z, Zhao D, Li Q. PRMT7 Inhibitor SGC3027 Enhances Radiotherapy Efficacy via Activating ATM Kinase in Non-Small Cell Lung Carcinoma. Radiat Res 2025; 203:284-292. [PMID: 40015317 DOI: 10.1667/rade-24-00242.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 02/20/2025] [Indexed: 03/01/2025]
Abstract
Non-small-cell lung cancer (NSCLC) is the leading cause of tumor-related death in humans. Radiotherapy is a crucial strategy for NSCLC treatment, although its effectiveness is limited by the radio-resistance of tumor cells. Our current research finds that the protein arginine methyltransferase 7 (PRMT7) is upregulated in NSCLC and correlates with poor prognosis. Pharmacological inhibition of PRMT7 by SGC3027, a specific small-molecule PRMT7 inhibitor, suppresses the proliferation, migration and invasion of NSCLC. Combining irradiation with SGC3027 strengthens the impact of irradiation on the biological behaviors of NSCLC cells. We also find that SGC3027 specifically activates ATM kinase and its downstream cell cycle checkpoint kinases to enhance radiobiological response in NSCLC. These findings underscore the promising therapeutic potential of PRMT7 inhibitors as well as combining PRMT7 inhibition with irradiation exposure for effective NSCLC therapies.
Collapse
Affiliation(s)
- Ya Heng
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| | - Feifei Wang
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| | - Zhonghui Zhang
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| | - Zebang Lin
- Department of Thoracic Surgery, the Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, P.R. China
| | - Dahai Zhao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital, Anhui Medical University, Hefei 230601, P.R. China
| | - Qiuling Li
- Institute of Department of Health Sciences and Technology, Institutes of Physical Science and Information Technology, Anhui University, Hefei, 230601, P.R. China
| |
Collapse
|
562
|
Jiang H, Qi J, Wang J, Chen J, Feng D, Yang J, Liu X, Liu M, Zhou X, An Z, Lu Y, Ge C, Wang Y. Terramide A: a novel ironophore targeting Acinetobacter baumannii with mechanistic insights into bacterial iron deprivation. J Antibiot (Tokyo) 2025; 78:295-313. [PMID: 40164737 DOI: 10.1038/s41429-025-00816-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/01/2025] [Accepted: 03/15/2025] [Indexed: 04/02/2025]
Abstract
Acetobacter baumannii poses escalating clinical challenges due to its exceptional adaptability, demanding innovative antimicrobial strategies. This study pioneers an investigation into the antibacterial efficacy and molecular mechanism of Terramide A, a hydroxamate siderophore isolated from Aspergillus terreus, against notorious A. baumannii. Employing a multidisciplinary approach integrating phenotypic characterization with mechanistic interrogation, we demonstrate that Terramide A exerts significant inhibitory effects against A. baumannii and P. aeruginosa, pathogens critically dependent on siderophore-mediated iron acquisition for survival and virulence. Structural characterization underlines the hydroxamate moieties of Terramide A presumably supports its hypothesized role as a fungal siderophore, involving competitive iron sequestration and bacterial homeostasis. Subsequently, multi-omics investigation of susceptible strain AB19606 delineated a metabolic collapse cascade due to iron acquisition competition: (1) impairment of central metabolism and energy production through oxidative phosphorylation (OXPHO) inhibitions; (2) compromised stress adaptation and bacterial flexibility; (3) compensatory overactivation of siderophores biosynthesis and transportation, depleting metabolic intermediates and exacerbating stress; (4) coordinated suppression of virulence determinants, such as secretory systems and biofilm formation. These molecular derangements translated into phenotypic deficits, including quorum sensing, diminished autoinducer peptides production, and morphological/functional abnormalities. In vivo evaluation in a rat skin wound infection model further demonstrated that Terramide A promotes wound healing and mitigates inflammation, supporting its antibacterial efficacy. These findings establish Terramide A as a promising antibacterial agent and provide critical insights into iron-competitive antimicrobial strategies to exploit micro-nutrient deprivation and metabolic dysfunction. However, further research is needed to optimize the siderophore-based scaffold, clarify its mechanisms, and assess therapeutic potential.
Collapse
Affiliation(s)
- Hanxiang Jiang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jiangfeng Qi
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jiwen Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Jiaqin Chen
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Dong Feng
- Nanjing Southern Pharmaceutical Technology Co. Ltd., Nanjing, China
| | - Junbiao Yang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xinna Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Mengqun Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Xvzhe Zhou
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Zhilong An
- Nanjing Southern Pharmaceutical Technology Co. Ltd., Nanjing, China
| | - Yuanyuan Lu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China
| | - Chun Ge
- Department of Pharmacy, Nanjing First Hospital, China Pharmaceutical University, Nanjing, China.
- Department of Clinical Pharmacy, School of Basic Medicine & Clinical Pharmacy, China Pharmaceutical University, Nanjing, China.
| | - Ying Wang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, China.
| |
Collapse
|
563
|
Ray B, Roy KK. Deciphering insights into the binding mechanism and plasticity of Telacebec with M. tuberculosis cytochrome bcc-aa3 supercomplex through an unbiased molecular dynamics simulation, free-energy analysis, and DFT study. J Biomol Struct Dyn 2025; 43:2968-2981. [PMID: 38111165 DOI: 10.1080/07391102.2023.2294833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 12/02/2023] [Indexed: 12/20/2023]
Abstract
The cytochrome bcc-aa3 supercomplex, a key component in the electron transport chain pathway involved in bacterial energy production and homeostasis, is a clinically validated target for tuberculosis (TB), leading to Telacebec (Q203). Telacebec is a potent candidate drug under Phase II clinical development for the treatment of drug-sensitive and drug-resistant TB. Recently, the cryo-electron microscopy structure of this supercomplex from Mycobacterium tuberculosis (Mtb) complexed with Q203 was resolved at 6.9 Å resolution (PDB ID: 7E1W). To understand the binding site (QP site) flexibility and Q203's stability at the QP site of the Mtb cytochrome bcc complex, we conducted molecular dynamics (MD) simulation and free energy analysis on this complex in an explicit hydrated lipid bilayer environment for 500 ns. Through this study, the persistence of a range of direct and indirect interactions was observed over the course of the simulation. The significance of the interactions with His375, Tyr161, Ala178, Ala179, Ile183, His355, Leu356, and Thr313 is underlined. Electrostatic energy was the primary source of the net binding free energy, regardless of the important interacting residues. The overall binding free energy for Q203 was -112.84 ± 7.73 kcal/mol, of which the electrostatic and lipophilic energy contributions were -116.31 ± 1.14 and -21.32 ± 2.35 kcal/mol, respectively. Meanwhile, DFT calculations were utilized to elucidate Q203's molecular properties. Overall, this study deciphers key insights into the cytochrome bcc-aa3 supercomplex with Q203 on the ground of molecular mechanics and quantum mechanics that may facilitate structure-based drug design and optimization for the discovery of the next-generation antitubercular drug(s).
Collapse
Affiliation(s)
- Bedabrata Ray
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, India
| | - Kuldeep K Roy
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun, Uttarakhand, India
| |
Collapse
|
564
|
Flores NG, Fernández‐Aroca DM, Garnés‐García C, Domínguez‐Calvo A, Jiménez‐Suárez J, Sabater S, Fernández‐Aroca P, Andrés I, Cimas FJ, de Cárcer G, Belandia B, Palmero I, Huertas P, Ruiz‐Hidalgo MJ, Sánchez‐Prieto R. The CDK12-BRCA1 signaling axis mediates dinaciclib-associated radiosensitivity through p53-mediated cellular senescence. Mol Oncol 2025; 19:1265-1280. [PMID: 39626031 PMCID: PMC11977655 DOI: 10.1002/1878-0261.13773] [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: 05/08/2024] [Revised: 10/14/2024] [Accepted: 11/15/2024] [Indexed: 04/09/2025] Open
Abstract
Pan-cyclin-dependent-kinase (CDK) inhibitors are a new class of targeted therapies that can act on multiple CDKs, with dinaciclib being one of the most promising compounds. Although used as a monotherapy, an interesting approach could be to combine it with radiotherapy. Here, we show that dinaciclib increases radiosensitivity in some experimental models of lung and colon cancer (A549 or HCT 116) but not in others (H1299 or HT-29). Dinaciclib did not alter serine-protein kinase ATM signalling or cell cycle profiling after ionising-radiation exposure, which have been described for other CDK inhibitors. Interestingly, in terms of apoptosis, although the combination renders a clear increase, no potentiation of the ionising-radiation-induced apoptosis was observed. Mechanistically, inhibition of CDK12 by dinaciclib diminishes BRCA1 expression, which decreases homologous recombination (HR) and probably promotes the nonhomologous end joining repair process (NHEJ), which ultimately promotes the induction of ionising-radiation-associated cellular senescence in a TP53-dependent manner, explaining the lack of effect observed in some experimental models. In conclusion, our report proposes a molecular mechanism, based on the signalling axis CDK12-BRCA1, involved in this newly identified therapeutic effect of dinaciclib, although other players implicated in HR should not be discarded. In addition, our data provide a rationale for more selective and personalised chemo/radiotherapy treatment according to the genetic background of the tumour.
