851
|
Becerra D, Castillo JC. Recent advances in the synthesis of anticancer pyrazole derivatives using microwave, ultrasound, and mechanochemical techniques. RSC Adv 2025; 15:7018-7038. [PMID: 40041378 PMCID: PMC11878059 DOI: 10.1039/d4ra08866b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/21/2025] [Indexed: 03/06/2025] Open
Abstract
Pyrazole and its derivatives have attracted considerable attention in pharmaceutical and medicinal chemistry, as reflected in their presence in numerous FDA-approved drugs and clinical candidates. This review presents a comprehensive analysis of articles published between 2014 and 2024, focusing on the microwave-, ultrasound-, and mechanochemical-assisted synthesis of pyrazole derivatives with anticancer activity. It explores synthetic methodologies, anticancer efficacy, and molecular docking studies, underscoring the significance of pyrazole derivatives in drug discovery and medicinal chemistry. Notably, microwave irradiation stands out as the most widely employed technique, providing high efficiency by significantly reducing reaction times while maintaining moderate temperatures. Ultrasound irradiation serves as a valuable alternative, particularly for processes that require milder conditions, whereas mechanochemical activation, though less frequently employed, offers distinct advantages in terms of sustainability.
Collapse
Affiliation(s)
- Diana Becerra
- Escuela de Ciencias Químicas, Universidad Pedagógica y Tecnológica de Colombia Avenida Central del Norte 39-115 Tunja Colombia
| | - Juan-Carlos Castillo
- Escuela de Ciencias Químicas, Universidad Pedagógica y Tecnológica de Colombia Avenida Central del Norte 39-115 Tunja Colombia
| |
Collapse
|
852
|
Liu XH, Fu CH, Wang J, Wei YC, Tan CX, Weng JQ, Min LJ, Xu TM, Wu N. Novel 5-(Trifluoromethyl)-1,2,4-oxadiazole-Based Pyrimidin-4-ether Histone Deacetylase Inhibitors for Controlling Rust Disease: Design, Synthesis, Activity, and Structure-Activity Relationship. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:4563-4573. [PMID: 39939838 DOI: 10.1021/acs.jafc.4c09039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2025]
Abstract
Rust disease, an important plant pathogen, can lead to reduced crop or fruit production. Trifluoromethyloxadiazole (TFMO) is a class of histone deacetylase inhibitors (HDACs). Herein, a series of 5-(trifluoromethyl)-1,2,4-oxadiazole (TFMO)-based pyrimidin-4-ether derivatives were designed and synthesized. Antirust bioassay results of TFMOs showed that some of them possessed excellent activities against plant rust pathogens, such as Puccinia sorghi, Phakopsora pachyrhizi, and Puccinia rubigo. The most active compound, 3-(5-(((6-(difluoromethyl)pyrimidin-4-yl)oxy)methyl)thiophen-2-yl)-5-(trifluoromethyl)-1,2,4-oxadiazole (XII6), exhibited 50% control against P. pachyrhizi at 0.780 mg/L, which was significantly better than the commercial fungicide azoxystrobin (0%) at the same concentration. The field trial results indicated that the compound exhibited an excellent control effect against P. rubigo at 116 g a.i./ha. The acute toxic results indicated that compound XII6 has low toxicity. Furthermore, the enzyme activity results showed that compound XII6 is a strong, nonselective HDAC inhibitor. Finally, the structure-activity relationship was established, and the compound XII6-HDAC binding mode was carried out based on the crystal structure of hHDAC1, hHDAC4, and hHDAC6. This work provided an excellent fungicide against rust for further optimization.
Collapse
Affiliation(s)
- Xing-Hai Liu
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Chen-Hao Fu
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - Jian Wang
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - You-Chang Wei
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - Cheng-Xia Tan
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jian-Quan Weng
- College of Chemical Engineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Li-Jing Min
- College of Life Science, Key Laboratory of Vector Biology and Pathogen Control of Zhejiang Province, Huzhou University, Huzhou, Zhejiang 313000, China
| | - Tian-Ming Xu
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| | - Ningjie Wu
- Zhejiang Base of National Southern Pesticide Research Centre, Zhejiang Research Institute of Chemical Industry, Hangzhou 310023, China
| |
Collapse
|
853
|
Mulligan M, Boudreau MW, Bouwens BA, Lee Y, Carrell HW, Zhu J, Mousses S, Shapiro DJ, Nelson ER, Fan TM, Hergenrother PJ. Single Dose of a Small Molecule Leads to Complete Regressions of Large Breast Tumors in Mice. ACS CENTRAL SCIENCE 2025; 11:228-238. [PMID: 40028352 PMCID: PMC11869136 DOI: 10.1021/acscentsci.4c01628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 11/25/2024] [Accepted: 12/19/2024] [Indexed: 03/05/2025]
Abstract
Patients with estrogen receptor α positive (ERα+) breast cancer typically undergo surgical resection, followed by 5-10 years of treatment with adjuvant endocrine therapy. This prolonged intervention is associated with a host of undesired side effects that reduce patient compliance, and ultimately therapeutic resistance and disease relapse/progression are common. An ideal anticancer therapy would be effective against recurrent and refractory disease with minimal dosing; however, there is little precedent for marked tumor regression with a single dose of a small molecule therapeutic. Herein we report ErSO-TFPy as a small molecule that induces quantitative or near-quantitative regression of tumors in multiple mouse models of breast cancer with a single dose. Importantly, this effect is robust and independent of tumor size with eradication of even very large tumors (500-1500 mm3) observed. Mechanistically, these tumor regressions are a consequence of rapid induction of necrotic cell death in the tumor and are immune cell independent. If successfully translated to human cancer patients, the benefits of such an anticancer drug that is effective with a single dose would be significant.
Collapse
Affiliation(s)
- Michael
P. Mulligan
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Matthew W. Boudreau
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Brooke A. Bouwens
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Yoongyeong Lee
- Department
of Comparative Biosciences, University of
Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Hunter W. Carrell
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Junyao Zhu
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Spyro Mousses
- Systems
Oncology, Scottsdale, Arizona 85255, United States
| | - David J. Shapiro
- Department
of Biochemistry, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Erik R. Nelson
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Molecular and Integrative Physiology, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Division
of Nutritional Sciences, University of Illinois
at Urbana-Champaign, Urbana, Illinois 61801, United States
- Beckman Institute
for Advanced Science and Technology, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Timothy M. Fan
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
- Department
of Veterinary Clinical Medicine, University
of Illinois at Urbana-Champaign, Urbana, Illinois 61802, United States
| | - Paul J. Hergenrother
- Department
of Chemistry, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Carl
R. Woese Institute for Genomic Biology, University of Illinois at
Urbana-Champaign, Urbana, Illinois 61801, United States
- Cancer Center
at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| |
Collapse
|
854
|
Patel Y, Helmann JD. A mutation in RNA polymerase imparts resistance to β-lactams by preventing dysregulation of amino acid and nucleotide metabolism. Cell Rep 2025; 44:115268. [PMID: 39908144 PMCID: PMC11975431 DOI: 10.1016/j.celrep.2025.115268] [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: 05/28/2024] [Revised: 11/19/2024] [Accepted: 01/15/2025] [Indexed: 02/07/2025] Open
Abstract
Resistance to diverse antibiotics can result from mutations in RNA polymerase (RNAP), but the underlying mechanisms remain poorly understood. In this study, we compare two Bacillus subtilis RNAP mutations: one in β' (rpoC G1122D) that increases resistance to cefuroxime (CEF; a model β-lactam) and one in β (rpoB H482Y) that increases sensitivity. CEF resistance is mediated by a decrease in branched-chain amino acid (BCAA), methionine, and pyrimidine pathways. These same pathways are upregulated by CEF, and their derepression increases CEF sensitivity and antibiotic-induced production of reactive oxygen species. The CEF-resistant rpoC G1122D mutant evades these metabolic perturbations, and repression of the BCAA and pyrimidine pathways may function to restrict membrane biogenesis, which is beneficial when cell wall synthesis is impaired. These findings provide a vivid example of how RNAP mutations, which commonly arise in response to diverse selection conditions, can rewire cellular metabolism to enhance fitness.
Collapse
Affiliation(s)
- Yesha Patel
- Department of Microbiology, Cornell University, Ithaca, NY 14853-8101, USA
| | - John D Helmann
- Department of Microbiology, Cornell University, Ithaca, NY 14853-8101, USA.
| |
Collapse
|
855
|
Trávníček Z, Vančo J, Čajan M, Malina T, Dvořák Z, Lenobel R, Beláková B, Schmid JA. Gold(I) N-heterocyclic carbene complexes show strong proapoptotic, antioxidant and anti-inflammatory effects in A2780 and endothelial cells. Chem Biol Interact 2025; 408:111381. [PMID: 39824433 DOI: 10.1016/j.cbi.2025.111381] [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: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/20/2025]
Abstract
A series of eight gold(I) N-heterocyclic carbene (NHC) complexes [Au(IMes)(Ln)] based on 1,3-bis(2,4,6-trimethylphenyl)imidazole-2-ylidene (IMes) and 7-azaindole derivatives (HLn), where n = 1-8 for HL1 = 5-fluoro-7-azaindole, HL2 = 5-bromo-7-azaindole, HL3 = 3-chloro-7-azaindole, HL4 = 3-iodo-7-azaindole, HL5 = 5-bromo-3-chloro-7-azaindole, HL6 = 5-bromo-3-iodo-7-azaindole, HL7 = 4-chloro-2-methyl-7-azaindole and HL8 = 7-azaindole, was prepared, characterised and studied for their in vitro anti-cancer and anti-inflammatory effects. The complexes showed significant cytotoxicity on human ovarian cancer cell lines (A2780, IC50 ≈ 8-19 μM and A2780R, IC50 ≈ 8-19 μM) and lowered toxicity in normal HaCat and MRC-5 cells. Cellular effects of the selected complexes 1 and 7 were evaluated in A2780 cells using flow cytometry. Moreover, the time-dependent cellular uptake in A2780 cells, a shotgun proteomic analysis, an ESI-MS study of hydrolysis and interactions with l-cysteine and reduced glutathione (GSH) were performed. Complexes 1 and 7 revealed remarkable anti-inflammatory effects via inhibition of NF-κB activity in human endothelial cells.
Collapse
Affiliation(s)
- Zdeněk Trávníček
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic.
| | - Ján Vančo
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Michal Čajan
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - Tomáš Malina
- Czech Advanced Technology and Research Institute, Regional Centre of Advanced Technologies and Materials, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic; Nanotechnology Centre, Centre for Energy and Environmental Technologies, VSB - Technical University of Ostrava, 17. listopadu 2172/15, CZ-708 00, Ostrava-Poruba, Czech Republic
| | - Zdeněk Dvořák
- Department of Cell Biology and Genetics, Faculty of Science, Palacký University, Šlechtitelů 27, CZ-779 00, Olomouc, Czech Republic
| | - René Lenobel
- Laboratory of Growth Regulators, Institute of Experimental Botany of the Czech Academy of Sciences, and Faculty of Science, Palacký University, Šlechtitelů 27, CZ-783 71, Olomouc, Czech Republic
| | - Barbora Beláková
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090, Vienna, Austria
| | - Johannes A Schmid
- Institute of Vascular Biology and Thrombosis Research, Center for Physiology and Pharmacology, Medical University of Vienna, Schwarzspanierstrasse 17, 1090, Vienna, Austria.
| |
Collapse
|
856
|
Lasota M, Jankowski D, Wiśniewska A, Szeleszczuk Ł, Misterka-Kozaka A, Kaczor-Kamińska M, Zarzycka M, Patena M, Brzozowski T. Interaction of Avapritinib with Congo Red in Pancreatic Cancer Cells: Molecular Modeling and Biophysical Studies. Int J Mol Sci 2025; 26:1980. [PMID: 40076604 PMCID: PMC11901030 DOI: 10.3390/ijms26051980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/19/2025] [Accepted: 02/22/2025] [Indexed: 03/14/2025] Open
Abstract
Pancreatic cancer is a malignant tumor with one of the worst prognoses among solid tumors, characterized by resistance to treatment. Therefore, there is an urgent need for new methods of targeted therapy. Previous studies have shown that the overexpression of receptor tyrosine kinases such as c-KIT or PDGFR can increase proliferation, migration, and invasion of cancer cells. The aim of our study was to analyze aggregates between a supramolecular carrier (Congo red, CR) and a tyrosine kinase inhibitor (BLU-258) as well as to investigate the effect of the free inhibitor and its aggregate with Congo red (CR-BLU-258) on selected properties of pancreatic cells, including these cells' viability and three-dimensional cell spheroid cultures. To better understand the interactions between Congo red and BLU-258, we used molecular modeling in addition to biophysical methods. These attempts allowed us to determine the optimal molar ratio, which we used for in vitro studies on pancreatic cancer cell lines. A significantly greater decrease in the viability of the tested 3D cultures was observed after 48 h of incubation with CR-BLU-258, which resulted in a lower IC50 value for the tested co-aggregate compared with BLU-258 alone. Moreover, a higher resistance of PANC-1 and BxPC3 spheroid cells to the tested compounds was noted compared with the 2D culture model. A significantly lower response was observed in 3D cell cultures (BxPC3 and PANC-1) treated with BLU-258 alone compared with the 2D culture. Thus, our results showed that both BLU-258 (alone) and in its co-aggregate with Congo red exhibit anticancer activity, inhibiting the growth of pancreatic cancer cells and reducing their viability, survival, and migration. Both tested compounds also affected the phosphorylation of the selected signaling proteins. We conclude that the selected tyrosine kinase inhibitor (alone) and in its co-aggregate with Congo red exhibit anticancer activity and should be considered as a novel effective therapy against pancreatic cancer.
Collapse
Affiliation(s)
- Małgorzata Lasota
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Daniel Jankowski
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Anna Wiśniewska
- Chair of Pharmacology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
| | - Łukasz Szeleszczuk
- Department of Organic and Physical Chemistry, Faculty of Pharmacy, Medical University of Warsaw, 1 Banacha Street, 02-097 Warsaw, Poland;
| | - Anna Misterka-Kozaka
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Marta Kaczor-Kamińska
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Marta Zarzycka
- Chair of Medical Biochemistry, Jagiellonian University Medical College, 7 Kopernika Street, 31-034 Krakow, Poland; (M.K.-K.); (M.Z.)
| | - Maksym Patena
- SSG of Targeted Therapy and Supramolecular Systems, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland; (D.J.); (M.P.)
| | - Tomasz Brzozowski
- Center for Biomedicine and Interdisciplinary Sciences, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland;
- Department of Physiology, Faculty of Medicine, Jagiellonian University Medical College, 16 Grzegórzecka Street, 31-531 Krakow, Poland
| |
Collapse
|
857
|
Mousso T, Pham K, Drewes R, Babatunde S, Jong J, Krug A, Inserra G, Biber J, Brazzo JA, Gupte S, Bae Y. Survivin in cardiovascular diseases and its therapeutic potential. Vascul Pharmacol 2025; 159:107475. [PMID: 40015658 DOI: 10.1016/j.vph.2025.107475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/20/2025] [Accepted: 02/23/2025] [Indexed: 03/01/2025]
Abstract
Aberrant changes in cell behaviors, such as proliferation, apoptosis, and migration, are some of the contributing factors to the development of various cardiovascular diseases (CVDs) and pathologies, including atherosclerosis, neointimal hyperplasia, and heart failure. In recent years, numerous studies have identified survivin, a key player in the anti-apoptotic pathway, to be extensively involved in modulating cellular functioning in cancer, with many reaching clinical trials. Though seemingly different, CVDs and cancer share abundant similarities regarding abnormal cell modifications and behaviors. This overlap has sparked growing interest in investigating survivin as a therapeutic target in the context of CVD. With new findings emerging rapidly, a comprehensive understanding of survivin's role in cardiovascular pathology is crucial to revealing its full therapeutic potential and translating these discoveries into effective treatments. This review discusses recent findings of survivin in CVDs and related pathologies, focusing on its dual role in promoting proliferation and inhibiting apoptosis, specifically in atherosclerosis, neointimal hyperplasia, stroke, hypertension, myocardial infarction, and heart failure. Across different cell types and pathological contexts, survivin plays a pivotal role throughout the disease progression-from the onset of disease development to the facilitation of compensatory mechanisms post-injury-primarily through its function in regulating cell proliferation and apoptosis. Furthermore, given the limited research on survivin as a therapeutic target for CVDs, potential clinical avenues, including YM155 (a survivin inhibitor) or adenoviral, adeno-associated, and lentiviral vectors, are also discussed. Overall, this review highlights survivin as a promising target for mitigating the detrimental effects of CVDs and to provide new perspectives to advance research on the intervention of CVDs and associated pathologies.
