723 results on '"rational drug design"'
Search Results
2. Discovery of novel tetracyclic quinazolines as multi-KRAS inhibitors for the treatment of solid tumors
- Author
-
Hong, Changhee, Lee, Jooyun, Kim, Dowon, Hong, Soojung, Park, Jongseon, Hong, Sungjun, An, Kyung mi, Lee, Myongjae, and Han, Gyoonhee
- Published
- 2025
- Full Text
- View/download PDF
3. cPEPmatch Webserver: A comprehensive tool and database to aid rational design of cyclic peptides for drug discovery
- Author
-
Brianda L. Santini, Stephanie Wendel, Niklas Halbwedl, Asha Knipp, and Martin Zacharias
- Subjects
Cyclic peptides ,Rational drug design ,Molecular dynamics ,Protein-protein complexes ,Protein binding modulation ,Biotechnology ,TP248.13-248.65 - Abstract
Cyclic peptides have emerged as versatile scaffolds in drug discovery due to their stability and specificity. Here, we present the cPEPmatch webserver (accessible at https://t38webservices.nat.tum.de/cpepmatch/), an easy-to-use interface for the rational design of cyclic peptides targeting protein-protein interactions combined with a semi-quantitative evaluation of binding stability. This platform also offers access to a comprehensive database of cyclic peptide crystal structures. We demonstrate the webserver's utility through a series of case studies involving medically relevant protein systems, highlighting its potential to significantly advance drug discovery efforts.
- Published
- 2024
- Full Text
- View/download PDF
4. Novel potent molecular glue degraders against broad range of hematological cancer cell lines via multiple neosubstrates degradation
- Author
-
Pengyun Li, Xiaotong Hu, Zhiya Fan, Shiyang Sun, Qijie Ran, Ting Wei, Pengli Wei, Qiyu Jiang, Jian Yan, Ning Yang, Changkai Jia, Tingting Yang, Yaqiu Mao, Xu Cai, Tingting Xu, Zhiyuan Zhao, Xiaohong Qian, Weijie Qin, Xiaomei Zhuang, Feng Fan, Junhai Xiao, Zhibing Zheng, and Song Li
- Subjects
Hematological cancer ,Molecular glue ,IMIDs ,Cereblon ,Rational drug design ,Neosubstrate ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Background Targeted protein degradation of neosubstrates plays a crucial role in hematological cancer treatment involving immunomodulatory imide drugs (IMiDs) therapy. Nevertheless, the persistence of inevitable drug resistance and hematological toxicities represents a significant obstacle to their clinical effectiveness. Methods Phenotypic profiling of a small molecule compounds library in multiple hematological cancer cell lines was conducted to screen for hit degraders. Molecular dynamic-based rational design and cell-based functional assays were conducted to develop more potent degraders. Multiple myeloma (MM) tumor xenograft models were employed to investigate the antitumor efficacy of the degraders as single or combined agents with standard of care agents. Unbiased proteomics was employed to identify multiple therapeutically relevant neosubstrates targeted by the degraders. MM patient-derived cell lines (PDCs) and a panel of solid cancer cell lines were utilized to investigate the effects of candidate degrader on different stage of MM cells and solid malignancies. Unbiased proteomics of IMiDs-resistant MM cells, cell-based functional assays and RT-PCR analysis of clinical MM specimens were utilized to explore the role of BRD9 associated with IMiDs resistance and MM progression. Results We identified a novel cereblon (CRBN)-dependent lead degrader with phthalazinone scaffold, MGD-4, which induced the degradation of Ikaros proteins. We further developed a novel potent candidate, MGD-28, significantly inhibited the growth of hematological cancer cells and induced the degradation of IKZF1/2/3 and CK1α with nanomolar potency via a Cullin-CRBN dependent pathway. Oral administration of MGD-4 and MGD-28 effectively inhibited MM tumor growth and exhibited significant synergistic effects with standard of care agents. MGD-28 exhibited preferentially profound cytotoxicity towards MM PDCs at different disease stages and broad antiproliferative activity in multiple solid malignancies. BRD9 modulated IMiDs resistance, and the expression of BRD9 was significant positively correlated with IKZF1/2/3 and CK1α in MM specimens at different stages. We also observed pronounced synergetic efficacy between the BRD9 inhibitor and MGD-28 for MM treatment. Conclusions Our findings present a strategy for the multi-targeted degradation of Ikaros proteins and CK1α against hematological cancers, which may be expanded to additional targets and indications. This strategy may enhance efficacy treatment against multiple hematological cancers and solid tumors.