Collapse
Affiliation(s)
- Natalia García Flores
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
| | - Diego M. Fernández‐Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Centre for Genomics and Child Health, Blizard Institute, Faculty of Medicine and DentistryQueen Mary University of LondonUK
| | - Cristina Garnés‐García
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
| | - Andrés Domínguez‐Calvo
- Facultad de BiologíaUniversidad de SevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSpain
| | - Jaime Jiménez‐Suárez
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
| | - Sebastià Sabater
- Servicio de Oncología RadioterápicaComplejo Hospitalario Universitario de AlbaceteSpain
| | - Pablo Fernández‐Aroca
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
| | - Ignacio Andrés
- Servicio de Oncología RadioterápicaComplejo Hospitalario Universitario de AlbaceteSpain
| | - Francisco J. Cimas
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
- Área de Bioquímica y Biología Molecular, Facultad de MedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Guillermo de Cárcer
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
- Departamento de Biología del CáncerInstituto de Investigaciones Biomédicas Sols‐Morreale (CSIC‐UAM)MadridEspaña
- CSIC Conexión‐Cáncer HubMadridSpain
| | - Borja Belandia
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
- Departamento de Biología del CáncerInstituto de Investigaciones Biomédicas Sols‐Morreale (CSIC‐UAM)MadridEspaña
- CSIC Conexión‐Cáncer HubMadridSpain
| | - Ignacio Palmero
- Laboratorio de Senescencia Celular y Supresión Tumoral, Departamento de Biología del CáncerInstituto de Investigaciones Biomédicas Sols‐Morreale (CSIC‐UAM)MadridSpain
| | - Pablo Huertas
- Facultad de BiologíaUniversidad de SevillaSpain
- Centro Andaluz de Biología Molecular y Medicina Regenerativa‐CABIMERUniversidad de Sevilla‐CSIC‐Universidad Pablo de OlavideSpain
| | - María José Ruiz‐Hidalgo
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
- Área de Bioquímica y Biología Molecular, Facultad de MedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
| | - Ricardo Sánchez‐Prieto
- Laboratorio de Oncología Molecular, Unidad de Medicina Molecular, Instituto de BiomedicinaUniversidad de Castilla‐La ManchaAlbaceteSpain
- Unidad de Biomedicina de la UCLM, Unidad asociada al CSICAlbaceteSpain
- Translational Cancer Research Group, Chronic Diseases and Cancer, Area 3, Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS)MadridSpain
- Departamento de Biología del CáncerInstituto de Investigaciones Biomédicas Sols‐Morreale (CSIC‐UAM)MadridEspaña
- CSIC Conexión‐Cáncer HubMadridSpain
| |
Collapse
|
565
|
Zi CT, Wu YL, Liu ZH, Niu Y, Yuan WJ, Yang ZW, Wang XJ, Sun XL, Yang L, Sheng J. Novel (-)-eigallocatechin-3-gallate-erlotinib conjugates via triazole rings inhibit non-small cell lung cancer cells through EGFR signaling pathway. Bioorg Chem 2025; 157:108263. [PMID: 39938444 DOI: 10.1016/j.bioorg.2025.108263] [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: 11/19/2024] [Revised: 01/19/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
EGFR is frequently overexpressed in non-small cell lung cancer, and EGFR plays a crucial role in the occurrence and progression of malignant tumors. Currently, drug resistance often develops following treatment with EGFR tyrosine kinase inhibitors, such as erlotinib and gefitinib. Therefore, It is essential to investigate new compounds that can effectively target EGFR overexpression. The polyphenols epigallocatechin-3-gallate (EGCG), found in tea, have demonstrated anti-cancer properties. In this study, we linked EGCG and erlotinib through a click reaction using polyglycol to form an EGCG-erlotinib conjugated compounds (EGCG-Erls). We then explored its biological activity through various experiments. The results indicated that the compound 10 exhibited a superior inhibitory effect on NCI-H1975 cells, reduced their cloning and migratory capabilities, promoted cell apoptosis, and inhibited cell cycle progression. Furthermore, it was observed that compound 10 can bind to the EGFR protein and effectively inhibit the expression of phosphorylated EGFR (p-EGFR) and its downstream signaling proteins. Overall, the study suggests that compound 10 may induce apoptosis and inhibit cell proliferation via the EGFR signaling pathway, providing a promising avenue for the development of new EGFR inhibitors.
Collapse
Affiliation(s)
- Cheng-Ting Zi
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Yi-Long Wu
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Zhen-Hao Liu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yun Niu
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Wen-Juan Yuan
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China; Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Zi-Wei Yang
- Research Center for Agricultural Chemistry, College of Science, Yunnan Agricultural University, Kunming 650201, China; Institute of Biofabrication Research, Yunnan Agricultural University, College of Science, Kunming 650201, China
| | - Xuan-Jun Wang
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Xiu-Li Sun
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| | - Liu Yang
- State Key Laboratory of Phytochemistry and Plant Resources in West China, Kunming Institute of Botany, Chinese Academy of Sciences, Kunming 650201, China.
| | - Jun Sheng
- Key Laboratory of Pu-erh Tea Science, Ministry of Education, College of Food Science and Technology, Yunnan Agricultural University, Kunming 650201, China.
| |
Collapse
|
566
|
Rahman SMA, Singh G, Khan MS, Balasubramaniam AK, Monga V. Recent developments of pyrimidine appended HIV-1 non-nucleoside reverse transcriptase inhibitors. Bioorg Chem 2025; 157:108273. [PMID: 40037028 DOI: 10.1016/j.bioorg.2025.108273] [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: 11/13/2024] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 03/06/2025]
Abstract
Acquired Immune Deficiency Syndrome (AIDS) is an ailment that progressively weakens the immune system and is responsible for being the sole cause of 630,000 deaths worldwide in 2023. It is a potentially fatal condition that promotes the growth of malignancies and secondary infection. Viruses like Human Immunodeficiency Virus (HIV-1) and Hepatitis B virus (HBV) employ an enzyme, reverse transcriptase (RT), to replicate their genomes and spread across the host genome. RT has proved to be one of the most important therapeutic targets for the treatment of AIDS as well as for the development of new HIV-1 medications. The pyrimidine nucleus has been described as a dynamic cornerstone in developing new anti-HIV-1 medications and represents a familiar motif found in various marketed anti-HIV-1 drugs, such as diaryl pyrimidines (DAPYs). The rapid emergence of drug-resistant viral strains due to mutations in the HIV-1 RT structure along with their unfavourable pharmacokinetics present new challenges. Recent years have witnessed tremendous progress in the design and discovery of new substituted pyrimidines as potent and selective non-nucleoside reverse transcriptase inhibitors (NNRTIs). Further, the current developments in the field of X-ray crystallography and molecular modeling have remarkably augmented the design strategies, with simultaneous improvement in the resistance profiles. This article comprehensively reviews recent trends in the design and development of pyrimidine-based HIV-1 NNRTIs. The study emphasizes their biological activities, structure-activity relationship, and docking studies to guide the rational design of NNRTIs with desired potency, safety, and efficacy.
Collapse
Affiliation(s)
- S Maheen Abdul Rahman
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India
| | - Gurpreet Singh
- Department of Pharmaceutical Chemistry, ISF College of Pharmacy, GT Road, Ghal Kalan, Moga 142001, India
| | - Mhd Shabbu Khan
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India
| | - Arun Kumar Balasubramaniam
- Department of Pharmaceutical Sciences, Joan M. Lafleur College of Pharmacy and Health Sciences, Texas Southern University, Houston, Texas 77004, USA
| | - Vikramdeep Monga
- Department of Pharmaceutical Sciences and Natural Products, Central University of Punjab, VPO-Ghudda, Bathinda 151401, Punjab, India.
| |
Collapse
|
567
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
568
|
Manzanelli FA, Clemente CM, Campagno LP, Beltramo DM, Robledo SM, Ravetti S, Garro AG. Sodium ibuprofenate: antibacterial activities and potential β-lactamase inhibition in critical Gram-negative bacteria. Future Microbiol 2025; 20:395-407. [PMID: 40059403 PMCID: PMC11980465 DOI: 10.1080/17460913.2025.2475639] [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: 12/23/2024] [Accepted: 03/03/2025] [Indexed: 04/04/2025] Open
Abstract
AIMS To evaluate the antibacterial and antibiofilm activities of sodium ibuprofenate (NaI) and its hypertonic variant (NaIHS) against multidrug-resistant Gram-negative bacteria (MDR-GNB) and explore their potential to inhibit β-lactamase enzymes. METHODS Antibacterial activity was assessed using minimum inhibitory concentration (MIC), minimum bactericidal concentration (MBC), and time-kill assays. Antibiofilm activity was evaluated by measuring bacterial viability and biomass reduction in preformed biofilms. Scanning electron microscopy (SEM) was used to observe membrane effects. Molecular docking and molecular dynamics simulations were conducted to analyze the binding affinity of ibuprofen to the active sites of β-lactamases (CTX-M-15, KPC-2, OXA-23). RESULTS NaI exhibited bactericidal activity at concentrations of 25-75 mm, with Acinetobacter baumannii being the most susceptible. NaCl (≥0.5 M) enhanced bactericidal efficacy and lowered MBCs. Time-kill assays indicated rapid bacterial eradication within 2 hours, with NaIHS achieving similar results at lower concentrations. SEM confirmed membrane disruption. Both formulations reduced bacterial viability in biofilms, with NaIHS showing greater efficiency. In silico studies suggest ibuprofen may inhibit β-lactamases, with enhanced interactions in saline environments. CONCLUSION Sodium ibuprofenate, particularly in its hypertonic form, demonstrates strong antibacterial, antibiofilm, and potential β-lactamase inhibitory activity, making it a promising candidate for treating MDR-GNB infections.
Collapse
Affiliation(s)
- Franco A. Manzanelli
- Centro de Investigaciones y Transferencia de Villa María (CIT VM), Villa María, Argentina
| | - Camila M. Clemente
- Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires (FCEyN-UBA) e Instituto de Química Biológica de la Facultad de Ciencias Exactas y Naturales (IQUIBICEN) CONICET, Ciudad de Buenos Aires, Argentina
- Structural Studies Division, MRC Laboratory of Molecular Biology, Cambridge, UK
| | - Luciana P. Campagno
- Facultad de Ciencias Químicas,Universidad Nacional de Córdoba, UNITEFA, CONICET, Córdoba, Argentina
| | - Dante M. Beltramo
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Sara M. Robledo
- PECET-Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia
| | - Soledad Ravetti
- Centro de Investigaciones y Transferencia de Villa María (CIT VM), Villa María, Argentina
- Instituto Académico Pedagógico de Ciencias Humanas, Universidad Nacional de Villa María, Villa María, Argentina
| | - Ariel G. Garro
- Instituto Académico Pedagógico de Ciencias Humanas, Universidad Nacional de Villa María, Villa María, Argentina
- Ministerio de Producción, Ciencia e Innovación Tecnológica de la Provincia de Córdoba, Córdoba, Argentina
| |
Collapse
|
569
|
Floresta G. Leading designs of peptide-based chemical probes for medical imaging- the dawn of precision diagnostics. Future Med Chem 2025; 17:861-863. [PMID: 40072224 PMCID: PMC12036484 DOI: 10.1080/17568919.2025.2479415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Affiliation(s)
- Giuseppe Floresta
- Department of Drug and Health Sciences, University of Catania, Catania, Italy
| |
Collapse
|
570
|
Chen O, Fu L, Wang Y, Li J, Liu J, Wen Y. Targeting HSP90AA1 to overcome multiple drug resistance in breast cancer using magnetic nanoparticles loaded with salicylic acid. Int J Biol Macromol 2025; 298:139443. [PMID: 39756742 DOI: 10.1016/j.ijbiomac.2024.139443] [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: 07/24/2024] [Revised: 12/31/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
Multiple drug resistance (MDR) remains a major obstacle in effective breast cancer chemotherapy. This study explores the role of HSP90AA1 in driving MDR and evaluates the potential of magnetic nanoparticles (Fe3O4@SA) loaded with salicylic acid (SA) to counteract drug resistance. A comprehensive screening of 200 SA-related target genes identified nine core genes, including HSP90AA1. Pharmacophore analysis revealed that SA interacts with HSP90AA1, a key regulator of mitochondrial K+ channels. Fe3O4@SA nanoparticles demonstrated efficient cellular uptake and lysosomal escape, markedly improving the chemosensitivity of resistant breast cancer cells and promoting apoptosis. In vivo experiments further confirmed the anticancer efficacy of Fe3O4@SA, highlighting its potential as a promising therapeutic strategy to overcome MDR in breast cancer.