Collapse
Affiliation(s)
- Thomas Mousso
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Khanh Pham
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Rhonda Drewes
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Sefunmi Babatunde
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Jessica Jong
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Alanna Krug
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Gabrielle Inserra
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - John Biber
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Joseph A Brazzo
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Sachin Gupte
- Department of Pharmacology, New York Medical College, Valhalla, NY, USA
| | - Yongho Bae
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA; Department of Biomedical Engineering, School of Engineering and Applied Sciences, University at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
858
|
Petrov V, Aleksandrova T, Pashev A. Synthetic Approaches to Novel DPP-IV Inhibitors-A Literature Review. Molecules 2025; 30:1043. [PMID: 40076268 PMCID: PMC11902039 DOI: 10.3390/molecules30051043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Revised: 02/19/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Dipeptidyl peptidase IV (DPP-IV) is a serine protease whose inhibition has been an object of considerable interest in the context of developing novel treatments for type 2 diabetes mellitus. The development of novel DPP-IV inhibitors from natural or synthetic origin has seen a growing scientific interest in recent years, especially during the SARS-CoV-2 pandemic, when DPP-IV inhibitors were found to be of beneficial therapeutic value for COVID-19 patients. The present manuscript aims to summarize the most recent information on the synthesis of different DPP-IV inhibitors, emphasizing the various heterocyclic scaffolds that can be found in them. Special attention is devoted to DPP-IV inhibitors that are currently in clinical trials. Different synthetic approaches for the construction of DPP-IV inhibitors are discussed, as well as the most recent developments in the field.
Collapse
Affiliation(s)
| | | | - Aleksandar Pashev
- Department of Chemistry and Biochemistry, Faculty of Pharmacy, Medical University—Pleven, 1 St. Kliment Ohridski Str., 5800 Pleven, Bulgaria; (V.P.); (T.A.)
| |
Collapse
|
859
|
Dong Y, Wang C, Gao Y, Xu J, Ping H, Liu F, Niu A. Bleaching of Idesia polycarpa Maxim. Oil Using a Metal-Organic Framework-Based Adsorbent: Kinetics and Adsorption Isotherms. Foods 2025; 14:787. [PMID: 40077489 PMCID: PMC11898808 DOI: 10.3390/foods14050787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/07/2025] [Accepted: 02/21/2025] [Indexed: 03/14/2025] Open
Abstract
Idesia polycarpa Maxim. is a woody oil crop with great potential for edible oil production. While crude oil is rich in pigments, traditional bleaching methods have limited effectiveness in improving its color. In this study, a metal-organic framework (MOF) material, MIL-88B(Fe), was synthesized and used for the bleaching of Idesia polycarpa Maxim. oil. The adsorption selectivity of MIL-88B(Fe) and the adsorption process of carotenoids and chlorophyll were investigated. The results demonstrated that the synthesized MIL-88B(Fe) exhibited excellent bleaching capability, achieving a bleaching rate of 97.67% in 65 min. It showed a strong adsorption effect on pigments, particularly carotenoids. The content of lutein decreased from 118.27 mg/kg to 0.01 mg/kg after 65 min of bleaching. The squalene and phytosterol contents in the oil were minimally affected by the bleaching process, while the free fatty acid content slightly increased due to the high reaction temperature and the adsorbent properties. The adsorption process of MIL-88B(Fe) was best described by a pseudo-first-order kinetic model, indicating that the adsorption was a spontaneous and endothermic chemical process. Moreover, MIL-88B(Fe) demonstrated good safety and reusability, making it a promising novel adsorbent for the bleaching of Idesia polycarpa Maxim. oil and other oils with a high pigment content for the vegetable oil industry.
Collapse
Affiliation(s)
- Yiyang Dong
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
| | - Chengming Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
- Key Laboratory of Environment Correlative Dietology, Huazhong Agricultural University, Ministry of Education, Wuhan 430070, China
| | - Yu Gao
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
| | - Jing Xu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
| | - Hongzheng Ping
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
| | - Fangrong Liu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
| | - Aifeng Niu
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (Y.D.); (Y.G.); (J.X.); (H.P.); (F.L.); (A.N.)
| |
Collapse
|
860
|
Rabb JD, Kruse LE, Lin Q. Design of Cell-Penetrating Domain Antibodies via a Genetically Encoded β-Lactam Amino Acid. Angew Chem Int Ed Engl 2025; 64:e202424076. [PMID: 39777952 PMCID: PMC11850181 DOI: 10.1002/anie.202424076] [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: 12/09/2024] [Revised: 01/05/2025] [Accepted: 01/07/2025] [Indexed: 01/11/2025]
Abstract
Domain antibodies such as monobodies provide an attractive immunoglobin fold for evolving high-affinity protein binders targeting the intracellular proteins implicated in cell signalling. However, it remains a challenge to endow cell permeability to these small and versatile protein binders. Here, we report a streamlined approach combining orthogonal crosslinking afforded by a genetically encoded β-lactam-lysine (BeLaK) and genetic supercharging to generate cell-penetrating monobodies. When introduced to the N-terminal β-strand of a series of supercharged monobodies, BeLaK enabled efficient inter-strand crosslinking with the neighbouring lysine. Compared to its non-crosslinked counterpart, a BeLaK-crosslinked, +18-charged monobody exhibited enhanced thermostability and greater cellular uptake at 40 nM. Moreover, this structurally rigidified, supercharged monobody inhibited ERK1/2 phosphorylation in KYSE-520 esophageal cancer cell line at sub-micromolar concentration, indicating significant endosomal escape after endocytosis. Together, the discovery of this BeLaK-encoded, rigidified immunoglobin fold should facilitate the design of cell-penetrating monobodies targeting intracellular signalling proteins.
Collapse
Affiliation(s)
- Johnathan D Rabb
- Department of Chemistry, State University of New York at Buffalo, Buffalo, New York, 14260-3000, United States
| | - Lucas E Kruse
- Department of Chemistry, State University of New York at Buffalo, Buffalo, New York, 14260-3000, United States
| | - Qing Lin
- Department of Chemistry, State University of New York at Buffalo, Buffalo, New York, 14260-3000, United States
| |
Collapse
|
861
|
Hartog PB, Westerlund AM, Tetko IV, Genheden S. Investigations into the Efficiency of Computer-Aided Synthesis Planning. J Chem Inf Model 2025; 65:1771-1781. [PMID: 39889203 PMCID: PMC11863376 DOI: 10.1021/acs.jcim.4c01821] [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: 10/04/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 02/02/2025]
Abstract
The efficiency of machine learning (ML) models is crucial to minimize inference times and reduce the carbon footprints of models deployed in production environments. Current models employed in retrosynthesis to generate a synthesis route from a target molecule to purchasable compounds are prohibitively slow. The model operates in a single-step fashion in a tree search algorithm by predicting reactant molecules given a product molecule as input. In this study, we investigate the ability of alternative transformer architectures, knowledge distillation (KD), and simple hyper-parameter optimization to decrease inference times of the Chemformer model. Initially, we assess the ability of closely related transformer architectures and conclude that these models under-performed when using KD. Additionally, we investigate the effects of feature-based and response-based KD together with hyper-parameters optimized based on inference sample time and model accuracy. We find that although reducing model size and improving single-step speed are important, our results indicate that multi-step search efficiency is more significantly influenced by the diversity and confidence of single-step models. Based on this work, further research should use KD in combination with other techniques, as multi-step speed continues to prevent proper integration of synthesis planning. However, in Monte Carlo-based (MC) multi-step retrosynthesis, other factors play a crucial role in balancing exploration and exploitation during the search process, often outweighing the direct impact of single-step model speed and carbon footprints.
Collapse
Affiliation(s)
- Peter B.R. Hartog
- Molecular
AI, Discovery Sciences, R&D, AstraZeneca, Pepparedsleden 1, 431 83 Mölndal, Sweden
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich - German Research Center for Environmental
Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Annie M. Westerlund
- Molecular
AI, Discovery Sciences, R&D, AstraZeneca, Pepparedsleden 1, 431 83 Mölndal, Sweden
| | - Igor V. Tetko
- Institute
of Structural Biology, Molecular Targets and Therapeutics Center, Helmholtz Munich - German Research Center for Environmental
Health (GmbH), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
| | - Samuel Genheden
- Molecular
AI, Discovery Sciences, R&D, AstraZeneca, Pepparedsleden 1, 431 83 Mölndal, Sweden
| |
Collapse
|
862
|
Ceylan B, Adam J, Toews S, Kaiser F, Dörr J, Scheppa D, Tants JN, Smart A, Schoth J, Philipp S, Stirnal E, Ferner J, Richter C, Sreeramulu S, Caliskan N, Schlundt A, Weigand JE, Göbel M, Wacker A, Schwalbe H. Optimization of Structure-Guided Development of Chemical Probes for the Pseudoknot RNA of the Frameshift Element in SARS-CoV-2. Angew Chem Int Ed Engl 2025; 64:e202417961. [PMID: 39887818 DOI: 10.1002/anie.202417961] [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: 09/19/2024] [Revised: 12/12/2024] [Accepted: 01/28/2025] [Indexed: 02/01/2025]
Abstract
Targeting the RNA genome of SARS-CoV-2 is a viable option for antiviral drug development. We explored three ligand binding sites of the core pseudoknot RNA of the SARS-CoV-2 frameshift element. We iteratively optimized ligands, based on improved affinities, targeting these binding sites and report on structural and dynamic properties of the three identified binding sites. Available experimental 3D structures of the pseudoknot element were compared to SAXS and NMR data to validate its dominant folding state in solution. In order to experimentally map in silico predicted binding sites, NMR assignments of the majority of nucleobases were achieved by segmental labeling of the pseudoknot RNA and isotope-filtered NMR experiments at 1.2 GHz, demonstrating the value of NMR spectroscopy to supplement modelling and docking data. Optimized ligands with enhanced affinity were shown to specifically inhibit frameshifting without affecting 0-frame translation in cell-free translation assays, establishing the frameshift element as target for drug-like ligands of low molecular weight.
Collapse
Affiliation(s)
- Betül Ceylan
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Jennifer Adam
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Sabrina Toews
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Frank Kaiser
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Jonas Dörr
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
| | - Daniel Scheppa
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
| | - Jan-Niklas Tants
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
- Institute for Molecular Biosciences, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Alexandria Smart
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research (HIRI-HZI), Josef-Schneider-Straße 2/D15, 97080, Würzburg, Germany
- Regensburg Center for Biochemistry (RCB), University of Regensburg, Universitätsstraße 31, 93053, Regensburg
| | - Julian Schoth
- Institute of Pharmaceutical Chemistry, University of Marburg, 35032, Marburg, Germany
| | - Susanne Philipp
- Institute of Pharmaceutical Chemistry, University of Marburg, 35032, Marburg, Germany
| | - Elke Stirnal
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Jan Ferner
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Christian Richter
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Sridhar Sreeramulu
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Neva Caliskan
- Helmholtz Institute for RNA-based Infection Research, Helmholtz Centre for Infection Research (HIRI-HZI), Josef-Schneider-Straße 2/D15, 97080, Würzburg, Germany
- Regensburg Center for Biochemistry (RCB), University of Regensburg, Universitätsstraße 31, 93053, Regensburg
| | - Andreas Schlundt
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
- Institute for Molecular Biosciences, Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
- Institute of Biochemistry, University of Greifswald, 17489, Greifswald, Germany
| | - Julia E Weigand
- Institute of Pharmaceutical Chemistry, University of Marburg, 35032, Marburg, Germany
| | - Michael Göbel
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
| | - Anna Wacker
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Harald Schwalbe
- Institute for Organic Chemistry and Chemical Biology, Goethe University Frankfurt, Max-von-Laue-Straße 7, 60438, Frankfurt am Main, Germany
- Center of Biomolecular Magnetic Resonance (BMRZ), Goethe University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| |
Collapse
|
863
|
Zeng Q, Shi W, Kleij AW. Highly Functional Allyl-Bicyclo[1.1.1]pentane Synthesis by Radical-Initiated Three-Component Stereoselective Allylation. JACS AU 2025; 5:913-921. [PMID: 40017744 PMCID: PMC11863170 DOI: 10.1021/jacsau.4c01129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 01/09/2025] [Accepted: 01/17/2025] [Indexed: 03/01/2025]
Abstract
Rapid access to highly functional allylated BCP synthons can be achieved with good selectivity and yield through a radical, three-component reaction (3CR) regime using various combinations of radical precursors and vinyl-appended heterocycles acting as versatile and modular precursors. This practical process combines mild operating conditions, a wide scope of reaction partners, and the ability to diversify the functionalized allylic scaffolds further using the allyl and other functional groups as synthetic branching points. The developed protocol allows structural alteration and increases the molecular complexity through late-stage drug modifications and drug conjugation approaches. Mechanistic probes demonstrate that the 3CR process is initiated by a selective, light-promoted radical addition to [1.1.1]-propellane, followed by coupling with the vinyl-substituted heterocycle, which represents a formal decarboxylative radical addition/double bond relay/protonation sequence.