- Published
- 2024
- Full Text
- View/download PDF
5. Development of Novel ROCK Inhibitors via 3D-QSAR and Molecular Docking Studies: A Framework for Multi-Target Drug Design.
- Author
-
Beljkas, Milan, Petkovic, Milos, Vuletic, Ana, Djuric, Ana, Santibanez, Juan Francisco, Srdic-Rajic, Tatjana, Nikolic, Katarina, and Oljacic, Slavica
- Subjects
- *
RHO-associated kinases , *COMPUTER-assisted drug design , *DRUG design , *CYTOSKELETON , *MOLECULAR docking - Abstract
Background/Objectives: Alterations in the actin cytoskeleton correlates to tumor progression and affect critical cellular processes such as adhesion, migration and invasion. Rho-associated coiled-coil-containing protein kinases (ROCK1 and ROCK2), important regulators of the actin cytoskeleton, are frequently overexpressed in various malignancies. The aim of this study was therefore to identify the key structural features of ROCK1/ROCK2 inhibitors using computer-aided drug design (CADD) approaches. In addition, new developed ROCK inhibitors provided a significant framework for the development of multitarget therapeutics—ROCK/HDAC (histone deacetylases) multitarget inhibitors. Methods: 3D-QSAR (Quantitative structure-activity relationship study) and molecular docking study were employed in order to identify key structural features that positively correlate with ROCK inhibition. MDA-MB-231, HCC1937, Panc-1 and Mia PaCa-2 cells were used for evaluation of anticancer properties of synthesized compounds. Results: C-19 showed potent anti-cancer properties, especially enhancement of apoptosis and cell cycle modulation in pancreatic cancer cell lines. In addition, C-19 and C-22 showed potent anti-migratory and anti-invasive effects comparable to the well-known ROCK inhibitor fasudil. Conclusions: In light of the results of this study, we propose a novel multi-target approach focusing on developing dual HDAC/ROCK inhibitors based on the structure of both C-19 and C-22, exploiting the synergistic potential of these two signaling pathways to improve therapeutic efficacy in metastatic tumors. Our results emphasize the potential of multi-target ROCK inhibitors as a basis for future cancer therapies. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
6. Novel potent molecular glue degraders against broad range of hematological cancer cell lines via multiple neosubstrates degradation.
- Author
-
Li, Pengyun, Hu, Xiaotong, Fan, Zhiya, Sun, Shiyang, Ran, Qijie, Wei, Ting, Wei, Pengli, Jiang, Qiyu, Yan, Jian, Yang, Ning, Jia, Changkai, Yang, Tingting, Mao, Yaqiu, Cai, Xu, Xu, Tingting, Zhao, Zhiyuan, Qian, Xiaohong, Qin, Weijie, Zhuang, Xiaomei, and Fan, Feng
- Subjects
- *
DRUG resistance in cancer cells , *DRUG design , *CANCER cell growth , *HEMATOLOGIC malignancies , *ORAL drug administration - Abstract
Background: Targeted protein degradation of neosubstrates plays a crucial role in hematological cancer treatment involving immunomodulatory imide drugs (IMiDs) therapy. Nevertheless, the persistence of inevitable drug resistance and hematological toxicities represents a significant obstacle to their clinical effectiveness. Methods: Phenotypic profiling of a small molecule compounds library in multiple hematological cancer cell lines was conducted to screen for hit degraders. Molecular dynamic-based rational design and cell-based functional assays were conducted to develop more potent degraders. Multiple myeloma (MM) tumor xenograft models were employed to investigate the antitumor efficacy of the degraders as single or combined agents with standard of care agents. Unbiased proteomics was employed to identify multiple therapeutically relevant neosubstrates targeted by the degraders. MM patient-derived cell lines (PDCs) and a panel of solid cancer cell lines were utilized to investigate the effects of candidate degrader on different stage of MM cells and solid malignancies. Unbiased proteomics of IMiDs-resistant MM cells, cell-based functional assays and RT-PCR analysis of clinical MM specimens were utilized to explore the role of BRD9 associated with IMiDs resistance and MM progression. Results: We identified a novel cereblon (CRBN)-dependent lead degrader with phthalazinone scaffold, MGD-4, which induced the degradation of Ikaros proteins. We further developed a novel potent candidate, MGD-28, significantly inhibited the growth of hematological cancer cells and induced the degradation of IKZF1/2/3 and CK1α with nanomolar potency via a Cullin-CRBN dependent pathway. Oral administration of MGD-4 and MGD-28 effectively inhibited MM tumor growth and exhibited significant synergistic effects with standard of care agents. MGD-28 exhibited preferentially profound cytotoxicity towards MM PDCs at different disease stages and broad antiproliferative activity in multiple solid malignancies. BRD9 modulated IMiDs resistance, and the expression of BRD9 was significant positively correlated with IKZF1/2/3 and CK1α in MM specimens at different stages. We also observed pronounced synergetic efficacy between the BRD9 inhibitor and MGD-28 for MM treatment. Conclusions: Our findings present a strategy for the multi-targeted degradation of Ikaros proteins and CK1α against hematological cancers, which may be expanded to additional targets and indications. This strategy may enhance efficacy treatment against multiple hematological cancers and solid tumors. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