Collapse
Affiliation(s)
- Ou Chen
- Department of clinical laboratory, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Linlin Fu
- Neurosurgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yu Wang
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jinggui Li
- Liaoning Jiahe Hospital of Traditional Chinese Medicine, Medical Imaging Center, Shenyang, China
| | - Jun Liu
- Department of cardiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| | - Yanqing Wen
- Department of Pharmacy, The Fourth Affiliated Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
571
|
Hausner SH, Davis RA, Ganguly T, Harris R, Sutcliffe JL. Evaluation of [ 18F]AlF NOTA-5G, an Aluminum [ 18F]fluoride Labeled Peptide Targeting the Cell Surface Receptor Integrin Alpha(v)beta(6) for PET Imaging. Mol Imaging Biol 2025; 27:285-292. [PMID: 39979580 DOI: 10.1007/s11307-025-01989-3] [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: 10/08/2024] [Revised: 02/01/2025] [Accepted: 02/05/2025] [Indexed: 02/22/2025]
Abstract
PURPOSE Peptide-based probes targeting integrin αvβ6 have shown promise in clinical trials for cancer imaging based on the high over-expression of this epithelial-specific cell surface receptor in many cancerous tissues. Recently, the αvβ6-targeting gallium-68 labeled DOTA-5G peptide, [68Ga]Ga DOTA-5G, demonstrated diagnostic value in patients with metastatic pancreatic cancer. To facilitate adoption at sites without access to gallium-68 and take advantage of the characteristics of fluorine-18 through convenient [18F]fluoride chelation chemistry, this study evaluated the fluorine-18 labeled analog, [18F]AlF NOTA-5G, in vitro and in vivo in a tumor mouse model, and compared it to [68Ga]Ga DOTA-5G. PROCEDURES NOTA-5G was synthesized on solid phase and radiolabeled with aluminum [18F]fluoride to generate [18F]AlF NOTA-5G. Cell binding and internalization of [18F]AlF NOTA-5G were evaluated in paired DX3puroβ6 (αvβ6 +) and DX3puro (αvβ6 -), and pancreatic BxPC-3 (αvβ6 +) cells. Imaging (1-6 h) and biodistribution were performed in BxPC-3 tumor-bearing mice. RESULTS [18F]AlF NOTA-5G was obtained in > 93% radiochemical purity. Cell binding was αvβ6-targeted (1 h: 66% bound to DX3puroβ6, vs 2% to DX3puro), and ≥ 50% of bound activity was internalized; analogous to [68Ga]Ga DOTA-5G, PET imaging showed clearly delineated tumors. Excretion remained primarily renal (1 to 4 h: 18.6 to 12.5% ID/g). Tumor uptake remained relatively steady (1 to 4 h: 2.3 ± 0.4 to 1.8 ± 0.6% ID/g - closely matching [68Ga]Ga DOTA-5G with 2.6 ± 0.8 and 2.0 ± 0.6% ID/g at 1 and 2 h), resulting in tumor/pancreas, tumor/liver, and tumor/blood ratios of 18/1, 24/1, and 162/1, respectively (4 h); by comparison, for [68Ga]Ga DOTA-5G the values were 21/1, 20/1, and 22/1 (2 h). CONCLUSIONS [18F]AlF NOTA-5G demonstrated selective αvβ6-targeting and tumor uptake similar to [68Ga]Ga DOTA-5G. The tumor-to-background ratio resulted high-contrast PET images, with an extended imaging window compared to [68Ga]Ga DOTA-5G. The synthesis of [18F]AlF NOTA-5G is currently being optimized for clinical production.
Collapse
Affiliation(s)
- Sven H Hausner
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis, Sacramento, CA, 95817, USA
| | - Ryan A Davis
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Tanushree Ganguly
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA
| | - Rebecca Harris
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis, Sacramento, CA, 95817, USA
| | - Julie L Sutcliffe
- Department of Internal Medicine, Division of Hematology/Oncology, University of California Davis, Sacramento, CA, 95817, USA.
- Department of Biomedical Engineering, University of California Davis, Davis, CA, USA.
- Center for Molecular and Genomic Imaging, University of California Davis, Davis, CA, USA.
| |
Collapse
|
572
|
Dou Y, Liu Y, Han R, Zheng J, Wang L, Hu C, Huang D, He C, Zhang Y, Lin C, Lu C, Wu D, Tang H, He T, Tang L, He Y. Multieffect Specific Nanovesicles for Homing Resistant Tumors and Overcoming Osimertinib-Acquired Resistance in NSCLC. Adv Healthc Mater 2025; 14:e2404087. [PMID: 39967371 DOI: 10.1002/adhm.202404087] [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: 10/30/2024] [Revised: 01/15/2025] [Indexed: 02/20/2025]
Abstract
Acquired resistance to osimertinib (Osi) remains a major obstacle in the treatment of patients with EGFR-mutant non-small cell lung cancer (NSCLC). AXL elevation is a known key mechanism of Osi-resistance, and therapeutic strategies remain scarce. Emerging evidence reveals that an increased intracellular glutathione (GSH) level induces Osi resistance. In this study, a new mechanism is identified by which GSH regulates AXL expression via glutathione peroxidase 4 (GPX4) in Osi-resistant cells. A multifunctional covalent organic framework (COF) nanoplatform for GSH consumption, AXL inhibition, and co-delivery of the AXL inhibitor (Brigatinib) and Osi is creatively constructed to confirm whether Osi sensitivity improves by simultaneously targeting GSH-AXL resistance mechanisms. Furthermore, it is coated, for the first time, the COF carrier system with specific vesicles to precisely home it into resistant tumors, where CDH2 adhesion molecules play a crucial role. The engineered multifunctional antiresistance-specific nanovesicles effectively inhibited the GSH-AXL axis, induced apoptosis in Osi-resistant cells both in vitro and in vivo, and delayed the progression of Osi-resistant tumors. Overall, these findings provide a novel strategy to overcome the Osi-acquired resistance caused by high AXL levels in NSCLC.
Collapse
Affiliation(s)
- Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Rui Han
- Department of Respiratory Disease, Bishan hospital of Chongqing Medical University, Bishan Hospital of Chongqing, Chongqing, 402760, China
| | - Jie Zheng
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
- School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Liping Wang
- Department of pain treatment, The seventh people's Hospital of Chongqing, Chongqing, 401320, China
| | - Chen Hu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Daijuan Huang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
- School of Medicine, Chongqing University, Chongqing, 400044, China
| | - Chao He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Yimin Zhang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Caiyu Lin
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Conghua Lu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Di Wu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huan Tang
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Tingting He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Liling Tang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
- School of Medicine, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
573
|
Salem MA, Abusaif MS, Gohar NA, Ammar YA, Ragab A. Novel 3-Substituted-2H-Chromene Scaffold Based Fluorinated Hydrophobic Fragment as In-Vitro Antiproliferative Agents and Apoptosis Inducers Targeting Both VEGFR-2/BRAF V600E and h-DHFR With Molecular Docking Simulation. Drug Dev Res 2025; 86:e70085. [PMID: 40152211 DOI: 10.1002/ddr.70085] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 02/16/2025] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
Recently, there has been an increasing interest in the use of protein kinase inhibitors as a therapeutic strategy for the treatment of cancer. In this study, a new series of 2H-chromene derivatives (2-5 and 6-8) and 3H-benzo[f]chromene carbohydrazide derivative (9) were synthesized. The structure of the designed derivatives was characterized by IR, 1H/13C NMR, and elemental analysis. Moreover, the cytotoxic activity of the newly synthesized chromenes was evaluated against breast cancer cell lines (MDA-MB-231 and MCF-7) and a cervical cancer cell line (HeLa). The results of these evaluations demonstrated promising activity, ranging from good to moderate. Additionally, the lung fibroblast cell line (WI-38), as a normal cell line, was also utilized to assess the active derivatives' selectivity. Among the compounds tested, chromene derivative 3 demonstrated the highest potency, exhibiting IC50 values of 5.36 ± 0.50, 7.82 ± 0.60, and 9.28 ± 0.70 µM against the MDA-MB 231, MCF-7, and HeLa cell lines, respectively. The potential of chromone 3 as a multi-targeted anticancer agent was assessed by evaluating its activity against BRAF and VEGFR-2. Notably, the most promising chromene derivative 3 demonstrated significant VEGFR2 activity with an IC50 value of 0.224 µM compared to sorafenib's 0.045 µM, while exhibiting inhibitory activity against BRAF with an IC50 value of 1.695 µM relative to Vemurafenib's IC50 value of 0.468 µM. In addition, compound 3 inhibits the DHFR enzyme with an IC50 value of 2.217 ± 0.014 µM, compared to methotrexate (IC50 = 0.4315 ± 0.019 µM). These results revealed that the compound has multifaceted mechanisms of action that may augment its therapeutic effectiveness. In addition, compound 3 causes overexpression of caspase-3 and Bax by 6.13 and 8.85-fold, respectively. It also downregulates the antiapoptotic Bcl-2 level by 0.4775-fold compared to the untreated MDA-MB 231 cells. Flow cytometry analysis of MDA-MB-231 cells indicates that compound 3 induces cell cycle arrest in the G0-G1 phase, with an observed percentage of 73.15%. The in-silico toxicity prediction was evaluated and demonstrated a good toxicity profile. Finally, molecular docking studies supported these findings by confirming strong binding affinities of the derivatives to VEGFR-2, BRAF, and DHFR.