Collapse
Affiliation(s)
- Qian Zeng
- Institute
of Chemical Research of Catalonia (ICIQ-Cerca), The Barcelona Institute of Science and Technology, Av. Països Catalans 16, 43007 Tarragona, Spain
- Departament
de Química Física i Inorgànica, Universitat Rovira i Virgili, Marcel·lí Domingo s/n, 43007 Tarragona, Spain
| | - Wangyu Shi
- Institute
of Chemical Research of Catalonia (ICIQ-Cerca), The Barcelona Institute of Science and Technology, Av. Països Catalans 16, 43007 Tarragona, Spain
- Departament
de Química Física i Inorgànica, Universitat Rovira i Virgili, Marcel·lí Domingo s/n, 43007 Tarragona, Spain
| | - Arjan W. Kleij
- Institute
of Chemical Research of Catalonia (ICIQ-Cerca), The Barcelona Institute of Science and Technology, Av. Països Catalans 16, 43007 Tarragona, Spain
- Catalan
Institute of Research and Advanced Studies (ICREA), Pg. Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
864
|
Uguen M, Shell DJ, Silva M, Deng Y, Li F, Szewczyk MM, Yang K, Zhao Y, Stashko MA, Norris-Drouin JL, Waybright JM, Beldar S, Rectenwald JM, Mordant AL, Webb TS, Herring LE, Arrowsmith CH, Ackloo S, Gygi SP, McGinty RK, Barsyte-Lovejoy D, Liu P, Halabelian L, James LI, Pearce KH, Frye SV. Potent and selective SETDB1 covalent negative allosteric modulator reduces methyltransferase activity in cells. Nat Commun 2025; 16:1905. [PMID: 39994194 PMCID: PMC11850789 DOI: 10.1038/s41467-025-57005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/04/2025] [Indexed: 02/26/2025] Open
Abstract
A promising drug target, SETDB1, is a dual methyl-lysine (Kme) reader and methyltransferase implicated in cancer and neurodegenerative disease progression. To help understand the role of the triple Tudor domain (3TD) of SETDB1, its Kme reader, we first identify a low micromolar potency small molecule ligand, UNC6535, which occupies simultaneously both the TD2 and TD3 reader binding sites. Further optimization leads to the discovery of UNC10013, a covalent 3TD ligand targeting Cys385 of SETDB1. UNC10013 is potent with a kinact/KI of 1.0 × 106 M-1s-1 and demonstrates proteome-wide selectivity. In cells, negative allosteric modulation of SETDB1-mediated Akt methylation occurs after treatment with UNC10013. Therefore, UNC10013 is a potent, selective, and cell-active covalent ligand for the 3TD of SETDB1, demonstrating negative allosteric modulator properties and making it a promising tool to study the biological role of SETDB1 in disease progression.
Collapse
Affiliation(s)
- Mélanie Uguen
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Devan J Shell
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Yu Deng
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Ka Yang
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Yani Zhao
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Michael A Stashko
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jacqueline L Norris-Drouin
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jarod M Waybright
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Design Therapeutics, Carlsbad, CA, USA
| | | | - Justin M Rectenwald
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Angie L Mordant
- UNC Metabolomics and Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Thomas S Webb
- UNC Metabolomics and Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Laura E Herring
- UNC Metabolomics and Proteomics Core Facility, Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Robert K McGinty
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Pengda Liu
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Lindsey I James
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Kenneth H Pearce
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Stephen V Frye
- UNC Eshelman School of Pharmacy, Center for Integrative Chemical Biology and Drug Discovery, Chemical Biology and Medicinal Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA.
| |
Collapse
|
865
|
Xie Y, Liu F, Wu Y, Zhu Y, Jiang Y, Wu Q, Dong Z, Liu K. Inflammation in cancer: therapeutic opportunities from new insights. Mol Cancer 2025; 24:51. [PMID: 39994787 PMCID: PMC11849313 DOI: 10.1186/s12943-025-02243-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/20/2025] [Indexed: 02/26/2025] Open
Abstract
As one part of the innate immune response to external stimuli, chronic inflammation increases the risk of various cancers, and tumor-promoting inflammation is considered one of the enabling characteristics of cancer development. Recently, there has been growing evidence on the role of anti-inflammation therapy in cancer prevention and treatment. And researchers have already achieved several noteworthy outcomes. In the review, we explored the underlying mechanisms by which inflammation affects the occurrence and development of cancer. The pro- or anti-tumor effects of these inflammatory factors such as interleukin, interferon, chemokine, inflammasome, and extracellular matrix are discussed. Since FDA-approved anti-inflammation drugs like aspirin show obvious anti-tumor effects, these drugs have unique advantages due to their relatively fewer side effects with long-term use compared to chemotherapy drugs. The characteristics make them promising candidates for cancer chemoprevention. Overall, this review discusses the role of these inflammatory molecules in carcinogenesis of cancer and new inflammation molecules-directed therapeutic opportunities, ranging from cytokine inhibitors/agonists, inflammasome inhibitors, some inhibitors that have already been or are expected to be applied in clinical practice, as well as recent discoveries of the anti-tumor effect of non-steroidal anti-inflammatory drugs and steroidal anti-inflammatory drugs. The advantages and disadvantages of their application in cancer chemoprevention are also discussed.
Collapse
Affiliation(s)
- Yifei Xie
- Department of Pathology and Forensic Medicine, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Fangfang Liu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Medical Genetics and Cell Biology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Yunfei Wu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yuer Zhu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
| | - Yanan Jiang
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Qiong Wu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China
| | - Zigang Dong
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China.
| | - Kangdong Liu
- State Key Laboratory of Metabolic Dysregulation & the Prevention and Treatment of Esophageal Cancer, Zhengzhou, Henan, 450052, China.
- Department of Pathophysiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450000, China.
- China-US (Henan) Hormel Cancer Institute, Zhengzhou, Henan, 450007, China.
- The Collaborative Innovation Center of Henan Province for Cancer Chemoprevention, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
866
|
Currie GM, Rohren EM. Potential of Technetium and Rhenium Theranostics. Semin Nucl Med 2025:S0001-2998(25)00006-6. [PMID: 40000268 DOI: 10.1053/j.semnuclmed.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 01/23/2025] [Accepted: 01/24/2025] [Indexed: 02/27/2025]
Abstract
While theranostics has transformed the precision medicine landscape over the last decade, there is scope for the development of true theranostic pairs, e.g. diagnostic and therapeutic partners in which any physical, chemical, and biological differences are negligible to in vivo application. Although simple to state in theory, there are, in fact, limited options exhibiting optimal physical characteristics and wholly shared elements. Further compounding real-world application of the traditional theranostic method are additional barriers. The use of PET/CT as the cornerstone of the diagnostic pair in theranostics creates inequity of access and opportunity based on socioeconomic and geographic factors, and the growing demand for both 68Ga and 177Lu is straining production capabilities globally. Improving access to theranostics globally will require novel thinking and infrastructure investment to ensure that patients of all economic and social backgrounds have access to this transformative technology. An approach which is underdeveloped, but which may address gaps in health inequities and improve outcomes, is the application of the widely available generator-produced 99mTc for imaging and 188Re for therapy. Despite favourable and near identical radiochemistry, the search for the next generation of theranostic radionuclide pairs seldom references technetium or rhenium radionuclides. Advances in SPECT/CT instrumentation and radiochemistry provide an opportunity to deliver theranostics to communities not serviced by PET-based theranostics. The 188Re and 99mTc supply by daily elution of a generator affords significant convenience, flexibility and delayed biomolecule imaging. Low abundance gamma emissions of 188Re allow serial imaging and dosimetry calculations. 99mTc / 188Re theranostics could address inequity in access and opportunity to cutting edge theranostics.
Collapse
Affiliation(s)
- Geoffrey M Currie
- School of Dentistry and Medical Sciences, Charles Sturt University, NSW, Australia; Department of Radiology, Baylor College of Medicine, TX, USA.
| | - Eric M Rohren
- School of Dentistry and Medical Sciences, Charles Sturt University, NSW, Australia; Department of Radiology, Baylor College of Medicine, TX, USA
| |
Collapse
|
867
|
Choudhary D, Kumar B, Chandrasekaran B, Singh TG, Kaur R, Aldahish A, Vasudevan R, Balaji P. Microwave-Assisted Synthesis of Morpholine-Based Chalcones as Reversible MAO-A Inhibitors in the Management of Mental Depression. Pharmaceuticals (Basel) 2025; 18:309. [PMID: 40143088 PMCID: PMC11945657 DOI: 10.3390/ph18030309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/13/2025] [Accepted: 02/20/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Depression is one of the most serious and common health problems among the youth population and is responsible for the initiation of many diseases. As per the World Health Organization, 3.8% of the population suffers from mental depression, globally. The monoamine oxidase-A (MAO-A) enzyme is responsible for the degradation of neurotransmitters leading to lower levels of neurotransmitters. Methods: Chalcones (C1-C15) were synthesized by reacting substituted acetophenone with various benzaldehydes in a basic ethanolic solvent at 80 °C under microwave irradiation conditions. To compare the reaction time and product yield, a conventional method of synthesis of chalcones was also performed. The synthesized chalcones (C1-C15) were spectroscopically characterized and screened initially for inhibitory activities against MAO-A and MAO-B. The best active compounds were undertaken for IC50 determination against MAO-A enzyme followed by the reversibility of inhibition analysis and the antioxidant assay. Moreover, in silico molecular docking and ADME pharmacokinetic investigations were accomplished. Results: Most of the compounds inhibited MAO-A, specifically, compounds C14 and C6 exhibited the highest inhibition at IC50 values of 7.91 ± 0.08 μM and 8.45 ± 0.19 μM, respectively. Both these compounds exhibited a reversible MAO-A inhibition displaying up to 60% recovery of enzymatic activity when diluted with substrate (Tyramine). The results of the in silico study indicated docking scores of -9.56 Kcal/mol (C14) and -9.45 Kcal/mol (C6) and exhibited a π-π stacking interaction with the crucial amino acid Trp-397. The compounds were determined to cross the blood-brain barrier (BBB) and displayed favorable gastrointestinal (GI) absorption. Further, the antioxidant assay results demonstrated that the synthesized compounds possess modest free radical scavenging potential. Conclusions: This study displayed the MAO-A inhibitory potential of morpholine-substituted chalcones as a promising pharmacophore for the development of novel antidepressant lead compounds.
Collapse
Affiliation(s)
- Diksha Choudhary
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Bhupinder Kumar
- Department of Pharmaceutical Sciences, HNB Garhwal University, Chauras Campus, Srinagar 246174, Jammu and Kashmir, India
| | | | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Rajwinder Kaur
- Chitkara College of Pharmacy, Chitkara University, Rajpura 140401, Punjab, India
| | - Afaf Aldahish
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia (R.V.)
| | - Rajalakshimi Vasudevan
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia (R.V.)
| | - Prasanalakshmi Balaji
- Department of Computer Science, College of Computer Science, King Khalid University, Abha 61421, Saudi Arabia
| |
Collapse
|
868
|
Paul A, Terrell JR, Farahat AA, Ogbonna EN, Kumar A, Boykin DW, Neidle S, Wilson WD. Alternative Approach to Sequence-Specific Recognition of DNA: Cooperative Stacking of Dication Dimers─Sensitivity to Compound Curvature, Aromatic Structure, and DNA Sequence. ACS Chem Biol 2025; 20:489-506. [PMID: 39920086 PMCID: PMC11851451 DOI: 10.1021/acschembio.4c00800] [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: 11/23/2024] [Revised: 01/24/2025] [Accepted: 01/28/2025] [Indexed: 02/09/2025]
Abstract
With the growing number and diversity of known genome sequences, there is an increasing opportunity to regulate gene expression through synthetic, cell-permeable small molecules. Enhancing the DNA sequence recognition abilities of minor groove compounds has the potential to broaden their therapeutic applications with significant implications for areas such as modulating transcription factor activity. While various classes of minor groove binding agents can selectively identify pure AT and mixed AT and GC base pair(s) containing sequences, there remains a lack of compounds capable of distinguishing between different AT sequences. In this work, we report on the design compounds that exhibit selective binding to -TTAA- or -TATA- containing DNA minor groove sequences compared with other AT ones. Several studies have shown that the -AATT- and -TTAA- sequences have distinct physical and interaction properties, especially in terms of their different requirements for recognition in the minor groove. Achieving strong, selective minor groove binding at -TTAA- sequences has been challenging, but DB1003, a benzimidazole-furan-furan diamidine, has demonstrated cooperative dimeric binding activity at -TTAA-. It has significantly less binding preference for AATT. To better understand and modify the selectivity, we synthesized a set of rationally designed analogs of DB1003 by altering the position of the five-membered heterocyclic structure. Binding affinities and stoichiometries obtained from biosensor-surface plasmon resonance experiments show that DB1992, a benzimidazolefuran-thiophene diamidine, binds strongly to -TTAA- as a positive cooperative dimer with high cooperativity. The high-resolution crystal structure of the TTAA-DNA-DB1992 complex reveals that DB1992 binds as an antiparallel π-stacked dimer with numerous diverse contacts to the DNA minor groove. This distinctive binding arrangement and the properties of diamidines at the -TTAA- minor groove demonstrate that benzimidazole-furan-thiophene is a unique DNA binding pharmacophore. Competition mass spectroscopy and circular dichroism studies confirmed the binding stoichiometry and selectivity preference of the compounds for the -TTAA- sequence.
Collapse
Affiliation(s)
- Ananya Paul
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - J. Ross Terrell
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - Abdelbasset A. Farahat
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
- Department
of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
- Master
of Pharmaceutical Sciences Program, California
North State University, Elk Grove, California 95757, United States
| | - Edwin N. Ogbonna
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - Arvind Kumar
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - David W. Boykin
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| | - Stephen Neidle
- School
of Pharmacy, University College London, London WC1N 1AX, U.K.
| | - W. David Wilson
- Department
of Chemistry and Center for Diagnostics and Therapeutics Georgia State
University, Atlanta, Georgia 30303, United States
| |
Collapse
|
869
|
Nemoto M, Ando W, Mano T, Lee M, Yuzawa S, Mizuno T. Identification of Key Amino Acids in the A Domains of Polymyxin Synthetase Responsible for 2,4-Diaminobutyric Acid Adenylation in Paenibacillus polymyxa NBRC3020 Strain. ACS Chem Biol 2025; 20:321-331. [PMID: 39818748 DOI: 10.1021/acschembio.4c00553] [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: 01/19/2025]
Abstract
Developing novel nonribosomal peptides (NRPs) requires a comprehensive understanding of the enzymes involved in their biosynthesis, particularly the substrate amino acid recognition mechanisms in the adenylation (A) domain. This study focused on the A domain responsible for adenylating l-2,4-diaminobutyric acid (l-Dab) within the synthetase of polymyxin, an NRP produced by Paenibacillus polymyxa NBRC3020. To date, investigations into recombinant proteins that selectively adenylate l-Dab─exploring substrate specificity and enzymatic activity parameters─have been limited to reports on A domains found in enzymes synthesizing l-Dab homopolymers (pldA from S. celluloflavus USE31 and pddA from S. hindustanus NBRC15115), which remain exceedingly rare. The polymyxin synthetase in NBRC3020 contains five A domains specific to l-Dab, distributed across five distinct modules (modules 1, 3, 4, 5, 8, and 9). In this study, we successfully obtained soluble A domain proteins from modules 1, 5, 8, and 9 by preparing module-specific recombinant proteins. These proteins were expressed in E. coli BAP-1, purified via Ni-affinity chromatography, and demonstrated high specificity for l-Dab. Through sequence homology analysis, three-dimensional structural modeling, docking simulations to estimate substrate-binding sites, and functional validation using alanine mutants, we identified Glu281 and Asp344 as critical residues for recognizing the side chain amino group of l-Dab, and Asp238 as essential for recognizing its main chain amino group in the A domain. Notably, these key residues were conserved not only across the A domains in modules 1, 5, 8, and 9 of P. polymyxa NBRC3020 but also in those of the P. polymyxa PKB1 strain, as confirmed by sequence homology analysis. Interestingly, in pldA and pddA, the key residues involved in recognizing the side-chain amino group of l-Dab, which are conserved among polymyxin synthetases of NBRC3020 and PKB1 strain, were not observed. This suggests a potentially different mechanism for l-Dab recognition.
Collapse
Affiliation(s)
- Mai Nemoto
- Department of Life Science and Applied Chemistry, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya, Aichi 466-8555, Japan
| | - Wataru Ando
- Department of Life Science and Applied Chemistry, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya, Aichi 466-8555, Japan
| | - Taichi Mano
- Department of Life Science and Applied Chemistry, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya, Aichi 466-8555, Japan
| | - Minjae Lee
- Faculty of Environment and Information Studies, Keio University, Fujisawa, Kanagawa 252-0882, Japan
- Department of Chemistry, University of California, Berkeley, Berkeley, California 94720, United States
| | - Satoshi Yuzawa
- Systems Biology Program, Graduate School of Media and Governance, Keio University, Fujisawa, Kanagawa 252-0882, Japan
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Toshihisa Mizuno
- Department of Life Science and Applied Chemistry, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho, Showa-ku, Nagoya, Aichi 466-8555, Japan
- Department of Nanopharmaceutical Sciences, Graduate School of Engineering, Nagoya Institute of Technology, Gokiso-cho Showa-ku, Nagoya, Aichi 466-8555, Japan
| |
Collapse
|
870
|
Noori Z, Solà M, Viñas C, Teixidor F, Poater J. Unraveling aromaticity: the dual worlds of pyrazole, pyrazoline, and 3D carborane. Beilstein J Org Chem 2025; 21:412-420. [PMID: 39996167 PMCID: PMC11849550 DOI: 10.3762/bjoc.21.29] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/15/2025] [Indexed: 02/26/2025] Open
Abstract
A new series of o-carborane-fused pyrazoles has been recently successfully synthesized. This fusion was expected to create a hybrid 3D/2D aromatic system, combining the 3D aromaticity of o-carborane with the 2D aromaticity of pyrazole. However, while the boron cage retains its aromatic character, the pyrazole's aromaticity is lost. As a result, rather than forming o-carborane-fused pyrazoles, the synthesis yielded o-carborane-fused pyrazolines, which are non-aromatic. The limited overlap between the π molecular orbitals (MOs) of the planar heterocycle and the n + 1 MOs of the carborane prevents significant electronic delocalization between the two fused components. This contrasts with the fusion of pyrazole and benzene to form indazole, where both rings maintain their 2D aromaticity. Our findings demonstrate that the peripheral σ-aromaticity of carborane and the π-aromaticity of the heterocycle are orthogonal, making a true 3D/2D aromatic system unachievable. The carborane is highly aromatic, generating highly negative NICS values (-25 to -30 ppm). We have observed that these high NICS values extend to fused rings, leading to incorrect estimations of aromaticity. Therefore, relying solely on NICS can be misleading, and other computational indicators, along with experimental or structural data, should be used to accurately assess aromaticity.
Collapse
Affiliation(s)
- Zahra Noori
- Departament de Química Inorgànica i Orgànica & Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain
| | - Miquel Solà
- Institut de Química Computacional i Catàlisi and Departament de Química, Universitat de Girona, Maria Aurèlia Capmany 69, 17003 Girona, Catalonia, Spain
| | - Clara Viñas
- Institut de Ciència de Materials de Barcelona, Consejo Superior de Investigaciones Científicas, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Francesc Teixidor
- Institut de Ciència de Materials de Barcelona, Consejo Superior de Investigaciones Científicas, Campus Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Jordi Poater
- Departament de Química Inorgànica i Orgànica & Institut de Química Teòrica i Computacional (IQTCUB), Universitat de Barcelona, Martí i Franquès 1-11, 08028 Barcelona, Spain
- ICREA, Pg. Lluís Companys 23, 08010 Barcelona, Spain
| |
Collapse
|
871
|
Huang L, Zhang Y, Liu P, Lan L, Yang L, Wang B, Cao T, Hu L, Qin X. Design, synthesis and biological evaluation of 2 H-[1,4]oxazino-[2,3- f]quinazolin derivatives as potential EGFR inhibitors for non-small cell lung cancer. RSC Med Chem 2025:d4md01016g. [PMID: 40093516 PMCID: PMC11907643 DOI: 10.1039/d4md01016g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/17/2025] [Indexed: 03/19/2025] Open
Abstract
Epidermal growth factor receptor tyrosine kinase inhibitors (EGFR-TKIs) have emerged as the first-line treatment for patients with EGFR-mutant non-small cell lung cancer (NSCLC). A series of 2H-[1,4]oxazino[2,3-f]quinazolin derivatives were synthesized and evaluated as irreversible EGFR-TKIs for the treatment of NSCLC. Most of the synthesized compounds demonstrated strong inhibitory activity against the EGFR kinase and the tested cancer cells. Notably, compound 4a exhibited considerable inhibitory effects against the EGFR kinase and the EGFRL858R/T790M mutant NCI-H1975 cancer cells. Compound 4a was found to suppress cell proliferation, colony formation, cell invasion, and migration, while also inducing G0/G1 phase arrest of the cell cycle in NCI-H1975 cells. Compound 4a was docked into the active pocket of the EGFR mutant to ascertain the probable binding conformation. Overall, compound 4a was identified as a promising irreversible EGFR-TKI for the treatment of NSCLC.
Collapse
Affiliation(s)
- Linchang Huang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University Nanning 530008 China
| | - Ying Zhang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University Nanning 530008 China
| | - Peng Liu
- Guangzhou Institute of Biomedicine and Health, Chinese Academy of Science Guangzhou 510530 China
| | - Lihong Lan
- School of Chemistry and Chemical Engineering, Guangxi Minzu University Nanning Guangxi 530008 China
| | - Lifang Yang
- School of Chemistry and Chemical Engineering, Guangxi Minzu University Nanning Guangxi 530008 China
| | - Bo Wang
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University Nanning 530008 China
| | - Tingting Cao
- College of Chemistry and Life Science, Beijing University of Technology Beijing 100124 China
| | - Liming Hu
- College of Chemistry and Life Science, Beijing University of Technology Beijing 100124 China
| | - Xuemei Qin
- Guangxi Key Laboratory for Polysaccharide Materials and Modifications, School of Marine Sciences and Biotechnology, Guangxi Minzu University Nanning 530008 China
| |
Collapse
|
872
|
Yang DD, Chutiwitoonchai N, Wang F, Tian P, Sureram S, Lei X, Mahidol C, Ruchirawat S, Kittakoop P. Effects of organic salts of virucidal and antiviral compounds from Nelumbo nucifera and Kaempferia parviflora against SARS-CoV-2. Sci Rep 2025; 15:6380. [PMID: 39984611 PMCID: PMC11845762 DOI: 10.1038/s41598-025-89736-0] [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: 12/11/2024] [Accepted: 02/07/2025] [Indexed: 02/23/2025] Open
Abstract
The present work investigates virucidal and antiviral compounds in the extracts of seed embryos of a lotus, Nelumbo nucifera, and a Thai ginseng, Kaempferia parviflora. Separation of the extracts led to the identification of antiviral compounds against SARS-CoV-2. Neferine (1) and nuciferine (3) from N. nucifera, as well as their respective HCl salts (2 and 4), exhibited virucidal and antiviral activities against SARS-CoV-2. Virucidal activity of neferine salt (2) (EC50 4.78 µM) was 7.5 times better than its free-base, neferine (1) (EC50 36.01 µM), and the salt (2) also improved the selectivity index (SI), showing less cytotoxicity than 1. This work demonstrates that organic salts have an impact on biological activities. A crude extract of K. parviflora rhizomes displayed virucidal activity (EC50 42.11 µg/mL) and antiviral activity (EC50 39.28 µg/mL). Isolation of a crude extract of K. parviflora rhizomes led to the identification of nine flavonoids (5-13). Among these flavonoids, only 5,7,4'-trimethoxyflavone (8) was found to show virucidal (EC50 437.90 µM) and antiviral (EC50 50.97 µM) activities against SARS-CoV-2. However, flavonoids (5-13) did not inhibit SARS-CoV-2 3CLpro enzyme at the concentrations of 10 µM and 100 µM. In conclusion, our data underscores the therapeutic potential of N. nucifera and K. parviflora derived bioactive compounds against SARS-CoV-2.
Collapse
Affiliation(s)
- Dan-Dan Yang
- Chulabhorn Graduate Institute, Program in Chemical Sciences, Laksi, Bangkok, 10210, Thailand
| | - Nopporn Chutiwitoonchai
- Virology and Cell Technology Research Team, National Center for Genetic Engineering and Biotechnology (BIOTEC), National Science and Technology Development Agency (NSTDA), 113 Thailand Science Park, Phahonyothin Rd, Pathumthani, 12120, Thailand.
| | - Feng Wang
- The Research Center of Chiral Drugs, China-Thailand Joint Research Institute of Natural Medicine, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, 201203, Shanghai, China
| | - Ping Tian
- The Research Center of Chiral Drugs, China-Thailand Joint Research Institute of Natural Medicine, Innovation Research Institute of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, 201203, Shanghai, China
| | - Sanya Sureram
- Chulabhorn Research Institute, Kamphaeng Phet 6, Talat Bang Khen, Lak Si, 10210, Bangkok, Thailand
| | - Xiaoguang Lei
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Bioorganic Chemistry and Molecular Engineering of Ministry of Education, College of Chemistry and Molecular Engineering, Peking University, 100871, Beijing, China
| | - Chulabhorn Mahidol
- Chulabhorn Research Institute, Kamphaeng Phet 6, Talat Bang Khen, Lak Si, 10210, Bangkok, Thailand
| | - Somsak Ruchirawat
- Chulabhorn Graduate Institute, Program in Chemical Sciences, Laksi, Bangkok, 10210, Thailand
- Chulabhorn Research Institute, Kamphaeng Phet 6, Talat Bang Khen, Lak Si, 10210, Bangkok, Thailand
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, Science, Research and Innovation, Ministry of Higher Education, 10400, Bangkok, Thailand
| | - Prasat Kittakoop
- Chulabhorn Graduate Institute, Program in Chemical Sciences, Laksi, Bangkok, 10210, Thailand.
- Chulabhorn Research Institute, Kamphaeng Phet 6, Talat Bang Khen, Lak Si, 10210, Bangkok, Thailand.
- Center of Excellence on Environmental Health and Toxicology (EHT), OPS, Science, Research and Innovation, Ministry of Higher Education, 10400, Bangkok, Thailand.
| |
Collapse
|
873
|
Kasula M, Toyama M, Samunuri R, Jha AK, Okamoto M, Baba M, Sharon A. Pyrazolo[3,4- d]pyrimidine-based neplanocin analogues identified as potential de novo pharmacophores for dual-target HBV inhibition. RSC Med Chem 2025:d4md00932k. [PMID: 39990166 PMCID: PMC11840712 DOI: 10.1039/d4md00932k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Accepted: 01/16/2025] [Indexed: 02/25/2025] Open
Abstract
The discovery of selective and potent inhibitors through de novo pathways is essential to combat drug resistance in chronic hepatitis B (CHB) infections. Recent studies have highlighted that neplanocin A (NepA) derivatives are biologically selective inhibitors of the hepatitis B virus (HBV). In this study, we designed, synthesized, and evaluated various pyrazolo[3,4-d]pyrimidine-based NepA analogues (4a-h) for their anti-HBV activity. Notably, analogue 4g demonstrated significant activity against HBV replication, with EC50 (HBV DNA) = 0.96 μM, CC50 > 100 μM and EC50 (HBsAg) = 0.82 μM, showing selective inhibition of HBsAg secretion. The SAR analysis concluded that replacing the polar 4-NH2 group with -CH3 also acted as a weak H-bonding donor, and the presence of 3-iodo was found to be desirable for the activity/toxicity profile. The nucleoside analogues exhibited a distinct mechanism of action compared to existing nucleoside analogues for the selective inhibition of HBsAg secretion. Based on these findings, compound 4g represents a promising lead molecule for the development of new anti-HBV agents with unique mechanisms of action.
Collapse
Affiliation(s)
- Mohan Kasula
- Department of Chemistry, Forbesganj College, Purnea Univeristy-854318 India
- Department of Chemistry, Birla Institute of Technology Mesra Ranchi-835215 India
| | - Masaaki Toyama
- Department of Virology II, National Institute of Infectious Diseases Tokyo Japan
| | - Ramakrishnamraju Samunuri
- Department of Chemistry, Birla Institute of Technology Mesra Ranchi-835215 India
- Aragen Life Sciences Ltd IDA, Nacharam Hyderabad-500076 India
| | - Ashok Kumar Jha
- Aragen Life Sciences Ltd IDA, Nacharam Hyderabad-500076 India
| | - Mika Okamoto
- Division of Infection Control Research, Center for Advanced Research and Promotion, Kagoshima University Kagoshima Japan
| | - Masanori Baba
- Division of Infection Control Research, Center for Advanced Research and Promotion, Kagoshima University Kagoshima Japan
| | - Ashoke Sharon
- Department of Chemistry, Birla Institute of Technology Mesra Ranchi-835215 India
| |
Collapse
|
874
|
Bailey HJ, Eisert J, Kazi R, Gerhartz J, Pieńkowska DE, Dressel I, Vollrath J, Kondratov I, Matviyuk T, Tolmachova N, Shah VJ, Giuliani G, Mosler T, Geiger TM, Esteves AM, Santos SP, Sousa RL, Bandeiras TM, Leibrock EM, Bauer U, Leuthner B, Langer JD, Wegener AA, Nowak RP, Sorrell FJ, Dikic I. An engineered cereblon optimized for high-throughput screening and molecular glue discovery. Cell Chem Biol 2025; 32:363-376.e10. [PMID: 39610248 DOI: 10.1016/j.chembiol.2024.11.002] [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: 05/15/2024] [Revised: 09/24/2024] [Accepted: 11/01/2024] [Indexed: 11/30/2024]
Abstract
The majority of clinical degraders utilize an immunomodulatory imide drug (IMiD)-based derivative that directs their target to the E3 ligase receptor cereblon (CRBN); however, identification of IMiD molecular glue substrates has remained underexplored. To tackle this, we design human CRBN constructs, which retain all features for ternary complex formation, while allowing generation of homogenous and cost-efficient expression in E. coli. Extensive profiling of the construct shows it to be the "best of both worlds" in terms of binding activity and ease of production. We next designed the "Enamine focused IMiD library" and demonstrated applicability of the construct to high-throughput screening, identifying binders with high potency, ligand efficiency, and specificity. Finally, we adapt our construct for proof of principle glue screening approaches enabling IMiD cellular interactome determination. Coupled with our IMiD binding landscape the methods described here should serve as valuable tools to assist discovery of next generation CRBN glues.