Collapse
Affiliation(s)
- Mohamed A Salem
- Department of Chemistry, Faculty of Science and Arts, King Khalid University, Mohail, Saudi Arabia
| | - Moustafa S Abusaif
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Cairo, Egypt
| | - Nirvana A Gohar
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Modern University for Technology and Information (MTI), Cairo, Egypt
| | - Yousry A Ammar
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Cairo, Egypt
| | - Ahmed Ragab
- Department of Chemistry, Faculty of Science (boys), Al-Azhar University, Cairo, Egypt
- Chemistry Department, Faculty of Science, Galala City, Galala University, Suez, Egypt
| |
Collapse
|
574
|
Zůvalová I, Vyhlídalová B, Ondrová K, Nádvorník P, Hrubý J, Illés P, Soural M, Šebela M, Šindlerová L, Kubala L, Mani S, Dvořák Z. Decoding structural determinants of aryl hydrocarbon receptor antagonism by monoterpenoids. Bioorg Chem 2025; 157:108265. [PMID: 39952059 DOI: 10.1016/j.bioorg.2025.108265] [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: 12/14/2024] [Revised: 02/08/2025] [Accepted: 02/08/2025] [Indexed: 02/17/2025]
Abstract
Monocyclic monoterpenoids carvones have been recently identified as atypical negative allosteric modulators of aryl hydrocarbon receptor (AhR). In the current work, we performed AhR antagonist activity screening of 100 natural and synthetic monoterpenoids, and their analogues. Using SAR approach, structural determinants of AhR antagonist activity were assigned, including CO presence/position, planarity, and C3/C5-alkylation. Applying pyramidal selection criteria, including absence of residual agonist activity, no cytotoxicity, strong antagonist potency, and pan-antagonism against diverse AhR agonists, we distilled four lead AhR antagonists (carvacrol, o-cresol, 3-methyl-S-carvone, EN-2). Whereas 3-methyl-S-carvone and EN-2 were non-competitive AhR pan-antagonists, carvacrol and o-cresol were ligand-selective AhR antagonists acting by unclear mechanism. We characterized in detail the effects of lead compounds at cellular functions of AhR, including AhR nuclear translocation, AhR dimerization with ARNT, and the expression of AhR-regulated genes. As a proof of concept, effects of monoterpenoids in the murine macrophages were investigated.
Collapse
Affiliation(s)
- Iveta Zůvalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic.
| | - Barbora Vyhlídalová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Karolína Ondrová
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Petr Nádvorník
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Jiří Hrubý
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Peter Illés
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Miroslav Soural
- Department of Organic Chemistry, Faculty of Science, Palacký University, 17. Listopadu 12, Olomouc 771 46, Czech Republic
| | - Marek Šebela
- Department of Biochemistry, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic
| | - Lenka Šindlerová
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 00, Czech Republic
| | - Lukáš Kubala
- Institute of Biophysics of the Czech Academy of Sciences, Královopolská 135, Brno 612 00, Czech Republic
| | - Sridhar Mani
- Department of Medicine and Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, Olomouc 779 00, Czech Republic.
| |
Collapse
|
575
|
Ghoneim MM, Abdelgawad MA, Elkanzi NAA, Bakr RB. Review of the recent advances of pyrazole derivatives as selective COX-2 inhibitors for treating inflammation. Mol Divers 2025; 29:1789-1820. [PMID: 39014146 DOI: 10.1007/s11030-024-10906-9] [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: 02/09/2024] [Accepted: 05/30/2024] [Indexed: 07/18/2024]
Abstract
Pyrazole heterocycle is regarded as an extremely significant agent for the therapy of inflammation. Celecoxib, lonazolac, deracoxib, and phenylbutazone are examples of commercially approved pyrazole drugs with COX-2 inhibitory potential for curing inflammation. There have been recently many reviews for the biological significance of pyrazole derivatives. This review talks about pyrazole derivatives with anti-inflammatory activity and also sheds the light on the recent updates on pyrazole research with an emphasis on some synthetic pathways utilized to construct this privileged scaffold and structure activity relationship that accounts for the anti-inflammatory activity in an attempt to pave the opportunity for medicinal chemists to develop novel anti-inflammatory agents with better COX-2 selectivity.
Collapse
Affiliation(s)
- Mohammed M Ghoneim
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Ad Diriyah, 13713, Saudi Arabia
| | - Mohamed A Abdelgawad
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jouf University, Sakaka, 72388, Saudi Arabia.
| | - Nadia A A Elkanzi
- Chemistry Department, College of Science, Jouf University, P.O. Box: 2014, Sakaka, Saudi Arabia
| | - Rania B Bakr
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Beni-Suef University, 62514, Beni-Suef, Egypt.
| |
Collapse
|
576
|
Santos AM, Vieira EM, de Jesus JR, Santana Júnior CC, Nascimento Júnior JAC, Oliveira AMS, Araújo AADS, Picot L, Alves IA, Serafini MR. Development and characterization of farnesol complexed in β- and hydroxypropyl-β-cyclodextrin and their antibacterial activity. Carbohydr Res 2025; 550:109406. [PMID: 39864120 DOI: 10.1016/j.carres.2025.109406] [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: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Farnesol (FAR) belongs to terpenes group and is a sesquiterpene alcohol and a hydrophobic compound, which can be extracted from natural sources or obtained by organic chemical or biological synthesis. Recent advances in the field of nanotechnology allow the drawbacks of low drug solubility, which can improve the drug therapeutic index. Therefore, this study aimed to prepare the FAR inclusion complexes with β-cyclodextrin (β-CD) and hydroxypropyl-β-cyclodextrin (HP-β-CD) through freeze-drying method, proposing their physicochemical characterization, comparing their toxicity, and evaluating their in vitro antibacterial activity. Initially, physical mixture and freeze-dried inclusion complexes of FAR/β-CD and FAR/HP-β-CD were obtained in the molar ratio (1:1). The samples were characterized by DSC, TG/DTG, FTIR, PXRD, SEM, pHPZC, and the complexation efficiency were performed by HPLC. In vivo toxicity assay was performed using Tenebrio molitor larvae to determine the LD50 and toxic dose of the samples. Also, it was proposed that the evaluation of the fluorescence suppression of Bovine Serum Albumin and the antibacterial activity. The complexation of FAR was evidenced with β-CD and HP-β-CD by the characterization techniques analyzed. The complexation efficiency of FAR/β-CD and FAR/HP-β-CD were 73,53 % and 74.12 %, respectively. The inclusion complexes demonstrated a reduction in toxicity, as evidenced by lower toxic and LD50 doses compared to the free FAR. The inclusion complexes induced conformational changes in BSA, suggesting that they reached the subdomains containing tryptophan residues. In terms of antibacterial activity, FAR/β-CD and FAR/HP-β-CD did not exhibit significant MIC results compared to free FAR, except for FAR/HP-β-CD against S. aureus ATCC 25923.
Collapse
Affiliation(s)
- Anamaria Mendonça Santos
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil.
| | - Edileuza Marcelo Vieira
- Research Laboratory in Biomaterials, LPBio, Department of Chemistry, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | - Jemmyson Romário de Jesus
- Research Laboratory in Biomaterials, LPBio, Department of Chemistry, Federal University of Viçosa, Viçosa, Minas Gerais, Brazil
| | | | | | - Ana Maria Santos Oliveira
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil
| | | | - Laurent Picot
- La Rochelle Université, UMR CNRS 7266 LIENSs, La Rochelle, France
| | - Izabel Almeida Alves
- Postgraduate Program in Pharmaceutical Sciences, University of the State of Bahia and Faculty of Pharmacy, Federal University of Bahia, Salvador, Bahia, Brazil
| | - Mairim Russo Serafini
- Postgraduate Program in Health Sciences, Federal University of Sergipe, Aracaju, Sergipe, Brazil; Postgraduate Program in Pharmaceutical Sciences, Federal University of Sergipe, São Cristóvão, Sergipe, Brazil.
| |
Collapse
|
577
|
Lou H, Feng M, Al-Tamimi Z, Kuczera K, Hageman MJ. Predicting Distribution Coefficients (LogD) of Cyclic Peptides Using Molecular Dynamics Simulations. Pharm Res 2025; 42:613-622. [PMID: 40140127 DOI: 10.1007/s11095-025-03850-2] [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: 02/03/2025] [Accepted: 03/15/2025] [Indexed: 03/28/2025]
Abstract
PURPOSE The distribution coefficient (LogD) is a critical property for oral peptide drug design. In this study, we focused on cyclic peptides (octreotide and its analogs) and aimed to determine their LogD values at four pHs using both the simulation and experimental approaches. METHODS For the experimental approach, the shake-flask method with LCMS quantification was employed to determine LogD values. For the simulation approach, the partition coefficient (LogP) was obtained from the solvation free energy calculations using molecular dynamics (MD) simulation. The LogD values were then calculated from the obtained LogP values considering the predicted pKa and ionization states of each peptide residue. More peptide properties such as polar surface area (PSA), number of intramolecular hydrogen bonds, solvent accessible surface area (SASA), and radius of gyration (Rg) were also calculated with the aid of MD simulation. RESULTS For a total of 28 LogD values across four pHs, the predicted values from the simulation under the OPLS-AA forcefield agreed with the experimental values, with an average deviation of 1.39 ± 0.86 log units, displaying better predictions compared to the data generated under the CHARMM forcefield or using commercial software. In addition, the analysis of PSA, SASA, and Rg data suggested the peptides exhibited some conformational flexibility in both aqueous and organic phases. CONCLUSIONS The method developed in this study can predict the LogD values at a wide pH range covering multiple formulation/physiological conditions and therefore can provide insights into designing oral peptide drugs, especially for early-stage projects.
Collapse
Affiliation(s)
- Hao Lou
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA.
| | - Mei Feng
- Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, KS, 66047, USA
| | - Zahraa Al-Tamimi
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA
| | - Krzysztof Kuczera
- Department of Chemistry, University of Kansas, Lawrence, KS, 66045, USA
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Michael J Hageman
- Department of Pharmaceutical Chemistry, University of Kansas, Lawrence, KS, 66047, USA.