Collapse
Affiliation(s)
- Henry J Bailey
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60439 Frankfurt am Main, Germany
| | - Jonathan Eisert
- Merck Healthcare KGaA, Discovery and Development Technologies, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Rubina Kazi
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Jan Gerhartz
- Institute of Structural Biology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Dominika Ewa Pieńkowska
- Institute of Structural Biology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Ina Dressel
- Institute of Structural Biology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Joshua Vollrath
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, IMPRS on Cellular Biophysics, Max-von-Laue-Strasse 3, 60439 Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60439 Frankfurt am Main, Germany
| | - Ivan Kondratov
- Enamine Ltd., Winston Churchill Street 78, 02094 Kyiv, Ukraine; Enamine Germany GmbH, Industriepark Hoechst, G837, 65926 Frankfurt am Main, Germany; V. P. Kukhar Institute of Bioorganic Chemistry and Petrochemistry, National Academy of Sciences of Ukraine, Akademik Kukhar Street 1, 02094 Kyiv, Ukraine
| | | | | | - Varun Jayeshkumar Shah
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Giulio Giuliani
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Thorsten Mosler
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany
| | - Thomas M Geiger
- Institute of Structural Biology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Ana M Esteves
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Sandra P Santos
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Raquel L Sousa
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Tiago M Bandeiras
- iBET, Instituto de Biologia Experimental e Tecnológica, Apartado 12, 2781-901 Oeiras, Portugal
| | - Eva-Maria Leibrock
- Merck Healthcare KGaA, Discovery and Development Technologies, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Ulrike Bauer
- Merck Healthcare KGaA, Discovery and Development Technologies, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Birgitta Leuthner
- Merck Healthcare KGaA, Discovery and Development Technologies, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Julian D Langer
- Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60439 Frankfurt am Main, Germany; Max Planck Institute for Brain Research, Max-von-Laue-Strasse 4, 60439 Frankfurt am Main, Germany
| | - Ansgar A Wegener
- Merck Healthcare KGaA, Discovery and Development Technologies, Frankfurter Straße 250, 64293 Darmstadt, Germany
| | - Radosław P Nowak
- Institute of Structural Biology, University of Bonn, Venusberg Campus 1, 53127 Bonn, Germany
| | - Fiona J Sorrell
- Merck Healthcare KGaA, Discovery and Development Technologies, Frankfurter Straße 250, 64293 Darmstadt, Germany.
| | - Ivan Dikic
- Institute of Biochemistry II, Medical Faculty, Goethe-University, Frankfurt am Main and Buchmann Institute for Molecular Life Sciences, Frankfurt am Main, Germany; Max Planck Institute of Biophysics, Max-von-Laue-Strasse 3, 60439 Frankfurt am Main, Germany.
| |
Collapse
|
875
|
Krejcova K, Boura E. Structural basis for broad-spectrum binding of AT-9010 to flaviviral methyltransferases. Arch Virol 2025; 170:61. [PMID: 39976734 PMCID: PMC11842469 DOI: 10.1007/s00705-025-06227-3] [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: 08/27/2024] [Accepted: 12/06/2024] [Indexed: 02/23/2025]
Abstract
AT-9010 (2'-methyl-2'-fluoro guanosine triphosphate) is a GTP analog whose prodrug, AT-752 is under consideration in human medicine as a potential antiviral drug against certain flaviviruses. It was previously believed to inhibit viral replication by acting primarily as a chain terminator. However, it was discovered recently that it also binds the GTP binding site of the methyltransferase (MTase) domain of the orthoflavivirus polymerase, thus interfering with RNA capping. Here, we investigated the binding of AT-9010 to Ntaya and Zika virus MTases. Structural analysis using X-ray crystallography revealed similar interactions between the base and sugar moieties of AT-9010 and key residues in both MTases, although differences in hydrogen bonding were observed. Our analysis also suggested that the triphosphate part of AT-9010 is flexible. Despite minor variations, the overall binding mode of AT-9010 was found to be the same for all of the flaviviral MTases examined, suggesting a structural basis for the efficacy of AT-9010 against multiple orthoflavivirus MTases.
Collapse
Affiliation(s)
- Katerina Krejcova
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i, Flemingovo nám. 2, Prague, 16610, Czech Republic
| | - Evzen Boura
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, v.v.i, Flemingovo nám. 2, Prague, 16610, Czech Republic.
| |
Collapse
|
876
|
Sokal A, Mruczek P, Niedoba M, Dewalska A, Stocerz K, Kadela-Tomanek M. Anticancer Activity of Ether Derivatives of Chrysin. Molecules 2025; 30:960. [PMID: 40005270 PMCID: PMC11857933 DOI: 10.3390/molecules30040960] [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: 12/30/2024] [Revised: 02/15/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Chrysin, a naturally occurring flavonoid, exhibits a broad spectrum of biological activities, including showing anticancer properties. However, its clinical application is limited by poor bioavailability and low solubility. The introduction of an amine, amide, ester, or alkoxy group to a flavone skeleton influences the biological activity. This review also discusses hybrid compounds, such as the chrysin-porphyrin hybrid, which are characterized by higher biological activity and better bioavailability properties than single molecules. This review concentrates on the anticancer activity of chrysin and its derivatives against the most popular cancers, such as breast, lung, prostate, and gastrointestinal tumors.
Collapse
Affiliation(s)
- Arkadiusz Sokal
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland; (A.S.); (P.M.)
- Doctoral School, Medical University of Silesia in Katowice, 15 Poniatowskiego Str., 40-055 Katowice, Poland; (M.N.); (A.D.); (K.S.)
| | - Patryk Mruczek
- Department of Organic Chemistry, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 4 Jagiellońska Str., 41-200 Sosnowiec, Poland; (A.S.); (P.M.)
| | - Mateusz Niedoba
- Doctoral School, Medical University of Silesia in Katowice, 15 Poniatowskiego Str., 40-055 Katowice, Poland; (M.N.); (A.D.); (K.S.)
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 30 Ostrogórska Str., 41-200 Sosnowiec, Poland
| | - Agnieszka Dewalska
- Doctoral School, Medical University of Silesia in Katowice, 15 Poniatowskiego Str., 40-055 Katowice, Poland; (M.N.); (A.D.); (K.S.)
- Department of Microbiology and Immunology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana Str., 41-800 Zabrze, Poland
| | - Klaudia Stocerz
- Doctoral School, Medical University of Silesia in Katowice, 15 Poniatowskiego Str., 40-055 Katowice, Poland; (M.N.); (A.D.); (K.S.)
- Department of Community Pharmacy, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia in Katowice, 10 Jedności Str., 41-200 Sosnowiec, Poland
| | - Monika Kadela-Tomanek
- Department of Pathology, Faculty of Pharmaceutical Sciences in Sosnowiec, Medical University of Silesia, 30 Ostrogórska Str., 41-200 Sosnowiec, Poland
| |
Collapse
|
877
|
Kohl F, Vogl T, Hampel F, Dube H. Hemiphosphoindigos as a platform for chiroptical or water soluble photoswitching. Nat Commun 2025; 16:1760. [PMID: 39971955 PMCID: PMC11840110 DOI: 10.1038/s41467-025-56942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 02/06/2025] [Indexed: 02/21/2025] Open
Abstract
Photoswitches are important molecular tools to precisely control the behavior of matter by using light irradiation. They have found application in virtually all applied chemical fields from chemical biology to material sciences. However, great challenges remain in advanced property design including tailored chiroptical responses or water solubility. Here, hemiphosphoindigo (HPI) photoswitches are presented as capable phosphorus-based photoswitches and a distinct addition to the established indigoid chromophore family. Phosphinate is embedded in the core indigoid chromophore and the resulting optimized photoswitches display high thermal stabilities, excellent fatigue resistance and high isomer enrichment. A series of planar, twisted and heterocyclic HPIs are investigated to probe design strategies for advantageous photophysical properties. The phosphinate provides a platform for easily accessible, water-soluble photoswitches, especially interesting for biological applications. Its chiral nature further allows light-induced modulation of chiroptical properties. HPIs therefore open up a distinct structural space for photoswitch generation and advanced light-responsive applications.
Collapse
Affiliation(s)
- Fabien Kohl
- Friedrich-Alexander Universität Erlangen-Nurnberg, Department of Chemistry and Pharmacy, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Theresa Vogl
- Friedrich-Alexander Universität Erlangen-Nurnberg, Department of Chemistry and Pharmacy, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Frank Hampel
- Friedrich-Alexander Universität Erlangen-Nurnberg, Department of Chemistry and Pharmacy, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany
| | - Henry Dube
- Friedrich-Alexander Universität Erlangen-Nurnberg, Department of Chemistry and Pharmacy, Nikolaus-Fiebiger-Str. 10, 91058, Erlangen, Germany.
| |
Collapse
|
878
|
Li AY, Xie R, Zhou Q, Huang PF, Liu Y. Photoinduced radical cyclization reaction of isocyanides with α-carbonyl bromides to access 11-alkyl-substituted 1,4-dibenzodiazepines. Org Biomol Chem 2025; 23:1874-1882. [PMID: 39807070 DOI: 10.1039/d4ob01985g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
1,4-Dibenzodiazepines, an important component of nitrogen-containing heterocycles, are widely present in drugs. Herein, we developed a photochemical radical cascade cyclization reaction of isocyanides with α-carbonyl bromides under mild conditions. A sequence of 11-alkyl-substituted 1,4-dibenzodiazepines were produced in 53%-85% yields, demonstrating excellent tolerance towards various functional groups. Primary, secondary, and tertiary α-carbonyl bromides were effectively employed as alkyl reagents.
Collapse
Affiliation(s)
- Ao-Yun Li
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China.
| | - Rong Xie
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China.
| | - Quan Zhou
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China.
| | - Peng-Fei Huang
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China.
| | - Yu Liu
- Department of Chemistry and Chemical Engineering, Hunan Institute of Science and Technology, Yueyang 414006, China.
| |
Collapse
|
879
|
Sobhani N, Pittacolo M, D’Angelo A, Marchegiani G. Recent Anti-KRAS G12D Therapies: A "Possible Impossibility" for Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2025; 17:704. [PMID: 40002297 PMCID: PMC11853620 DOI: 10.3390/cancers17040704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/13/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) is an aggressive cancer, able to thrive in a challenging tumor microenvironment. Current standard therapies, including surgery, radiation, chemotherapy, and chemoradiation, have shown a dismal survival prognosis, resulting in less than a year of life in the metastatic setting. Methods: The pressing need to find better therapeutic methods brought about the discovery of new targeted therapies against the infamous KRAS mutations, the major oncological drivers of PDAC. Results: The most common KRAS mutation is KRASG12D, which causes a conformational change in the protein that constitutively activates downstream signaling pathways driving cancer hallmarks. Novel anti-KRASG12D therapies have been developed for solid-organ tumors, including small compounds, pan-RAS inhibitors, protease inhibitors, chimeric T cell receptors, and therapeutic vaccines. Conclusions: This comprehensive review summarizes current knowledge on the biology of KRAS-driven PDAC, the latest therapeutic options that have been experimentally validated, and developments in ongoing clinical trials.
Collapse
Affiliation(s)
- Navid Sobhani
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Matteo Pittacolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy;
| | - Alberto D’Angelo
- Department of Medicine, Northern General Hospital, Sheffield S5 7AT, UK;
| | - Giovanni Marchegiani
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35122 Padova, Italy;
| |
Collapse
|
880
|
Girgis AS, Zhao Y, Nkosi A, Ismail NSM, Bekheit MS, Aboshouk DR, Aziz MN, Youssef MA, Panda SS. The Therapeutic Potential of Spirooxindoles in Cancer: A Focus on p53-MDM2 Modulation. Pharmaceuticals (Basel) 2025; 18:274. [PMID: 40006086 PMCID: PMC11859340 DOI: 10.3390/ph18020274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The p53, often referred to as the "guardian of the genome", is a well-established tumor-suppressor protein that plays a critical role in regulating the cell cycle, DNA repair, differentiation, and apoptosis, with its activity primarily modulated by the MDM2 protein (murine double minute 2, also known as HDM2 in humans). Disrupting the protein-protein interaction between p53 and MDM2 represents a promising therapeutic strategy for developing anticancer agents. Recent studies have shown that several spirooxindole-containing compounds exhibit significant antitumor properties, primarily by inhibiting the p53-MDM2 interaction. This review provides an overview of structure-based spirooxindoles that could have therapeutic potential. It highlights findings from the past decade concerning their antiproliferative properties and implications for interfering with the p53-MDM2 interaction. The discussion includes various analogs of spirooxindoles as promising candidates for optimizing leads in drug discovery programs aimed at developing novel and clinically effective agents.
Collapse
Affiliation(s)
- Adel S. Girgis
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (D.R.A.); (M.N.A.)
| | - Yujun Zhao
- State Key Laboratory of Drug Research and Small-Molecule Drug Research Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Rd, Shanghai 201203, China;
| | - Angel Nkosi
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA;
| | - Nasser S. M. Ismail
- Pharmaceutical Chemistry Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt;
| | - Mohamed S. Bekheit
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (D.R.A.); (M.N.A.)
| | - Dalia R. Aboshouk
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (D.R.A.); (M.N.A.)
| | - Marian N. Aziz
- Department of Pesticide Chemistry, National Research Centre, Dokki, Giza 12622, Egypt; (M.S.B.); (D.R.A.); (M.N.A.)
| | - M. Adel Youssef
- Department of Chemistry, Faculty of Science, Helwan University, Helwan 11795, Egypt;
| | - Siva S. Panda
- Department of Chemistry and Biochemistry, Augusta University, Augusta, GA 30912, USA;
- Department of Biochemistry and Molecular Biology, Augusta University, Augusta, GA 30912, USA
| |
Collapse
|
881
|
Pflégr V, Konečná K, Stolaříková J, Ősterreicher J, Janďourek O, Krátký M. Enhancing the antimycobacterial efficacy of pyridine-4-carbohydrazide: linkage to additional antimicrobial agents via oxocarboxylic acids. RSC Med Chem 2025; 16:767-778. [PMID: 39568598 PMCID: PMC11575622 DOI: 10.1039/d4md00663a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/13/2024] [Indexed: 11/22/2024] Open
Abstract
This study evaluates the antimycobacterial potential of novel "mutual" bioactive amides, combining pyridine-4-carbohydrazide (isoniazid, INH) with various antimicrobial agents (sulphonamides, 4-aminosalicylic acid, thiosemicarbazide, diphenyl (thio)ethers) via oxocarboxylic acids. The aim was to enhance activity against both drug-susceptible and multidrug-resistant (MDR) Mycobacterium tuberculosis and non-tuberculous strains, while overcoming drug resistance through dual-action mechanisms. Many derivatives exhibited potent antimycobacterial activity, with minimum inhibitory concentrations (MICs) as low as ≤0.25 μM, outperforming INH, especially diphenyl (thio)ethers and biphenyl analogues. Additionally, the compounds were effective against M. kansasii (MICs ≤1 μM) and inhibited MDR strains at higher concentrations (≥8 μM). The cytotoxicity assay indicated a favourable safety profile, with no significant haemolysis at 125 μM, and some compounds were even protective. Selectivity for mycobacteria was confirmed by low inhibition of Gram-positive bacteria and inactivity against Gram-negative bacteria or fungi, highlighting the potential for further development as antimycobacterial agents.
Collapse
Affiliation(s)
- Václav Pflégr
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic +420 495067166 +420 495067302
| | - Klára Konečná
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic
| | - Jiřina Stolaříková
- Laboratory for Mycobacterial Diagnostics and Tuberculosis, Regional Institute of Public Health in Ostrava Partyzánské náměstí 7 Ostrava Czech Republic
| | - Jan Ősterreicher
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic
| | - Ondřej Janďourek
- Department of Biological and Medical Sciences, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic
| | - Martin Krátký
- Department of Organic and Bioorganic Chemistry, Faculty of Pharmacy in Hradec Králové, Charles University Akademika Heyrovského 1203 500 03 Hradec Králové Czech Republic +420 495067166 +420 495067302
| |
Collapse
|
882
|
Pandey V, Pandey T. Mechanistic understanding of pH as a driving force in cancer therapeutics. J Mater Chem B 2025; 13:2640-2657. [PMID: 39878033 DOI: 10.1039/d4tb02083a] [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: 01/31/2025]
Abstract
The development of pH-directed nanoparticles for tumor targeting represents a significant advancement in cancer biology and therapeutic strategies. These innovative materials have the ability to interact with the unique acidic microenvironment of tumors. They enhance drug delivery, increase therapeutic efficacy, and reduce systemic toxicity. The acidic conditions within tumors trigger the release of drugs from pH-responsive nanoparticles, ensuring targeted and controlled delivery directly to cancer cells while minimizing damage to healthy tissues. This review comprehensively explores the design, synthesis, and application of pH-stabilized nanoparticles in cancer therapy. It delves into the mechanisms of pH-responsive behavior, such as the use of pH-sensitive polymers and cleavable linkages that respond to the acidic tumor environment. Current strategies for nanoparticle stabilization, including surface coating, core-shell nanostructures, and hybrid nanoparticles, are discussed in detail, highlighting how these approaches enhance the stability and functionality of the nanoparticles in biological systems. Recent advancements in nanoparticle-based drug delivery systems are examined, showcasing multi-functional nanoparticles that combine therapeutic and diagnostic functions, as well as those designed for combination therapy to overcome drug resistance. This review identifies future directions in the field, such as the need for improved stability and biocompatibility, controlled and predictable drug release, and overcoming regulatory and manufacturing hurdles. Herein, we have highlighted the transformative potential of pH-stabilized nanoparticles in cancer therapy, offering a pathway towards more effective and targeted cancer treatments.