- Biopharmaceutical Innovation and Optimization Center, University of Kansas, Lawrence, KS, 66047, USA.
| |
Collapse
|
578
|
Dilweg MA, Gorostiola González M, de Ruiter MD, Meijboom NJ, van Veldhoven JPD, Liu R, Jespers W, van Westen GJP, Heitman LH, IJzerman AP, van der Es D. Exploring novel dilazep derivatives as hENT1 inhibitors and potentially covalent molecular tools. Purinergic Signal 2025; 21:289-316. [PMID: 38879664 PMCID: PMC12061832 DOI: 10.1007/s11302-024-10026-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Accepted: 05/22/2024] [Indexed: 05/09/2025] Open
Abstract
The human equilibrative nucleoside transporter 1 (SLC29A1, hENT1) is a solute carrier that modulates the passive transport of nucleosides and nucleobases, such as adenosine. This nucleoside regulates various physiological processes, such as vasodilation and -constriction, neurotransmission and immune defense. Marketed drugs such as dilazep and dipyridamole have proven useful in cardiovascular afflictions, but the application of hENT1 inhibitors can be beneficial in a number of other diseases. In this study, 39 derivatives of dilazep's close analogue ST7092 were designed, synthesized and subsequently assessed using [3H]NBTI displacement assays and molecular docking. Different substitution patterns of the trimethoxy benzoates of ST7092 reduced interactions within the binding pocket, resulting in diminished hENT1 affinity. Conversely, [3H]NBTI displacement by potentially covalent compounds 14b, 14c, and 14d resulted in high affinities (Ki values between 1.1 and 17.5 nM) for the transporter, primarily by the ability of accommodating the inhibitors in various ways in the binding pocket. However, any indication of covalent binding with amino acid residue C439 remained absent, conceivably as a result of decreased nucleophilic residue reactivity. In conclusion, this research introduces novel dilazep derivatives that are active as hENT1 inhibitors, along with the first high affinity dilazep derivatives equipped with an electrophilic warhead. These findings will aid the rational and structure-based development of novel hENT1 inhibitors and pharmacological tools to study hENT1's function, binding mechanisms, and its relevance in (patho)physiological conditions.
Collapse
Affiliation(s)
- Majlen A Dilweg
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Marina Gorostiola González
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Martijn D de Ruiter
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Nadine J Meijboom
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Jacobus P D van Veldhoven
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Rongfang Liu
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Willem Jespers
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Gerard J P van Westen
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Laura H Heitman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
- Oncode Institute, Leiden, The Netherlands
| | - Adriaan P IJzerman
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands
| | - Daan van der Es
- Division of Drug Discovery and Safety, Leiden Academic Centre for Drug Research, Leiden University, PO Box 9502, Leiden, The Netherlands.
| |
Collapse
|
579
|
Ailawadhi S, Pafundi D, Peterson J. Advances and future directions in radiopharmaceutical delivery for cancer treatment. Expert Rev Anticancer Ther 2025; 25:351-361. [PMID: 40019019 DOI: 10.1080/14737140.2025.2472859] [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: 09/09/2024] [Revised: 01/29/2025] [Accepted: 02/24/2025] [Indexed: 03/01/2025]
Abstract
INTRODUCTION Targeted radiopharmaceutical therapies (RPTs) have emerged as a promising approach for the precise treatment of various cancers. Delivering ionizing radiation directly to cancer cells while sparing surrounding healthy tissue, radiopharmaceuticals offer enhanced efficacy and reduced toxicity compared to conventional external beam radiation therapy (i.e. photons and electrons). AREAS COVERED In the current era of personalized cancer care, the appropriate choice of RPTs for a clinical condition and the specific patient's care needs to be better understood. Several available RPT agents with their respective clinical applicability along with rapidly ongoing research in this field have now given RPTs the ability to lend themselves to a personalized medicine focus. This review provides an overview of recent advancements in RPT, including nuclide selection and development, molecular targeting strategies, radiopharmaceutical development, and clinical applications. EXPERT OPINION We discuss the underlying principles, challenges, and opportunities for future development. Furthermore, we explore emerging technologies and future directions in the field, highlighting the potential impact on personalized cancer care.
Collapse
Affiliation(s)
| | - Deanna Pafundi
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Jennifer Peterson
- Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, USA
| |
Collapse
|
580
|
Baranova AA, Alferova VA, Korshun VA, Tyurin AP. Imaging-based profiling for elucidation of antibacterial mechanisms of action. Biotechnol Appl Biochem 2025; 72:542-569. [PMID: 39467068 DOI: 10.1002/bab.2681] [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: 05/29/2024] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
In this review, we aim to summarize experimental data and approaches to identifying cellular targets or mechanisms of action of antibacterials based on imaging techniques. Imaging-based profiling methods, such as bacterial cytological profiling, dynamic bacterial morphology imaging, and others, have become a useful research tool for mechanistic studies of new antibiotics as well as combinations with conventional ones and other therapeutic options. The main methodological and experimental details and obtained results are summarized and discussed. The review covers the literature up to February 2024.
Collapse
Affiliation(s)
- Anna A Baranova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vera A Alferova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
| | - Vladimir A Korshun
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anton P Tyurin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| |
Collapse
|
581
|
Nguyen VT, Birhanu BT, Miguel-Ruano V, Kim C, Batuecas M, Yang J, El-Araby AM, Jiménez-Faraco E, Schroeder VA, Alba A, Rana N, Sader S, Thomas CA, Feltzer R, Lee M, Fisher JF, Hermoso JA, Chang M, Mobashery S. Restoring susceptibility to β-lactam antibiotics in methicillin-resistant Staphylococcus aureus. Nat Chem Biol 2025; 21:482-489. [PMID: 39060390 DOI: 10.1038/s41589-024-01688-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 06/26/2024] [Indexed: 07/28/2024]
Abstract
Infections by Staphylococcus aureus have been treated historically with β-lactam antibiotics. However, these antibiotics have become obsolete in methicillin-resistant S. aureus by acquisition of the bla and mec operons. The presence of the β-lactam antibiotic is detected by the sensor domains of BlaR and/or MecR, and the information is transmitted to the cytoplasm, resulting in derepression of the antibiotic-resistance genes. We hypothesized that inhibition of the sensor domain would shut down this response system, and β-lactam susceptibility would be restored. An in silico search of 11 million compounds led to a benzimidazole-based hit and, ultimately, to the boronate 4. The X-ray structure of 4 is covalently engaged with the active-site serine of BlaR. Compound 4 potentiates by 16- to 4,096-fold the activities of oxacillin and of meropenem against methicillin-resistant S. aureus strains. The combination of 4 with oxacillin or meropenem shows efficacy in infected mice, validating the strategy.
Collapse
Affiliation(s)
- Van T Nguyen
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Biruk T Birhanu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Vega Miguel-Ruano
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Choon Kim
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Mayte Batuecas
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Jingdong Yang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Amr M El-Araby
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Eva Jiménez-Faraco
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Valerie A Schroeder
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Alejandra Alba
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Neha Rana
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Safaa Sader
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Caitlyn A Thomas
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Rhona Feltzer
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Mijoon Lee
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Jed F Fisher
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Juan A Hermoso
- Department of Crystallography and Structural Biology, Instituto de Química-Física 'Blas Cabrera', Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Mayland Chang
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Shahriar Mobashery
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
582
|
Wandji SR, Abshire DA, Davis JE, Tavakoli AS, Pope R. Substance use disorders among African-American men in the rural south: A scoping review. J Ethn Subst Abuse 2025; 24:327-352. [PMID: 37655703 DOI: 10.1080/15332640.2023.2248029] [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] [Indexed: 09/02/2023]
Abstract
African American (AA) men in the rural South may be at high risk for experiencing adverse health outcomes from substance use (SU). We conducted a scoping review to explore the research on SU among rural AA men in the rural South of the United States (US). Ten articles addressed the following thematic areas pertaining to SU: factors associated with SU (n = 6), associations between substance use and health outcomes (n = 2), and the influence of impulsivity on SU (n = 2). Additional research on SU among AA men in the rural South is needed, particularly pertaining to treatment-related considerations.
Collapse
Affiliation(s)
| | | | - Jean E Davis
- University of South Carolina, Columbia, South Carolina
| | | | - Robert Pope
- Dominican University of California, San Rafael, California
| |
Collapse
|
583
|
Xiao Y, Luo J, Wang J, Lu L, Prakash O, Srivastava S, Muddassir M, Kumar A. New 1,4-naphthalenedicarboxylic acid- and 3,5-bis(benzimidazol-2-yl)pyridine-appended d 10-configuration-based coordination polymers as recoverable turn-off luminescent sensors for tetracycline. Dalton Trans 2025; 54:5888-5895. [PMID: 40085162 DOI: 10.1039/d5dt00025d] [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: 03/16/2025]
Abstract
Coordination polymers (CPs) possessing Zn2+ and Cd2+ display interesting photoluminescent properties, thereby finding utility as luminescent sensors to detect traces of molecular substances. Herein, new Cd(II) and Zn(II)-based coordination polymers (CPs) with formulae [Cd3(L)3(3,5-bipy)6(H2O)3·H2L·5H2O] (1) and [Zn(HL)2(3,5-bipy)2] (2) were synthesized utilizing 1,4-naphthalenedicarboxylic acid (H2L) as the main ligand and 3,5-bis(benzimidazol-2-yl)pyridine (3,5-bipy) as the co-ligand and characterized. These CPs exhibited luminescent properties. Upon exciting their aqueous suspensions at 360 nm, both 1 and 2 exhibited broad emissions at 461 nm and 457 nm, respectively, with 1 displaying more intense emission than 2. Furthermore, employing these CPs as luminescent sensors for sensing antibiotics in aqueous media suggest that both 1 and 2 display selective and sensitive sensing towards tetracycline (TCY). 1 and 2 demonstrated limit of detection (LOD) values of 6.21 × 10-6 M and 1.07 × 10-5 M, respectively, accompanied with Stern-Volmer constants (Ksv) of 2.08 × 104 M-1 and 1.24 × 104 M-1. Additionally, recoveries in the luminescent responses of both 1 and 2 were successfully achieved through titration-back experiments using salicylic acid (SA). The integrated experimental and computational techniques suggest that the decline in emission intensity of CPs in the presence of antibiotics is due to energy/charge transfer and interaction with TCY, inducing minor alterations in the framework. These alterations form a non-fluorescent ground state complex (GSC) that alters the electron communication and decreases the photoluminescent intensity. Also, restoration in the emissive responses of TCY@CP arises due to the SA-assisted release of TCY from TCY@CP, which leads to restoration of the emissive responses.