Collapse
Affiliation(s)
- Vivek Pandey
- Department of Chemistry, School for Chemical engineering and Physical Sciences, Lovely Professional University, Phagwara, Punjab, India.
| | - Tejasvi Pandey
- Department of Forensic Science, School for Bio Engineering and Bio Sciences, Lovely Professional University, Phagwara, Punjab, India
| |
Collapse
|
883
|
Tredup C, Ackloo S, Beck H, Brown PJ, Bullock AN, Ciulli A, Dikic I, Edfeldt K, Edwards AM, Elkins JM, Farin HF, Fon EA, Gstaiger M, Günther J, Gustavsson AL, Häberle S, Isigkeit L, Huber KVM, Kotschy A, Krämer O, Leach AR, Marsden BD, Matsui H, Merk D, Montel F, Mulder MPC, Müller S, Owen DR, Proschak E, Röhm S, Stolz A, Sundström M, von Delft F, Willson TM, Arrowsmith CH, Knapp S. Toward target 2035: EUbOPEN - a public-private partnership to enable & unlock biology in the open. RSC Med Chem 2025; 16:457-464. [PMID: 39618964 PMCID: PMC11605244 DOI: 10.1039/d4md00735b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 11/05/2024] [Indexed: 12/12/2024] Open
Abstract
Target 2035 is a global initiative that seeks to identify a pharmacological modulator of most human proteins by the year 2035. As part of an ongoing series of annual updates of this initiative, we summarise here the efforts of the EUbOPEN project whose objectives and results are making a strong contribution to the goals of Target 2035. EUbOPEN is a public-private partnership with four pillars of activity: (1) chemogenomic library collections, (2) chemical probe discovery and technology development for hit-to-lead chemistry, (3) profiling of bioactive compounds in patient-derived disease assays, and (4) collection, storage and dissemination of project-wide data and reagents. The substantial outputs of this programme include a chemogenomic compound library covering one third of the druggable proteome, as well as 100 chemical probes, both profiled in patient derived assays, as well as hundreds of data sets deposited in existing public data repositories and a project-specific data resource for exploring EUbOPEN outputs.
Collapse
Affiliation(s)
- Claudia Tredup
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Suzanne Ackloo
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
| | - Hartmut Beck
- Drug Discovery Sciences, Research & Development, Pharmaceuticals, Bayer AG Wuppertal Nordrhein-Westfalen Germany
| | - Peter J Brown
- Structural Genomics Consortium, University of North Carolina at Chapel Hill Campus Box 7356, 120 Mason Farm Road, GMB 1070 Chapel Hill North Carolina USA
| | - Alex N Bullock
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Alessio Ciulli
- Centre for Targeted Protein Degradation, University of Dundee, School of Life Sciences 1 James Lindsay Place DD1 5JJ Dundee UK
| | - Ivan Dikic
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty Frankfurt am Main Germany
- Buchmann Institute for Molecular Lifesciences, Goethe University Frankfurt Frankfurt am Main Germany
| | - Kristina Edfeldt
- Structural Genomics Consortium, Department of Medicine, Karolinska University Hospital and Karolinska Institutet Stockholm Sweden
| | - Aled M Edwards
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
| | - Jonathan M Elkins
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Henner F Farin
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy Frankfurt am Main Hessen Germany
| | - Edward A Fon
- Department of Neurology and Neurosurgery, Montreal Neurological Institute-Hospital (The Neuro), McGill University Montreal Canada
| | - Matthias Gstaiger
- Department of Biology, Institute of Molecular Systems Biology ETH Zürich Zurich ZH Switzerland
| | | | - Anna-Lena Gustavsson
- Chemical Biology Consortium Sweden, Department of Medical Biochemistry & Biophysics, Karolinska Institute Stockholm Sweden
| | - Sandra Häberle
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Laura Isigkeit
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Kilian V M Huber
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Andras Kotschy
- Servier Research Institute of Medicinal Chemistry Budapest Hungary
| | - Oliver Krämer
- Discovery Research Coordination, Boehringer Ingelheim International GmbH Binger Straße 173 55216 Ingelheim am Rhein Germany
| | - Andrew R Leach
- European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus Hinxton Cambridge UK
| | - Brian D Marsden
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
| | - Hisanori Matsui
- Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited Fujisawa Kanagawa Japan
| | - Daniel Merk
- Ludwig-Maximilians-Universitat Munchen Munchen Germany
| | - Florian Montel
- Discovery Research Coordination, Boehringer Ingelheim Pharma GmbH & Co. KG Birkendorfer Straße 65 88397 Biberach an der Riss Germany
| | - Monique P C Mulder
- Department of Cell and Chemical Biology, Leiden University Medical Center Leiden The Netherlands
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | | | - Ewgenij Proschak
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP Theodor-Stern-Kai 7 60596 Frankfurt am Main Germany
| | - Sandra Röhm
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| | - Alexandra Stolz
- Institute of Biochemistry II, Goethe University Frankfurt, Medical Faculty Frankfurt am Main Germany
- Buchmann Institute for Molecular Lifesciences, Goethe University Frankfurt Frankfurt am Main Germany
| | - Michael Sundström
- Structural Genomics Consortium, Department of Medicine, Karolinska University Hospital and Karolinska Institutet Stockholm Sweden
| | - Frank von Delft
- Centre for Medicines Discovery, University of Oxford NDM Research Building, Roosevelt Drive Oxford Oxfordshire UK
- Diamond Light Source, Harwell Science and Innovation Campus Didcot OX11 0DE UK
| | - Timothy M Willson
- Structural Genomics Consortium, University of North Carolina at Chapel Hill Campus Box 7356, 120 Mason Farm Road, GMB 1070 Chapel Hill North Carolina USA
| | - Cheryl H Arrowsmith
- Structural Genomics Consortium, University of Toronto - St George Campus 101 College Street, MaRS Center South Tower 7th Floor Toronto Canada
- Princess Margaret Cancer Centre Toronto Ontario M5G 1L7 Canada
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University Frankfurt Frankfurt 60438 Germany
- Structural Genomics Consortium, BMLS, Goethe University Frankfurt Frankfurt 60438 Germany
| |
Collapse
|
884
|
Battisti A, Ortore MG, Vilasi S, Sgarbossa A. FLIM-Phasor Analysis (FLIM-ϕ) of Aβ-Induced Membrane Order Alterations: Towards a Cell-Based Biosensor for Early Alzheimer's Disease Diagnosis. MICROMACHINES 2025; 16:234. [PMID: 40047700 PMCID: PMC11857758 DOI: 10.3390/mi16020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 03/09/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder, and its early detection can be critical for a prompt intervention that can potentially slow down the disease progression and improve the patient's quality of life. However, a diagnosis based solely on clinical symptoms can be challenging, especially in the early stages, while the detection of specific biomarkers such as amyloid-β peptide (Aβ) and tau proteins can provide objective evidence for diagnosis. In this work, we explored the effects of Aβ peptide on cell membrane properties thanks to fluorescence lifetime imaging (FLIM) combined with the phasor analysis (FLIM-ϕ). The results showed that the membrane viscosity is altered by the presence of Aβ peptide and that cells experience this effect even at nanomolar concentrations of peptide. This considerable sensitivity opens up the possibility of envisioning a cell-based biosensor able to detect very low concentrations of Aβ in a biological fluid, thus enabling timely diagnosis and intervention.
Collapse
Affiliation(s)
- Antonella Battisti
- NEST, Nanoscience Institute-CNR and Scuola Normale Superiore, p.zza San Silvestro 12, I-56127 Pisa, Italy;
- Biophysics Institute-CNR, Via Ugo La Malfa 153, I-90146 Palermo, Italy;
| | - Maria Grazia Ortore
- Dipartimento di Scienze Della Vita e dell’Ambiente, Università Politecnica delle Marche, Via Brecce Bianche, I-60131 Ancona, Italy;
| | - Silvia Vilasi
- Biophysics Institute-CNR, Via Ugo La Malfa 153, I-90146 Palermo, Italy;
| | - Antonella Sgarbossa
- NEST, Nanoscience Institute-CNR and Scuola Normale Superiore, p.zza San Silvestro 12, I-56127 Pisa, Italy;
| |
Collapse
|
885
|
Kumar G. Natural peptides and their synthetic congeners acting against Acinetobacter baumannii through the membrane and cell wall: latest progress. RSC Med Chem 2025; 16:561-604. [PMID: 39664362 PMCID: PMC11629675 DOI: 10.1039/d4md00745j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 11/18/2024] [Indexed: 12/13/2024] Open
Abstract
Acinetobacter baumannii is one of the deadliest Gram-negative bacteria (GNB), responsible for 2-10% of hospital-acquired infections. Several antibiotics are used to control the growth of A. baumannii. However, in recent decades, the abuse and misuse of antibiotics to treat non-microbial diseases have led to the emergence of multidrug-resistant A. baumannii strains. A. baumannii possesses a complex cell wall structure. Cell wall-targeting agents remain the center of antibiotic drug discovery. Notably, the antibacterial drug discovery intends to target the membrane of the bacteria, offering several advantages over antibiotics targeting intracellular systems, as membrane-targeting agents do not have to travel through the plasma membrane to reach the cytoplasmic targets. Microorganisms, insects, and mammals produce antimicrobial peptides as their first line of defense to protect themselves from pathogens and predators. Importantly, antimicrobial peptides are considered potential alternatives to antibiotics. This communication summarises the recently identified peptides of natural origin and their synthetic congeners acting against the A. baumannii membrane by cell wall disruption.
Collapse
Affiliation(s)
- Gautam Kumar
- Department of Pharmacy, Birla Institute of Technology and Science Pilani Pilani Campus Rajasthan 333031 India
| |
Collapse
|
886
|
Pandey P, Arya DK, Kumar A, Kaushik A, Mishra YK, Rajinikanth PS. Dual ligand functionalized pH-sensitive liposomes for metastatic breast cancer treatment: in vitro and in vivo assessment. J Mater Chem B 2025; 13:2682-2694. [PMID: 39841132 DOI: 10.1039/d4tb02570a] [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: 01/23/2025]
Abstract
This research demonstrates the design and development of a novel dual-targeting, pH-sensitive liposomal (pSL) formulation of 5-Fluorouracil (5-FU), i.e., (5-FU-iRGD-FA-pSL) to manage breast cancer (BC). The motivation to explore this formulation is to overcome the challenges of systemic toxicity and non-specific targeting of 5-FU, a conventional chemotherapeutic agent. The proposed formulation also combines folic acid (FA) and iRGD peptides as targeting ligands to enhance tumor cell specificity and penetration, while the pH-sensitive liposomes ensure the controlled drug release in the acidic tumor microenvironment. The physicochemical characterization revealed that 5-FU-iRGD-FA-pSL possesses optimal size, low polydispersity index, and favorable zeta potential, enhancing its stability and targeting capabilities. In vitro studies demonstrated significantly enhanced cellular uptake, cytotoxicity, and inhibition of cell migration in MCF-7 BC cells compared to free 5-FU and non-targeted liposomal formulations. DAPI staining revealed significant apoptotic features, including chromatin condensation (CC) and nuclear fragmentation (NF), with 5-FU-iRGD-FA-pSL inducing more pronounced apoptosis compared to 5-FU-pSL. Furthermore, in vivo analysis in a BC rat model showed superior anti-tumor efficacy, reduced systemic toxicity, and improved safety profile of the 5-FU-iRGD-FA-pSL formulation. This dual-targeting pSL system presents a promising approach for enhancing the therapeutic index of 5-FU, offering a potential strategy for more effective BC treatment.
Collapse
Affiliation(s)
- Prashant Pandey
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, T6G 2H7, Canada
| | - Dilip Kumar Arya
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| | - Anit Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland 33805, FL, USA
| | - Yogendra Kumar Mishra
- Smart Materials, NanoSYD, Mads Clausen Institute, University of Southern Denmark, Alsion 2, 6400 Sønderborg, Denmark
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh, 226025, India.
| |
Collapse
|
887
|
Lomuscio MC, Corriero N, Nanna V, Piccinno A, Saviano M, Lanzilotti R, Abate C, Alberga D, Mangiatordi GF. SIGMAP: an explainable artificial intelligence tool for SIGMA-1 receptor affinity prediction. RSC Med Chem 2025; 16:835-848. [PMID: 39618965 PMCID: PMC11605305 DOI: 10.1039/d4md00722k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 11/03/2024] [Indexed: 02/21/2025] Open
Abstract
Developing sigma-1 receptor (S1R) modulators is considered a valuable therapeutic strategy to counteract neurodegeneration, cancer progression, and viral infections, including COVID-19. In this context, in silico tools capable of accurately predicting S1R affinity are highly desirable. Herein, we present a panel of 25 classifiers trained on a curated dataset of high-quality bioactivity data of small molecules, experimentally tested as potential S1R modulators. All data were extracted from ChEMBL v33, and the models were built using five different fingerprints and machine-learning algorithms. Remarkably, most of the developed classifiers demonstrated good predictive performance. The best-performing model, which achieved an AUC of 0.90, was developed using the support vector machine algorithm with Morgan fingerprints. To provide additional, user-friendly information for medicinal chemists in the rational design of S1R modulators, two independent explainable artificial intelligence (XAI) approaches were employed, namely Shapley Additive exPlanations (SHAP) and Contrastive Explanation. The top-performing model is accessible through a user-friendly web platform, SIGMAP (https://www.ba.ic.cnr.it/softwareic/sigmap/), specifically developed for this purpose. With its intuitive interface, robust predictive power, and implemented XAI approaches, SIGMAP serves as a valuable tool for the rational design of new and more effective S1R modulators.