Collapse
Affiliation(s)
- Yao Xiao
- School of Chemistry and Environmental Engineering, Sichuan University of Science & Engineering, Zigong, 643000, PR China.
| | - Jing Luo
- School of Chemistry and Environmental Engineering, Sichuan University of Science & Engineering, Zigong, 643000, PR China.
| | - Jun Wang
- School of Chemistry and Environmental Engineering, Sichuan University of Science & Engineering, Zigong, 643000, PR China.
| | - Lu Lu
- School of Chemistry and Environmental Engineering, Sichuan University of Science & Engineering, Zigong, 643000, PR China.
| | - Om Prakash
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226007, India.
| | - Shreya Srivastava
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226007, India.
| | - Mohd Muddassir
- Department of Chemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abhinav Kumar
- Department of Chemistry, Faculty of Science, University of Lucknow, Lucknow 226007, India.
| |
Collapse
|
584
|
Xu Y, Zhang F, Li M, Zhang H, Yuan Y, Lin S, Yao L, Xu S, Yuan T, Yao H. Organelle-targeted BODIPY-conjugated platinum(IV) anticancer prodrugs for overcoming drug resistance. Dalton Trans 2025; 54:5849-5858. [PMID: 40079227 DOI: 10.1039/d4dt03249g] [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: 03/14/2025]
Abstract
Platinum-based chemotherapy, despite being a cornerstone of cancer treatment, faces significant challenges due to acquired drug resistance. To address this issue, we have designed three organelle-targeting platinum(IV) prodrugs conjugated with BODIPY fluorophores, enabling spatiotemporal control through green light irradiation. These BODIPY-Pt(IV) conjugates exhibit excellent stability in PBS buffer, demonstrating resilience under physiological conditions. Upon light exposure, these prodrugs undergo rapid activation, releasing axial ligands and generating cytotoxic platinum(II) species at specific cellular locations. In vitro studies across various cancer cell lines, including those resistant to conventional platinum therapies, show high efficacy of these prodrugs, attributed to their organelle-targeted delivery and fast photoactivation. Furthermore, the intrinsic fluorescence of these Pt(IV) prodrugs enables real-time tracking of their cellular distribution, providing valuable insights into their mechanism of action. Overall, our research presents a novel strategy that combines photoactivation of Pt(IV) prodrugs with organelle-specific targeting to overcome platinum drug resistance, enhancing the therapeutic efficacy of platinum-based prodrugs and offering a promising avenue for precision cancer therapy.
Collapse
Affiliation(s)
- Yuelun Xu
- Institute of Flow Chemistry and Engineering, School of Chemistry and Materials, Jiangxi Normal University, Nanchang 330022, China.
| | - Fengling Zhang
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Miaomiao Li
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Huimin Zhang
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Yuhong Yuan
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Siying Lin
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Liuxuan Yao
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Shunfang Xu
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Tao Yuan
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| | - Houzong Yao
- Institute of Flow Chemistry and Engineering, School of Chemistry and Materials, Jiangxi Normal University, Nanchang 330022, China.
- Jiangxi Province Key Laboratory of Natural and Biomimetic Drugs Research, School of Health, Jiangxi Normal University, Nanchang 330022, China
| |
Collapse
|
585
|
Gmedhin H, Schaefer S, Corrigan N, Wu P, Gu Z, Lenardon MD, Boyer C. Effect of Defined Block Sequence Terpolymers on Antifungal Activity and Biocompatibility. Macromol Biosci 2025; 25:e2400429. [PMID: 39764700 DOI: 10.1002/mabi.202400429] [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: 09/04/2024] [Revised: 12/16/2024] [Indexed: 04/15/2025]
Abstract
Invasive fungal infections cause over 3.7 million deaths worldwide annually, underscoring the critical need for new antifungal agents. Developing selective antifungal agents is challenging due to the shared eukaryotic nature of both fungal and mammalian cells. Toward addressing this, synthetic polymers designed to mimic host defense peptides are promising new candidates for combating fungal infections. This study investigates well-defined multiblock terpolymers with specific arrangements of cationic, hydrophobic, and hydrophilic groups, as potential antifungal agents. The block sequence in these copolymers significantly impacts their minimum inhibition concentration (MIC) against Candida albicans and biocompatibility. Furthermore, compared to their statistical counterparts, these block polymers exhibit lower MIC values in certain instances. Notably, triblock terpolymers containing a central hydrophobic block present an enhanced antifungal efficacy and biocompatibility. These findings highlight the potential of block sequence-controlled polymers as a versatile platform for developing customized and targeted antifungal therapies.
Collapse
Affiliation(s)
- Hatu Gmedhin
- Cluster for Advanced Macromolecular Design (CAMD) and Australian Centre for NanoMedicine (ACN), School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, 2052, Australia
- School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Sebastian Schaefer
- Cluster for Advanced Macromolecular Design (CAMD) and Australian Centre for NanoMedicine (ACN), School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, 2052, Australia
- School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Nathaniel Corrigan
- Cluster for Advanced Macromolecular Design (CAMD) and Australian Centre for NanoMedicine (ACN), School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Peifeng Wu
- School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| | - Zi Gu
- Cluster for Advanced Macromolecular Design (CAMD) and Australian Centre for NanoMedicine (ACN), School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - Megan D Lenardon
- School of Biotechnology and Biomolecular Sciences, UNSW, Sydney, NSW, 2052, Australia
| | - Cyrille Boyer
- Cluster for Advanced Macromolecular Design (CAMD) and Australian Centre for NanoMedicine (ACN), School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
- School of Chemical Engineering, UNSW, Sydney, NSW, 2052, Australia
| |
Collapse
|
586
|
Lu J, Yu D, Li H, Qin P, Chen H, Chen L. Promising natural products targeting protein tyrosine phosphatase SHP2 for cancer therapy. Phytother Res 2025; 39:1735-1757. [PMID: 38558278 DOI: 10.1002/ptr.8185] [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: 09/01/2023] [Revised: 02/27/2024] [Accepted: 02/29/2024] [Indexed: 04/04/2024]
Abstract
The development of Src homology-2 domain containing protein tyrosine phosphatase-2 (SHP2) inhibitors is a hot spot in the research and development of antitumor drugs, which may induce immunomodulatory effects in the tumor microenvironment and participate in anti-tumor immune responses. To date, several SHP2 inhibitors have made remarkable progress and entered clinical trials for the treatment of patients with advanced solid tumors. Multiple compounds derived from natural products have been proved to influence tumor cell proliferation, apoptosis, migration and other cellular functions, modulate cell cycle and immune cell activation by regulating the function of SHP2 and its mutants. However, there is a paucity of information about their diversity, biochemistry, and therapeutic potential of targeting SHP2 in tumors. This review will provide the structure, classification, inhibitory activities, experimental models, and antitumor effects of the natural products. Notably, this review summarizes recent advance in the efficacy and pharmacological mechanism of natural products targeting SHP2 in inhibiting the various signaling pathways that regulate different cancers and thus pave the way for further development of anticancer drugs targeting SHP2.
Collapse
Affiliation(s)
- Jiani Lu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danmei Yu
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongtao Li
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Pengcheng Qin
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Pharmacy, Henan University, Kaifeng, China
| | - Hongzhuan Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Chen
- Shanghai Frontiers Science Center of TCM Chemical Biology, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
587
|
Liang P, Wang Y, Liu J, Huang H, Li Y, Kang J, Li G, Wu H. Identification and Exploration of Immunity-Related Genes and Natural Products for Alzheimer's Disease Based on Bioinformatics, Molecular Docking, and Molecular Dynamics. Immun Inflamm Dis 2025; 13:e70166. [PMID: 40192032 PMCID: PMC11973734 DOI: 10.1002/iid3.70166] [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: 07/23/2024] [Revised: 01/14/2025] [Accepted: 02/23/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Recent research highlights the immune system's role in AD pathogenesis and promising prospects of natural compounds in treatment. This study explores immunity-related biomarkers and potential natural products using bioinformatics, machine learning, molecular docking, and kinetic simulation. METHODS Differentially expressed genes (DEGs) in AD were analyzed using GSE5281 and GSE132903 datasets. Important AD module genes were identified using a weighted co-expression algorithm (WGCNA), and immune-related genes (IRGs) were obtained from the ImmPortPortal database. Intersecting these genes yielded important IRGs. Then, the least absolute shrinkage and selection operator (LASSO) and other methods screened common immune-related AD markers. Biological pathways were explored through Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), and Gene Set Enrichment Analysis (GSEA). The accuracy of these markers was assessed by subject operator signature (ROC) curves and validated in the GSE122063 dataset. The datasets was then subjected to immunoinfiltration analysis. Multiple compound databases were used to analyze core Chinese medicines and components. Molecular docking and kinetic simulation verification were used for further verification. RESULTS A total of 1360 differential genes and 5 biomarkers (PGF, GFAP, GPI, SST, NFKBIA) were identified, showing excellent diagnostic efficiency. GSEA revealed markers associated with Oxidative phosphorylation, Nicotine addiction, and Hippo signaling pathway. Immune infiltration analysis showed dysregulation in multiple immune cell types in AD brains, with significant interactions between markers and 5 immune cell types. A total of 27 possible herbs and 7 core compounds were eventually identified. The binding environment of GPI-luteolin and GPI-stigasterol was relatively stable and showed good affinity. CONCLUSIONS PGF, GFAP, SST, GPI, and NFKBIA were identified for early AD diagnosis, associated with immune cells and pathways in AD brains. 7 promising natural compounds, including luteolin and stigmasterol, were screened for targeting these biomarkers.
Collapse
Affiliation(s)
- Pengpeng Liang
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| | - Yale Wang
- Shenzhen Longgang Second People's HospitalShenzhenChina
| | - Jiamin Liu
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| | - Hai Huang
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| | - Yue Li
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| | - Jinhua Kang
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| | - Guiyun Li
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| | - Hongyan Wu
- Shenzhen HospitalShanghai University of Traditional Chinese MedicineShenzhenChina
| |
Collapse
|
588
|
Almaamari A, Sultan M, Zhang T, Qaed E, Wu S, Qiao R, Duan Y, Ding S, Liu G, Su S. Sigma-1 Receptor Specific Biological Functions, Protective Role, and Therapeutic Potential in Cardiovascular Diseases. Cardiovasc Toxicol 2025; 25:614-630. [PMID: 39937319 DOI: 10.1007/s12012-025-09975-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/03/2025] [Indexed: 02/13/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of morbidity and mortality worldwide, and there is an urgent need for efficient and cost-effective treatments to decrease the risk of CVD. The sigma-1 receptor (S1R) plays a role in the development of cardiac hypertrophy, heart failure, ventricular remodeling, and various other cardiac diseases. Preclinical studies have shown that S1R activation has considerable beneficial effects on the cardiovascular system, and this knowledge might contribute to informing clinical trials associated with the prevention and treatment of CVDs. Therefore, the objective of this review was to investigate the mechanisms of S1R in CVD and how modulation of pathways contributes to cardiovascular protection to facilitate the development of new therapeutic agents targeting the cardiovascular system.
Collapse
Affiliation(s)
- Ahmed Almaamari
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Marwa Sultan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Tao Zhang
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Eskandar Qaed
- Department of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, 730000, People's Republic of China
| | - Shang Wu
- Breast Cancer Center, The Fourth Hospital, Hebei Medical University, Shijiazhuang, 050017, Hebei, People's Republic of China
| | - Ruoqi Qiao
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Yuxin Duan
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Shanshan Ding
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China
| | - Gang Liu
- Heart Center, The First Hospital of Hebei Medical University, Hebei Medical University, Shijiazhuang, China
| | - Suwen Su
- Department of Pharmacology, The Key Laboratory of Neural and Vascular Biology, Ministry of Education, The Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, Hebei, China.
| |
Collapse
|
589
|
Wen X, McLaren DG. High-throughput hit identification with acoustic ejection mass spectrometry. SLAS Technol 2025; 31:100245. [PMID: 39800101 DOI: 10.1016/j.slast.2025.100245] [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: 11/05/2024] [Revised: 12/17/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025]
Abstract
This mini-review provides an overview of recent developments in AEMS supporting hit identification in drug discovery, emphasizing its potential to enhance the quality and efficiency of label-free HTS. Future advancements that may further expand the role of AEMS in the drug discovery process will also be discussed.
Collapse
|
590
|
Liu Y, Zhang C, Zhang X, Wan C, Mao Z. Benzofuran-Chalcone Derivatives as VEGFR-2 Inhibitors: Synthesis and Anticancer Evaluation. Chem Biodivers 2025; 22:e202401991. [PMID: 39545925 DOI: 10.1002/cbdv.202401991] [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: 08/13/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 11/17/2024]
Abstract
The discovery and development of efficient VEGFR-2 inhibitors has become a research hotspot in cancer treatment. In this work, a series of new benzofuran-based chalcone derivatives have been prepared, and in vitro anticancer activities have been evaluated. The results revealed that derivatives showed selective cytotoxic activity against HCC1806, Hela, and A549 cell lines, especially 5c exhibited excellent inhibitory effect on VEFGR-2 (IC50 = 1.07 nM). The molecular docking study indicated that 5c had an obvious binding site with the target VEGFR-2 (PDB ID: 3V6B). Therefore, the benzofuran-based chalcone derivatives could be considered as potent VEGFR-2 inhibitors for further study.
Collapse
Affiliation(s)
- Yixin Liu
- School of Chinese Materia Medica, School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Chunfei Zhang
- School of Chinese Materia Medica, School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Xiao Zhang
- School of Chinese Materia Medica, School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| | - Chunping Wan
- School of Chinese Materia Medica, School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
- Chuxiong Autonomous Prefecture Hospital of Traditional Chinese Medicine, Chuxiong, China
| | - Zewei Mao
- School of Chinese Materia Medica, School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, China
| |
Collapse
|
591
|
Xiong F, Zhang Y, Jiao J, Zhu Y, Mo T, Li Y. Towards new bioactive fluorine-containing 1,3,4-oxadiazole-amide derivatives: synthesis, antibacterial activity, molecular docking and molecular dynamics simulation study. Mol Divers 2025; 29:1079-1089. [PMID: 38900333 DOI: 10.1007/s11030-024-10893-x] [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: 03/22/2024] [Accepted: 05/09/2024] [Indexed: 06/21/2024]
Abstract
Through the approach of molecular hybridization, this study rationally designed and synthesized new trifluoromethyl-1,3,4-oxadiazole amide derivatives, denoted as 1a-1n. The findings reveal that these novel molecules exhibit potent inhibitory effects against various bacterial strains. Thereinto, compounds 1c, 1d, 1i, 1j and 1n, demonstrate relatively superior antimicrobial performance against B. cereus FM314, with a minimum inhibitory concentration (MIC) of 0.03907 μg/mL. Molecular docking analysis suggests the potential importance of the Ser57 and Thr125 amino acid residues (PDB ID: 4EI9) in contributing to the inhibitory activity against B. cereus. The consistency of these results was further corroborated through subsequent molecular dynamics simulations and MMPBSA validations. The insights gained from this study serve to facilitate the rational design and efficient development of novel eco-friendly antimicrobial inhibitors based on the trifluoromethyl-1,3,4-oxadiazole amide scaffold.
Collapse
Affiliation(s)
- Fei Xiong
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China.
| | - Yanjun Zhang
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Jinlong Jiao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Yiren Zhu
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| | - Tianlu Mo
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China.
| | - Yeji Li
- Department of Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, People's Republic of China
| |
Collapse
|
592
|
Saini RS, Vaddamanu SK, Dermawan D, Bavabeedu SS, Khudaverdyan M, Mosaddad SA, Heboyan A. In Silico Docking of Medicinal Herbs Against P. gingivalis for Chronic Periodontitis Intervention. Int Dent J 2025; 75:1113-1135. [PMID: 39127518 PMCID: PMC11976485 DOI: 10.1016/j.identj.2024.06.019] [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: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/29/2024] [Indexed: 08/12/2024] Open
Abstract
OBJECTIVE This study aimed to explore the therapeutic potential of medicinal herbs for chronic periodontitis by examining the molecular interactions between specific herbal compounds and the heme-binding protein of Porphyromonas gingivalis, a key pathogen involved in the disease. METHODS The crystal structure of heme-binding protein was obtained from the Protein Data Bank. Herbal compounds were identified through an extensive literature review. Molecular docking simulations were performed to predict binding affinities, followed by Absorption, Distribution, Metabolism, and Excretion (ADME) parameter prediction. Drug-likeness was assessed based on Lipinski's Rule of Five, and pharmacophore modeling was conducted to identify key molecular interactions. RESULTS The molecular docking simulations revealed that chelidonine, rotenone, and myricetin exhibited significant binding affinities to the heme-binding protein, with docking scores of -6.5 kcal/mol, -6.4 kcal/mol, and -6.1 kcal/mol, respectively. These compounds formed stable interactions with key amino acid residues within the binding pocket. ADME analysis indicated that all 3 compounds had favourable pharmacokinetic properties, with no violations of Lipinski's rules and minimal predicted toxicity. Pharmacophore modeling further elucidated the interaction profiles, highlighting specific hydrogen bonds and hydrophobic interactions critical for binding efficacy. CONCLUSIONS Chelidonine, rotenone, and myricetin emerged as promising therapeutic candidates for chronic periodontitis due to their strong binding affinities, favorable ADME profiles, and lack of significant toxicity. The detailed pharmacophore modeling provided insights into the molecular mechanisms underpinning their inhibitory effects on the heme-binding protein of P. gingivalis. These findings suggest that these compounds have the potential for further development as effective treatments for chronic periodontitis. Future research should focus on in vitro and in vivo validation of these findings to confirm the efficacy and safety of these compounds in biological systems.
Collapse
Affiliation(s)
- Ravinder S Saini
- Department of Dental Technology, COAMS, King Khalid University, Abha, Saudi Arabia
| | | | - Doni Dermawan
- Applied Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Warsaw, Poland
| | - Shashit Shetty Bavabeedu
- Department of Restorative Dental Sciences, College of Dentistry, King Khalid University, Abha, Saudi Arabia
| | - Margarita Khudaverdyan
- The Center for Excellence in Dental Education, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia
| | - Seyed Ali Mosaddad
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India; Student Research Committee, School of Dentistry, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Conservative Dentistry and Bucofacial Prosthesis, Faculty of Odontology, Complutense University of Madrid, Madrid, Spain.
| | - Artak Heboyan
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, India; Department of Prosthodontics, Faculty of Stomatology, Yerevan State Medical University after Mkhitar Heratsi, Yerevan, Armenia; Department of Prosthodontics, School of Dentistry, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
593
|
O'Carroll SM, O'Neill LAJ. Recycling dead bacteria to fuel macrophage immunometabolism. Nat Immunol 2025; 26:529-530. [PMID: 40108422 DOI: 10.1038/s41590-025-02117-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Affiliation(s)
- Shane M O'Carroll
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
594
|
Chen S, Xi J, Xu Y, Zhang Y, Du W, Meng X, Zhang J, Hu X, Wang Y. Extracellular Matrix-Inspired Antibacterial Fibrous Hydrogels Containing Polyhexamethylene Biguanide and Gd 3. Macromol Rapid Commun 2025; 46:e2400696. [PMID: 39632411 DOI: 10.1002/marc.202400696] [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: 09/01/2024] [Revised: 10/30/2024] [Indexed: 12/07/2024]
Abstract
Traditional bulk hydrogels containing antibiotics or metal ions often fall short in effectively treating wound infections due to mechanical limitations, bacterial resistance, and potential cytotoxicity. To address these challenges, an extracellular matrix (ECM)-inspired antibacterial fibrous hydrogel featuring an anisotropic topological structure is developed that closely mimics the natural ECM environment. A novel antibacterial agent, PHMB-VAN-Gd (PVG), is synthesized by reacting polyhexamethylene biguanide (PHMB) with O-Vanillin (VAN) to form the Schiff base ligand PHMB-VAN (PV), followed by coordination with gadolinium ions (Gd3⁺). Employing silk fibroin (SF) as the matrix, the PVG complex is incorporated into fibrous hydrogels through electrospinning, generating structures that replicate the fibrous architecture of the ECM. The resulting SF-PVG fibrous hydrogels exhibited robust antibacterial activity, effectively inhibiting bacterial growth and biofilm formation. Furthermore, the aligned fiber orientation and substantial mechanical strength of these hydrogels facilitated cellular functions, promoting cell attachment and proliferation. This study underscores the significant potential of SF-PVG hydrogels for wound infection treatment.
Collapse
Affiliation(s)
- Siyuan Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jingyun Xi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuanyuan Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuxin Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Wenli Du
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xinyue Meng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jieyu Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xuefeng Hu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
595
|
Tena Meza A, Rivera CA, Shao H, Kelleghan AV, Houk KN, Garg NK. σ-Bond insertion reactions of two strained diradicaloids. Nature 2025; 640:683-690. [PMID: 39938569 DOI: 10.1038/s41586-025-08745-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Accepted: 02/05/2025] [Indexed: 02/14/2025]
Abstract
The development of new synthetic methodologies is instrumental for enabling the discovery of new medicines. The methods that provide efficient access to structural alternatives for aromatic compounds (that is, saturated arene bioisosteres) have become highly coveted1-4. The incorporation of these bioisosteres typically leads to favourable drug-like properties and represents an emerging field of research. Here we report a new synthetic method that furnishes a coveted motif, the bicyclo[2.1.1]hexane scaffold5,6, using mild reaction conditions and an operationally simple protocol. The methodology proceeds through the uncommon coupling of two strained fragments: transiently generated cyclic allenes and bicyclo[1.1.0]butanes, which possess considerable strain energies of about 30 kcal mol-1 (ref. 7) and about 60 kcal mol-1 (ref. 6), respectively. The reaction is thought to proceed by a σ-bond insertion through a diradical pathway. However, rather than requiring an external stimulus to generate radical species, reactivity is thought to arise as a result of innate diradical character present in each reactant. This diradicaloid character8, an underused parameter in reaction design, arises from the severe geometric distortions of each reactant. Our studies provide a means to access functionalized bicyclo[2.1.1]hexanes of value for drug discovery, underscore how geometric distortion of reactants can be used to enable uncommon modes of reactivity and should encourage the further exploration and strategic use of diradicaloids in chemical synthesis.