Collapse
Affiliation(s)
- Maria Cristina Lomuscio
- Dipartimento di Medicina di Precisione e Rigenerativa e Area Jonica (DiMePRe-J), Università degli Studi di Bari Aldo Moro Piazza Giulio Cesare, 11, Policlinico 70124 Bari Italy
| | - Nicola Corriero
- CNR - Institute of Crystallography Via Amendola 122/o 70126 Bari Italy
| | - Vittoria Nanna
- CNR - Institute of Crystallography Via Amendola 122/o 70126 Bari Italy
| | - Antonio Piccinno
- Department of Computer Science, University of Bari "Aldo Moro" Via E. Orabona, 4 I-70125 Bari Italy
| | - Michele Saviano
- CNR - Institute of Crystallography Via Vivaldi 43 81100 Caserta Italy
| | - Rosa Lanzilotti
- Department of Computer Science, University of Bari "Aldo Moro" Via E. Orabona, 4 I-70125 Bari Italy
| | - Carmen Abate
- CNR - Institute of Crystallography Via Amendola 122/o 70126 Bari Italy
- Department of Pharmacy - Pharmaceutical Sciences, University of Bari "Aldo Moro" Via E. Orabona, 4 I-70125 Bari Italy
| | - Domenico Alberga
- CNR - Institute of Crystallography Via Amendola 122/o 70126 Bari Italy
| | | |
Collapse
|
888
|
Reymova F, Sever B, Topalan E, Sevimli-Gur C, Can M, Tuyun AF, Başoğlu F, Ece A, Otsuka M, Fujita M, Demirci H, Ciftci H. Design, Synthesis, and Mechanistic Anticancer Evaluation of New Pyrimidine-Tethered Compounds. Pharmaceuticals (Basel) 2025; 18:270. [PMID: 40006082 PMCID: PMC11859636 DOI: 10.3390/ph18020270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/12/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Despite recent breakthroughs in cancer treatment, non-small cell lung cancer (NSCLC) and breast cancer remain major causes of death from all malignancies. The epidermal growth factor receptor (EGFR) is an important mediator of the pathways involved in cell proliferation, apoptosis, and angiogenesis. Thus, its overexpression triggers several types of cancer, including NSCLC and breast cancer. Methods: In the current study, we synthesized new pyrimidine-tethered compounds (chalcone derivative (B-4), pyrazoline-carbothioamide (B-9), and pyrazoline-thiazole hybrids (BH1-7)). These compounds were then tested for cytotoxicity against A549 NSCLC and MCF-7 breast cancer cells. Results: Of these, B-4 displayed significant cytotoxicity against both cells (IC50 = 6.70 ± 1.02 µM for MCF-7; IC50 = 20.49 ± 2.7 µM for A549) compared to the standard agent lapatinib (IC50 = 9.71 ± 1.12 µM for MCF-7; IC50 = 18.21 ± 3.25 µM for A549). The anticancer potential of B-4 between Jurkat leukemic T cells and peripheral blood mononuclear cells (PBMCs) (healthy) was found to be selective. Mechanistically, 11.9% and 10.2% of A549 and MCF-7 cells treated with B-4, respectively, underwent apoptosis and B-4 produced 46% EGFR inhibition at a concentration of 10 μM. The B-4/EGFR complex obtained after induced fit docking was subjected to 300 ns of molecular dynamics simulation, which confirmed the stability of the complex in a mimicked biological environment. On the other hand, B-4 was shown to have drug-like properties by in silico pharmacokinetic estimation. Conclusions: B-4 is an EGFR inhibitor and apoptosis inducer for future NSCLC and breast cancer studies.
Collapse
Affiliation(s)
- Farida Reymova
- Department of Bioengineering Sciences, Izmir Katip Celebi University, Izmir 35620, Türkiye; (F.R.); (M.C.)
| | - Belgin Sever
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Anadolu University, Eskisehir 26470, Türkiye;
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
| | - Edanur Topalan
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (E.T.); (H.D.)
| | - Canan Sevimli-Gur
- Department of Basic Pharmaceutical Sciences, Faculty of Pharmacy, Izmir Katip Celebi University, Izmir 35620, Türkiye;
| | - Mustafa Can
- Department of Bioengineering Sciences, Izmir Katip Celebi University, Izmir 35620, Türkiye; (F.R.); (M.C.)
| | - Amaç Fatih Tuyun
- Department of Chemistry, Faculty of Science, Istanbul University, Fatih, Istanbul 34126, Türkiye;
| | - Faika Başoğlu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, European University of Lefke, Northern Cyprus, TR-10, Mersin 99800, Türkiye;
| | - Abdulilah Ece
- Department of Medical Biochemistry, Faculty of Medicine, Biruni University, İstanbul 34015, Türkiye;
| | - Masami Otsuka
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
- Department of Drug Discovery, Science Farm Ltd., Kumamoto 862-0976, Japan
| | - Mikako Fujita
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
| | - Hasan Demirci
- Department of Molecular Biology and Genetics, Koc University, Istanbul 34450, Türkiye; (E.T.); (H.D.)
| | - Halilibrahim Ciftci
- Department of Bioengineering Sciences, Izmir Katip Celebi University, Izmir 35620, Türkiye; (F.R.); (M.C.)
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, Kumamoto 862-0973, Japan; (M.O.); (M.F.)
- Department of Drug Discovery, Science Farm Ltd., Kumamoto 862-0976, Japan
- Department of Molecular Biology and Genetics, Burdur Mehmet Akif Ersoy University, Istiklal Campus, Burdur 15030, Türkiye
| |
Collapse
|
889
|
Iizumi K, Yamaguchi J. Transformative reactions in nitroarene chemistry: C-N bond cleavage, skeletal editing, and N-O bond utilization. Org Biomol Chem 2025; 23:1746-1772. [PMID: 39831336 DOI: 10.1039/d4ob01928h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Nitroarenes are highly versatile building blocks in organic synthesis, playing a pivotal role in various reactions. Common transformations involving nitroarenes include nucleophilic aromatic substitution (SNAr) reactions, where the nitro group functions both as a potent electron-withdrawing group that activates the aromatic ring and as a leaving group facilitating the substitution. Additionally, the direct transformation of nitro groups, such as reduction-driven syntheses of amines and carboxylic acids, as well as ipso-substitution SNAr reactions, have been extensively explored. Interactions between ortho-nitro groups and neighboring substituents also provide unique opportunities for selective transformations. However, beyond these well-established processes, direct transformations of nitro groups have been relatively limited. In recent years, significant advancements have been made in alternative methodologies for nitro group transformations. This review focuses on the latest progress in novel transformations of nitroarenes, with emphasis on three major categories: (i) functional group transformations involving C-N bond cleavage in nitroarenes, (ii) skeletal editing via nitrene intermediates generated by N-O bond cleavage, and (iii) the utilization of nitroarenes as an oxygen source through N-O bond cleavage. These developments under-score the expanding utility of nitroarenes in modern organic synthesis.
Collapse
Affiliation(s)
- Keiichiro Iizumi
- Department of Applied Chemistry, Waseda University, 513 Wasedatsurumakicho, Shinjuku, Tokyo 162-0041, Japan.
| | - Junichiro Yamaguchi
- Department of Applied Chemistry, Waseda University, 513 Wasedatsurumakicho, Shinjuku, Tokyo 162-0041, Japan.
| |
Collapse
|
890
|
Porras M, Hernández D, Boto A. Short Synthesis of Structurally Diverse N-Acylhomoserine Lactone Analogs and Discovery of Novel Quorum Quenchers Against Gram-Negative Pathogens. Int J Mol Sci 2025; 26:1775. [PMID: 40004238 PMCID: PMC11855090 DOI: 10.3390/ijms26041775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Revised: 02/08/2025] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
Quorum quenchers are emerging as an alternative to conventional antimicrobials, since they hinder the development of virulence or resistance mechanisms but without killing the microorganisms, thus, reducing the risk of antimicrobial resistance. Many quorum quenchers are analogs of the natural quorum-sensing signaling molecules or autoinducers. Thus, different analogs of natural N-acylhomoserine lactones (AHLs) have been reported for controlling virulence or reducing the production of biofilms in Gram-negative pathogens. Herein we report the preparation of AHL analogs with a variety of N-substituents in just two steps from readily available N-substituted hydroxyproline esters. The substrates underwent an oxidative radical scission of the pyrrolidine ring. The resulting N-substituted β-aminoaldehyde underwent reduction and in situ cyclization to give a variety of homoserine lactones, with N- and N,N-substituted amino derivatives and with high optical purity. The libraries were screened for the inhibition of violacein production in Chromobacterium violaceum, a Gram-negative pathogen. For the first time, N,N-disubstituted AHL analogs were studied. Several N-sulfonyl derivatives, one carbamoyl, and one N-alkyl-N-sulfonyl homoserine lactone displayed a promising inhibitory activity. Moreover, they did not display microbicide action against S. aureus, C. jejuni, S. enterica, P. aeruginosa, and C. albicans, confirming a pure QQ activity. The determination of structure-activity relationships and in silico ADME studies are also reported, which are valuable for the design of next generations QQ agents.
Collapse
Affiliation(s)
| | - Dácil Hernández
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Tenerife, Spain;
| | - Alicia Boto
- Instituto de Productos Naturales y Agrobiología del CSIC, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Tenerife, Spain;
| |
Collapse
|
891
|
Hossain MA, Brahme RR, Miller BC, Amin J, de Barros M, Schneider JL, Auclair JR, Mattos C, Wang Q, Agar NYR, Greenblatt DJ, Manetsch R, Agar JN. Mass spectrometry methods and mathematical PK/PD model for decision tree-guided covalent drug development. Nat Commun 2025; 16:1777. [PMID: 39971904 PMCID: PMC11839910 DOI: 10.1038/s41467-025-56985-6] [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: 10/27/2023] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
Covalent drug discovery efforts are growing rapidly but have major unaddressed limitations. These include high false positive rates during hit-to-lead identification; the inherent uncoupling of covalent drug concentration and effect [i.e., uncoupling of pharmacokinetics (PK) and pharmacodynamics (PD)]; and a lack of bioanalytical and modeling methods for determining PK and PD parameters. We present a covalent drug discovery workflow that addresses these limitations. Our bioanalytical methods are based upon a mass spectrometry (MS) assay that can measure the percentage of drug-target protein conjugation (% target engagement) in biological matrices. Further we develop an intact protein PK/PD model (iPK/PD) that outputs PK parameters (absorption and distribution) as well as PD parameters (mechanism of action, protein metabolic half-lives, dose, regimen, effect) based on time-dependent target engagement data. Notably, the iPK/PD model is applicable to any measurement (e.g., bottom-up MS and other drug binding studies) that yields % of target engaged. A Decision Tree is presented to guide researchers through the covalent drug development process. Our bioanalytical methods and the Decision Tree are applied to two approved drugs (ibrutinib and sotorasib); the most common plasma off-target, human serum albumin; three protein targets (KRAS, BTK, SOD1), and to a promising SOD1-targeting ALS drug candidates.
Collapse
Affiliation(s)
- Md Amin Hossain
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
- Department of Neurosurgery and Radiology, Brigham and Women's Hospital, Harvard Medical School;Boston, Massachusetts, 02115, USA
| | - Rutali R Brahme
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
| | - Brandon C Miller
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Jakal Amin
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
| | - Marcela de Barros
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Jaime L Schneider
- Massachusetts General Hospital Cancer Center, Harvard Medical School;Boston, Massachusetts, 02114, USA
| | - Jared R Auclair
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA
| | - Carla Mattos
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Qingping Wang
- Sanofi US, Drug Metabolism and Pharmacokinetics;Cambridge, Massachusetts, 02141, USA
| | - Nathalie Y R Agar
- Department of Neurosurgery and Radiology, Brigham and Women's Hospital, Harvard Medical School;Boston, Massachusetts, 02115, USA
| | | | - Roman Manetsch
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA
- Department of Pharmaceutical Sciences, Northeastern University;Boston, Massachusetts, 02115, USA
| | - Jeffrey N Agar
- Department of Chemistry and Chemical Biology, Northeastern University;Boston, Massachusetts, 02115, USA.
- Barnett Institute of Chemical and Biological Analysis;Boston, Massachusetts, 02115, USA.
- Department of Pharmaceutical Sciences, Northeastern University;Boston, Massachusetts, 02115, USA.
| |
Collapse
|
892
|
Ma D, Wu Z, Zhang M, Mao J, Xu W, Jiang L, Wang Z. Glutathiones' life in multi-cancers: especially their potential micropetides in liver hepatocellular carcinoma. Discov Oncol 2025; 16:201. [PMID: 39966283 PMCID: PMC11836257 DOI: 10.1007/s12672-025-01945-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 02/05/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Glutathione plays critical roles in detoxifying xenobiotics, cell signaling, cell death and the antioxidant defence in an emerging body of evidence, the most abundant intracellular low molecular weight thiol in tissues. However, all glutathione metabolism pertinent genes (GMPGs) expression and their diagnostic/prognostic/micropeptide potential analyses have not been investigated to perform in pan-cancers. METHODS We gained GMPGs from the MsigDB 7.2, 12,123 samples were used to reveal the differentially expressed genes (DEGs) and the survival analysis in 32 types of cancers from TCGA, GTEx, and GEO datasets for the first time. All statistical analyses were performed by R for bioinformatics, such as DEGs, prognostic, diagnostic analysis, ceRNA, micropeptide prediction and immune infiltration. In addition, we utilized siRNA technology to target knockdown the expression of the G6PD gene in Huh7 hepatocellular carcinoma cells. RESULTS G6PD was significantly expressed and poor prognosis in liver hepatocellular carcinoma (LIHC) and predicted RBM26-AS1 encoded micropeptide might target G6PD in LIHC. In vitro experiments show that G6PD knockout in Huh7 cells reduces their proliferation, migration, and invasion capabilities. CONCLUSIONS We confirmed that G6PD played a crucial role in the occurrence and progression of LIHC. G6PD is positively associated with Th2 cells in LIHC, regulating immune responses in the immune system. We considered that micropeptide RBM26-AS1 might be a new player involved in LIHC by interacting with G6PD, might perform a key function in liver cancer.
Collapse
Affiliation(s)
- Didi Ma
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation (Wannan Medical College), Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Zhenguo Wu
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation (Wannan Medical College), Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Mengying Zhang
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation (Wannan Medical College), Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Jian Mao
- Yangtze River Delta Information Intelligence Innovation Research Institute, Wuhu, 241000, China
| | - Wenqin Xu
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation (Wannan Medical College), Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China
| | - Lan Jiang
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation (Wannan Medical College), Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China.
| | - Zuzhen Wang
- Anhui Province Key Laboratory of Non-Coding RNA Basic and Clinical Transformation (Wannan Medical College), Central Laboratory, Yijishan Hospital of Wannan Medical College, Wuhu, 241000, China.
- Center of Reproductive Medicine, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| |
Collapse
|
893
|
Lira RLDS, Nogueira FAB, Campos RDFPDC, Ferreira DRM, Roxo PLBT, de Azevedo CCS, Gimenes ECM, Bastos RLC, Nascimento CEC, Nunes FDO, Marques MCP, Campos CDL, Martinez CG, Zagmignan A, Silva LCN, Ribeiro RM, de Azevedo dos Santos APS, Carvalho RC, de Sousa EM. Mycobacterium abscessus subsp. massiliense: Biofilm Formation, Host Immune Response, and Therapeutic Strategies. Microorganisms 2025; 13:447. [PMID: 40005812 PMCID: PMC11858063 DOI: 10.3390/microorganisms13020447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/08/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Infection by Mycobacterium abscessus subsp. massiliense poses a growing public health threat, especially to immunocompromised individuals. The pathogenicity of this mycobacterium is directly linked to its ability to form biofilms, complex structures that confer resistance to antibiotics and the host immune response. The extracellular matrix of the biofilm acts as a physical barrier, hindering the penetration of drugs and the action of the immune system, while also inducing a slow-growth state that reduces susceptibility to antibiotics. Current therapies, which involve prolonged use of multiple antibiotics, are often ineffective and cause significant side effects. Therefore, it is essential to explore new strategies targeting bacterial resistance and biofilm destruction. This narrative review explores the biofilm-forming capacity of Mycobacterium abscessus subsp. massiliense and the potential of novel therapeutic strategies. Promising approaches include inhibiting biofilm formation, developing drugs with improved penetration of the extracellular matrix, combination therapies with agents that destabilize the biofilm structure, and modulating the host immune response. Investing in research and development of new therapeutic strategies is essential to combat this resistant bacterium and improve patient outcomes.