Collapse
Affiliation(s)
- Arismel Tena Meza
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Christina A Rivera
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Huiling Shao
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - Andrew V Kelleghan
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA
| | - K N Houk
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| | - Neil K Garg
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
596
|
Chowdhury FR, Mercado LD, Kharitonov K, Findlay BL. De novo evolution of antibiotic resistance to Oct-TriA 1. Microbiol Res 2025; 293:128056. [PMID: 39832423 DOI: 10.1016/j.micres.2025.128056] [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: 07/03/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/22/2025]
Abstract
The rise of antimicrobial resistance as a global health concern has led to a strong interest in compounds able to inhibit the growth of bacteria without detectable levels of resistance evolution. A number of these compounds have been reported in recent years, including the tridecaptins, a small family of lipopeptides typified by the synthetic analogue octyl-tridecaptin A1. Hypothesizing that prior reports of negligible resistance evolution have been due in part to limitations in the laboratory evolution systems used, we have attempted to select for resistant mutants using a soft agar gradient evolution (SAGE) system developed by our lab. Following optimization of the media conditions by incorporation of the anti-synaeresis agent xanthan gum into the agar matrix, we successfully evolved high-level resistance to both octyl-tridecaptin A1 as well as the challenging lipopeptide antibiotic polymyxin B. Decreased tridecaptin susceptibility was linked to mutations in outer membrane proteins ompC, lptD and mlaA, with the effect of these genes confirmed through a mix of allelic replacement and knockout studies. Overall, this work demonstrates the robust evolutionary potential of bacteria, even in the face of challenging antimicrobial agents.
Collapse
Affiliation(s)
- Farhan R Chowdhury
- Department of Biology, Concordia University, Montréal, Québec H4B 1R6, Canada
| | - Laura Domínguez Mercado
- Department of Chemistry and Biochemistry, Concordia University, Montréal, Québec H4B 1R6, Canada
| | - Katya Kharitonov
- Department of Biology, Concordia University, Montréal, Québec H4B 1R6, Canada
| | - Brandon L Findlay
- Department of Biology, Concordia University, Montréal, Québec H4B 1R6, Canada; Department of Chemistry and Biochemistry, Concordia University, Montréal, Québec H4B 1R6, Canada.
| |
Collapse
|
597
|
Wójcicka A, Bryndal I, Krupińska M, Wolska A, Milewski J, Pyra A, Becan L, Mączyński M, Matera-Witkiewicz A. 7-(4-Chlorophenyl)-1-hydroxy-5-methylpyrido[3,4-d]pyridazin-4(3H)-one: synthesis, solvatomorphism, in vitro anti-inflammatory and cytotoxic activity studies and in silico analysis. Acta Crystallogr C Struct Chem 2025; 81:198-211. [PMID: 40067331 DOI: 10.1107/s2053229625001858] [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: 11/04/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
The newly obtained compound 7-(4-chlorophenyl)-1-hydroxy-5-methylpyrido[3,4-d]pyridazin-4(3H)-one (CPM) was crystallized as two new variable solvates, namely, the dimethyl sulfoxide monosolvate, C14H10ClN3O2·C2H6SO (I), and the sesquisolvate, C14H10ClN3O2·1.5C2H6SO (II), and their structures were confirmed by single-crystal X-ray diffraction analysis. In previous work, 1-hydroxy-5-methyl-7-phenylpyrido[3,4-d]pyridazin-4(3H)-one (PM) was found to display anticancer activity. In the next step of our studies, we synthesized a new derivative of PM, introducing a Cl atom into the PM structure, obtaining CPM, which showed not only anticancer but also anti-inflammatory activity. CPM and the new semi-products of each step of the synthesis were examined by 1H NMR, 13C NMR and FT-IR spectroscopic analyses, and mass spectrometry. CPM forms (I) and (II) crystallize in the triclinic P1 and monoclinic C2/c space groups, respectively, and differ in the stoichiometry of the CPM and DMSO molecules in the crystal lattice, being 1:1 and 1:1.5 for (I) and (II), respectively. A powder X-ray diffraction analysis was performed only for solvate (I) due to the lack of stability of solvate (II). The potential cytotoxicity of CPM was evaluated against the normal cell lines L929 and RPTEC, as well as the cancer cell lines A172, AGS, CACO-2 and HepG2. The anti-inflammatory activity of CPM was also evaluated using colorimetric assay for the inhibition of COX-1 and COX-2. The same biological tests were carried out for PM to compare the activities of both compounds. The biological studies revealed that CPM does not exhibit more activity than PM. Moreover, in silico analysis of the bioavailability and molecular docking were performed.
Collapse
Affiliation(s)
- Anna Wójcicka
- Department of Organic Chemistry and Pharmaceutical Technology, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Iwona Bryndal
- Department of Organic Chemistry and Pharmaceutical Technology, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Magdalena Krupińska
- Screening of Biological Activity Assays and Collection of Biological Material Laboratory, Wrocław Medical University Biobank, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Aleksandra Wolska
- Screening of Biological Activity Assays and Collection of Biological Material Laboratory, Wrocław Medical University Biobank, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Jakub Milewski
- Screening of Biological Activity Assays and Collection of Biological Material Laboratory, Wrocław Medical University Biobank, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Anna Pyra
- Faculty of Chemistry, University of Wrocław, 14 F. Joliot-Curie, 50-383 Wrocław, Poland
| | - Lilianna Becan
- Department of Organic Chemistry and Pharmaceutical Technology, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Marcin Mączyński
- Department of Organic Chemistry and Pharmaceutical Technology, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| | - Agnieszka Matera-Witkiewicz
- Screening of Biological Activity Assays and Collection of Biological Material Laboratory, Wrocław Medical University Biobank, Faculty of Pharmacy, Wrocław Medical University, 211A Borowska, 50-556 Wrocław, Poland
| |
Collapse
|
598
|
Madesh S, Murugan R, Sau A, Jubie S, Swaroop AK, Rajagopal R, Kumaradoss KM, Arockiaraj J. Nano-Encapsulated Coumarin Derivative, CS-QM2 Inhibits Neoplasm Growth: Experimented in Zebrafish Model. J Biochem Mol Toxicol 2025; 39:e70239. [PMID: 40143626 DOI: 10.1002/jbt.70239] [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: 12/25/2024] [Revised: 03/05/2025] [Accepted: 03/13/2025] [Indexed: 03/28/2025]
Abstract
Cancer remains a significant global health challenge with limited therapeutic success, prompting the need for innovative treatment strategies. This study investigates the anticancer potential of nano-encapsulated metal derivatives (CS-QM2) using a zebrafish model with chemically induced cellular neoplasia. Characterization of CS-QM2 nanoparticles revealed successful synthesis with a high entrapment efficiency and enhanced drug release under acidic conditions. Zebrafish embryos exposed to 7,12-Dimethylbenz[a]anthracene (DMBA) exhibited significant malformations, macrophage accumulation, and abnormal tissue growth, which were markedly reduced by CS-QM2 treatment. CS-QM2 significantly increases intracellular ROS, resulting in higher LPO and induces apoptosis in neoplasm tissues. Furthermore, CS-QM2 treatment alters the tumor microenvironment, reducing macrophage accumulation by decreasing neutral lipid droplets, disrupting TAM metabolic support and limiting their protumorigenic activities. Biochemical assays demonstrated restored activities of antioxidant enzymes SOD, CAT, and GSH. Gene expression analysis showed upregulation of apoptosis and tumor suppressor genes (cas3, p53) and downregulation of inflammatory genes (cox-2, nf-kb). Histological assessment and SEM analysis confirmed reduced neoplasm occurrence and tissue abnormalities. These findings suggest that CS-QM2 nanoparticles effectively inhibit neoplasm growth and modulate the tumor microenvironment through oxidative stress induction and gene expression regulation.
Collapse
Affiliation(s)
- S Madesh
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Raghul Murugan
- Department of Pharmacology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| | - Avra Sau
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - S Jubie
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Ooty, India
| | | | - Rajakrishnan Rajagopal
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Kathiravan Muthu Kumaradoss
- Department of Pharmaceutical Chemistry, SRM College of Pharmacy, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Jesu Arockiaraj
- Toxicology and Pharmacology Laboratory, Department of Biotechnology, Faculty of Science and Humanities, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| |
Collapse
|
599
|
Rijwan, Arjmand F, Tabassum S. Synthesis, spectroscopic, single crystal X-ray diffraction studies of Bilastine, BLA(bpy)2Cu(II) & Zn(II) complexes as anticancer chemotherapeutic agents: Validation of their cytotoxic potency on cancer cell lines. J Mol Struct 2025; 1328:141244. [DOI: 10.1016/j.molstruc.2024.141244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
600
|
Fan Q, Chen H, Wei G, Wei D, Wang Z, Zhang L, Wang J, Zhu M. A review of conjugation technologies for antibody drug conjugates. Antib Ther 2025; 8:157-170. [PMID: 40491604 PMCID: PMC12146483 DOI: 10.1093/abt/tbaf010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/01/2025] [Accepted: 03/25/2025] [Indexed: 06/11/2025] Open
Abstract
Antibody-drug conjugates (ADCs) have gained significant attention in biotherapeutics after several years of steady development. Among the multiple factors influencing ADC design, the conjugation method is one of the most critical parameters. This review classifies conjugation strategies into three categories: non-specific, site-specific but non-selective, and fully site-specific and selective methods. The characteristics; advantages and disadvantages; chemistry, manufacturing, and controls (CMC) potential; and clinical status of each conjugation strategy are discussed in detail. The site-specific and selective methods will yield more homogeneous ADC, which may influence the stability and pharmacokinetics (PK) profile of the ADC and then influence the final therapeutic outcome. Additionally, the review also explores challenges and future directions for developing novel conjugation strategies. This review presents the most prevalent conjugation techniques, providing a valuable resource for researchers in selecting conjugation technologies and advancing ADC development.
Collapse
Affiliation(s)
- Qirui Fan
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Hu Chen
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Guoguang Wei
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Ding Wei
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Zekun Wang
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Lin Zhang
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Jun Wang
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| | - Marie Zhu
- Department of Discovery & Development, WuXi XDC Co., Ltd, 520 Fute North Road, Pilot Free Trade Zone, Pudong New Area, Shanghai, 200131, China
| |
Collapse
|