Collapse
Affiliation(s)
- Roseane Lustosa de Santana Lira
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Flávio Augusto Barros Nogueira
- Graduate Program in Biodiversity and Biotechnology, Amazônia—BIONORTE, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (F.A.B.N.); (A.Z.)
| | | | - Dayenne Regina Mota Ferreira
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Pedro Lucas Brito Tromps Roxo
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Caio César Santana de Azevedo
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Eleonôra Costa Monteiro Gimenes
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Ruan Lucas Costa Bastos
- Undergraduate in Medicine, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (P.L.B.T.R.); (C.C.S.d.A.); (E.C.M.G.); (R.L.C.B.)
| | - Camila Evangelista Carnib Nascimento
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Flávia Danyelle Oliveira Nunes
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Mayane Cristina Pereira Marques
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Carmem Duarte Lima Campos
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Camila Guerra Martinez
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Adrielle Zagmignan
- Graduate Program in Biodiversity and Biotechnology, Amazônia—BIONORTE, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (F.A.B.N.); (A.Z.)
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Luís Cláudio Nascimento Silva
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| | - Rachel Melo Ribeiro
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Ana Paula Silva de Azevedo dos Santos
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Rafael Cardoso Carvalho
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
| | - Eduardo Martins de Sousa
- Graduate Program in Health Sciences, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (R.L.d.S.L.); (C.E.C.N.); (F.D.O.N.); (M.C.P.M.); (C.D.L.C.); (R.M.R.); (A.P.S.d.A.d.S.); (R.C.C.)
- Graduate Program in Biodiversity and Biotechnology, Amazônia—BIONORTE, Federal University of Maranhão—UFMA, São Luís 65080-805, Brazil; (F.A.B.N.); (A.Z.)
- Graduate Program in Biosciences Applied to Health, CEUMA University—UNICEUMA, São Luís 65075-120, Brazil; (R.d.F.P.d.C.C.); (D.R.M.F.); (C.G.M.); (L.C.N.S.)
| |
Collapse
|
894
|
Parvin N, Joo SW, Mandal TK. Nanomaterial-Based Strategies to Combat Antibiotic Resistance: Mechanisms and Applications. Antibiotics (Basel) 2025; 14:207. [PMID: 40001450 PMCID: PMC11852044 DOI: 10.3390/antibiotics14020207] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Revised: 02/05/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The rapid rise of antibiotic resistance has become a global health crisis, necessitating the development of innovative strategies to combat multidrug-resistant (MDR) pathogens. Nanomaterials have emerged as promising tools in this fight, offering unique physicochemical properties that enhance antibiotic efficacy, overcome resistance mechanisms, and provide alternative therapeutic approaches. This review explores the diverse nanomaterial-based strategies used to combat antibiotic resistance, focusing on their mechanisms of action and practical applications. Nanomaterials such as metal nanoparticles, carbon-based nanomaterials, and polymeric nanostructures exhibit antibacterial properties through various pathways, including the generation of reactive oxygen species (ROS), disruption of bacterial membranes, and enhancement of antibiotic delivery. Additionally, the ability of nanomaterials to bypass traditional resistance mechanisms, such as biofilm formation and efflux pumps, has been demonstrated in numerous studies. This review also discusses the synergistic effects observed when nanomaterials are combined with conventional antibiotics, leading to increased bacterial susceptibility and reduced required dosages. By highlighting the recent advancements and clinical applications of nanomaterial-antibiotic combinations, this paper provides a comprehensive overview of how nanomaterials are reshaping the future of antibacterial therapies. Future research directions and challenges, including toxicity and scalability, are also addressed to guide the development of safer, more effective nanomaterial-based antibacterial treatments.
Collapse
Affiliation(s)
| | - Sang Woo Joo
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| | - Tapas K. Mandal
- School of Mechanical Engineering, Yeungnam University, Gyeongsan 38541, Republic of Korea;
| |
Collapse
|
895
|
Liu YQ, Li ZZ, Han YL, Wang QB. The role of efferocytosis in inflammatory bowel disease. Front Immunol 2025; 16:1524058. [PMID: 40040696 PMCID: PMC11876057 DOI: 10.3389/fimmu.2025.1524058] [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: 11/07/2024] [Accepted: 01/16/2025] [Indexed: 03/06/2025] Open
Abstract
Efferocytosis is the process by which various phagocytes clear apoptotic cells. In recent years, an increasing body of evidence has emphasized the importance of efferocytosis in maintaining internal homeostasis. Intestinal macrophages play a crucial role in modulating intestinal inflammation and promoting tissue repair. Inflammatory bowel disease (IBD) is a chronic, progressive, and relapsing condition, primarily marked by the presence of ulcers in the digestive tract. The exact mechanisms underlying IBD are not yet fully understood, and current treatment approaches mainly aim at repairing the damaged intestinal mucosa and reducing inflammatory responses to ease symptoms.This article provides new perspectives on IBD treatment and clinical management by examining the expression of macrophage efferocytosis-related molecules, the effects of efferocytosis on IBD development, the various roles of macrophage efferocytosis in IBD, and treatment strategies for IBD that focus on efferocytosis.
Collapse
Affiliation(s)
- Yi-Qian Liu
- Institute of Acupuncture and Moxibustion, Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Zhan-Zhan Li
- Academy of Traditional Chinese Medicine, Henan University of Chinese Medicine, Zhengzhou, China
| | - Yong-Li Han
- Acupuncture Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| | - Qing-Bo Wang
- Acupuncture Department, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
896
|
Wang J, Qin Y, Cui K, Li X, Cui M, Cao S, Zhang L, Shen Q, Wang T, Li F. Solvent-controlled silver catalyzed radical transformation of α-imino-oxy acids with cyclic aldimines. Chem Commun (Camb) 2025; 61:3359-3362. [PMID: 39882770 DOI: 10.1039/d4cc05675b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
A silver-catalyzed cross coupling of cyclic aldimines and α-imino-oxy acids has been developed. The solvent-dependent reaction could selectively deliver either cyclic imine moiety retained nitriles or ring-opened oxonitriles in moderate yields. The mechanistic studies show that the reaction undergoes a radical pathway.
Collapse
Affiliation(s)
- Jingjing Wang
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Yuran Qin
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Ke Cui
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Xueqi Li
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Mingyue Cui
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Sheng Cao
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Linbo Zhang
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| | - Qin Shen
- Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310027, P. R. China
| | - Teng Wang
- College of Chemistry, Beijing University of Chemical Technology, Beijing, 100029, P. R. China.
| | - Feng Li
- Lunan Institute of Intelligent Biomedical Engineering, College of Food Science and Pharmaceutical Engineering, Zaozhuang University, Zaozhuang, 277160, P. R. China.
| |
Collapse
|
897
|
Wang G, Seidler NJ, Röhm S, Pan Y, Liang XJ, Haarer L, Berger BT, Sivashanmugam SA, Wydra VR, Forster M, Laufer SA, Chaikuad A, Gehringer M, Knapp S. Probing the Protein Kinases' Cysteinome by Covalent Fragments. Angew Chem Int Ed Engl 2025; 64:e202419736. [PMID: 39716901 DOI: 10.1002/anie.202419736] [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: 10/12/2024] [Revised: 11/23/2024] [Accepted: 12/09/2024] [Indexed: 12/25/2024]
Abstract
Protein kinases are important drug targets, yet specific inhibitors have been developed for only a fraction of the more than 500 human kinases. A major challenge in designing inhibitors for highly related kinases is selectivity. Unlike their non-covalent counterparts, covalent inhibitors offer the advantage of selectively targeting structurally similar kinases by modifying specific protein side chains, particularly non-conserved cysteines. Previously, covalent fragment screens yielded potent and selective inhibitors for individual kinases such as ERK1/2 but have not been applied to the broader kinome. Furthermore, many of the accessible cysteine positions have not been addressed so far. Here, we outline a generalizable approach to sample ATP-site cysteines with fragment-like covalent inhibitors. We present the development of a kinase-focused covalent fragment library and its systematic screening against a curated selection of 47 kinases, with 60 active site-proximal cysteines using LC/MS and differential scanning fluorimetry (DSF) assays, followed by hit validation through various complementary techniques. Our findings expand the repertoire of targetable cysteines within protein kinases, provide insight into unique binding modes identified from crystal structures and deliver isoform-specific hits with promising profiles as starting points for the development of highly potent and selective covalent inhibitors.
Collapse
Affiliation(s)
- Guiqun Wang
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), DKTK Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| | - Nico J Seidler
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
| | - Sandra Röhm
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Yufeng Pan
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Xiaojun Julia Liang
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Lisa Haarer
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Benedict-Tilman Berger
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Saran Aswathaman Sivashanmugam
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Valentin R Wydra
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
| | - Michael Forster
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Stefan A Laufer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
- Tübingen Center for Academic Drug Discovery & Development (TüCAD2), Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Apirat Chaikuad
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
| | - Matthias Gehringer
- Institute of Pharmaceutical Sciences, Department of Pharmaceutical/Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Faculty of Medicine, Institute for Biomedical Engineering, Department for Medicinal Chemistry, Eberhard Karls University Tübingen, Auf der Morgenstelle 8, D-72076, Tübingen, Germany
- Cluster of Excellence iFIT (EXC 2180) 'Image-Guided & Functionally Instructed Tumor Therapies', Eberhard Karls University Tübingen, D-72076, Tübingen, Germany
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 9, D-60438, Frankfurt am Main, Germany
- Structure Genomics Consortium (SGC), Buchmann Institute for Life Sciences, Johann Wolfgang Goethe-University, Max-von-Laue-Str. 15, D-60438, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK), DKTK Site Frankfurt-Mainz, German Cancer Research Center (DKFZ), D-69120, Heidelberg, Germany
| |
Collapse
|
898
|
Chen S, Xu Z, Yuan B, Gou XY, Ackermann L. Difunctionalization of bicyclo[1.1.0]butanes enabled by merging C-C cleavage and ruthenium-catalysed remote C-H activation. NATURE SYNTHESIS 2025; 4:655-663. [PMID: 40375955 PMCID: PMC12075002 DOI: 10.1038/s44160-025-00745-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 01/22/2025] [Indexed: 05/18/2025]
Abstract
The high fraction of sp 3-hybridized carbon atom (Fsp 3) character of cyclobutane derivatives renders them as highly promising bioisosteres for otherwise typically flat arenes. Here, to address the current needs in medicinal chemistry for Fsp 3-rich molecules, we disclose a distinct strategy that exploits the merger of C-C scission in bicyclo[1.1.0]butanes (BCBs) with ruthenium-catalysed remote C-H functionalization of heteroarenes, affording densely substituted cyclobutanes in a chemo-controlled manner. This approach enabled the rapid and efficient synthesis of versatile tri- and tetrasubstituted cyclobutanes by coupling a wide range of mono- or disubstituted BCBs with heteroarenes and alkyl halides under mild reaction conditions, featuring ample substrate scope. The C-C/C-H functionalization was ensured by a multifunctional ruthenium(II) catalyst that enabled ruthenacycle-mediated halogen-atom transfer (Ru-XAT), as well as the selective functionalization of BCBs by strain release. Experimental and computational mechanistic studies unravelled a multi-catalysis manifold, while the C-H/C-C functionalization strategy allowed for telescoping late-stage modification.
Collapse
Affiliation(s)
- Shan Chen
- Wöhler-Research Institute for Sustainable Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Zhimin Xu
- Wöhler-Research Institute for Sustainable Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Binbin Yuan
- Wöhler-Research Institute for Sustainable Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Xue-Ya Gou
- Wöhler-Research Institute for Sustainable Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
| | - Lutz Ackermann
- Wöhler-Research Institute for Sustainable Chemistry, Georg-August-Universität Göttingen, Göttingen, Germany
| |
Collapse
|
899
|
van Wier SP, Beekman AM. Peptide design to control protein-protein interactions. Chem Soc Rev 2025; 54:1684-1698. [PMID: 39817557 PMCID: PMC11736853 DOI: 10.1039/d4cs00243a] [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: 05/20/2024] [Indexed: 01/18/2025]
Abstract
Targeting of protein-protein interactions has become of huge interest in every aspect of medicinal and biological sciences. The control of protein interactions selectively offers the opportunity to control biological processes while limiting off target effects. This interest has massively increased with the development of cryo-EM and protein structure prediction with tools such as RosettaFold and AlphaFold. When designing molecules to control protein interactions, either inhibition or stabilisation, a starting point is commonly peptide design. This tutorial review describes that process, highlighting the selection of an initial sequence with and without structural information. Subsequently, methods for how the sequence can be analysed for key residues and how this information can be used to optimise the ligand efficiency are highlighted. Finally a discussion on how peptides can be further modified to increase their affinity and cell permeability, improving their drug-like properties, is presented.
Collapse
Affiliation(s)
- Suzanne P van Wier
- School of Chemistry, Pharmacy & Pharmacology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| | - Andrew M Beekman
- School of Chemistry, Pharmacy & Pharmacology, University of East Anglia, Norwich Research Park, Norwich, NR4 7TJ, UK.
| |
Collapse
|
900
|
Sharma P, Paul K. Selective Recognition of Oncogene Promoter C-Myc G-Quadruplex: Design, Synthesis, and In Vitro Evaluation of Naphthalimide and Imidazo[1,2- a]pyrazines for Their Anticancer Activity. ACS APPLIED BIO MATERIALS 2025; 8:1377-1396. [PMID: 39844620 DOI: 10.1021/acsabm.4c01666] [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: 01/24/2025]
Abstract
c-Myc is a transcription factor that is overexpressed in most human cancers. Despite its challenging nature, we have developed a series of naphthalimide-imidazopyrazine conjugates to target c-Myc. The library of synthesized derivatives was tested for their anticancer activity against a nine-panel of cancer cell lines. Compound 8eb showed excellent cytotoxicity against all the tested cancer cell lines, with the range of growth inhibition from -98.79% to 96.62% at a single-dose concentration of 10-5 M. Further, 8eb was employed for a 5-dose assay against the same cancer cell lines, which showed efficacy at varying concentrations with an MG-MID GI50 value of 2.61 μM. Biophysical studies were performed to explore the interaction of 8eb with c-Myc Pu27 over ct-DNA, oncogene promotor Pu22, and human telomere, with a binding constant value of 1.3 × 107 M-1. Additionally, experiments were performed to get insights into the interaction mechanism between 8eb and the c-Myc oncogene promoter. A molecular docking study unveiled the stacking of the compound with G4 DNA through groove binding, where very few reports are available, with a favorable binding energy of -9.2 kcal/mol. Moreover, the pharmacokinetic study and HOMO-LUMO energy gap analysis underscored the potency of the active candidate. The compound's binding ability toward HSA was also assessed, where results suggested effective binding of the compound to HSA, revealing its potential for easy delivery to the target site. The above findings suggested that these newly synthesized candidates with potent anticancer activity offer a promising avenue as G4 DNA c-Myc stabilizers.
Collapse
Affiliation(s)
- Palak Sharma
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India
| | - Kamaldeep Paul
- Department of Chemistry and Biochemistry, Thapar Institute of Engineering and Technology, Patiala 147001, India
| |
Collapse
|