7. Safranal's Neurological Nexus: Unveiling Therapeutic Potential through Molecular Docking.
- Author
-
Muhammad Wasim, Zia, Syeda Rehana, and Ahmad, Saara
- Abstract
Safranal, a bioactive compound derived from saffron, has emerged as a subject of growing interest in the quest for novel therapeutics targeting neurological disorders. This research article explores safranal's neurological nexus by employing molecular docking techniques to elucidate its potential as a therapeutic agent. As several neurological disorders constitute a significant global health challenge, necessitating innovative approaches for effective treatment. Safranal, with its recognized neuroprotective, antioxidant, and anti-inflammatory properties, offers a promising avenue for intervention. To unravel its mechanisms of action, molecular docking studies have become invaluable tools. In this comprehensive investigation, an in-depth analysis of safranal's neuropharmacological properties and its interactions with critical molecular targets implicated in neurological disorders has been investigated. Through molecular docking simulations, specific binding affinities and modes of safranal with key receptors, enzymes, and signaling pathways in the central nervous system have been explored. Our study delves into safranal's potential applications in addressing neurodegenerative diseases, including Alzheimer's, Parkinson's, epilepsy, autism as well as its impact on mood disorders such as depression and anxiety. The findings presented herein underscore safranal's remarkable therapeutic potential within the realm of neurology, shedding light on its molecular interactions with pivotal targets. Safranal's neuroprotective attributes, as unveiled through molecular docking, offer novel prospects for addressing a spectrum of neurological disorders, warranting further exploration through animal modeling, preclinical and clinical investigations. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
8. cPEPmatch Webserver: A comprehensive tool and database to aid rational design of cyclic peptides for drug discovery
- Author
-
Santini, Brianda L., Wendel, Stephanie, Halbwedl, Niklas, Knipp, Asha, and Zacharias, Martin
- Published
- 2024
- Full Text
- View/download PDF
9. Discovery of low‐molecular‐weight phenylalanine derivatives as novel HIV capsid modulators with improved antiretroviral activity and metabolic stability.
- Author
-
Jiang, Xiangyi, Gao, Zhen, Sharma, Prem Prakash, Kumar, Sumit, Rathi, Brijesh, Ji, Xiangkai, Dai, Jiaojiao, Xie, Minghui, Dong, Guanyu, Xu, Shujing, De Clercq, Erik, Pannecouque, Christophe, Dick, Alexej, Zhan, Peng, and Liu, Xinyong
- Subjects
PHENYLALANINE ,SURFACE plasmon resonance ,HIV ,MOLECULAR dynamics ,MOLECULAR docking - Abstract
The HIV capsid (CA) protein is a promising target for anti‐AIDS treatment due to its critical involvement in viral replication. Herein, we utilized the well‐documented CA inhibitor PF74 as our lead compound and designed a series of low‐molecular‐weight phenylalanine derivatives. Among them, compound 7t exhibited remarkable antiviral activity with a high selection index (EC50 = 0.040 µM, SI = 2815), surpassing that of PF74 (EC50 = 0.50 µM, SI = 258). Furthermore, when evaluated against the HIV‐2 strain, 7t (EC50 = 0.13 µM) demonstrated approximately 14‐fold higher potency than that of PF74 (EC50 = 1.76 µM). Insights obtained from surface plasmon resonance (SPR) revealed that 7t exhibited stronger target affinity to the CA hexamer and monomer in comparison to PF74. The potential interactions between 7t and the HIV‐1 CA were further elucidated using molecular docking and molecular dynamics simulations, providing a plausible explanation for the enhanced target affinity with 7t over PF74. Moreover, the metabolic stability assay demonstrated that 7t (T1/2 = 77.0 min) significantly outperforms PF74 (T1/2 = 0.7 min) in human liver microsome, exhibiting an improvement factor of 110‐fold. In conclusion, 7t emerges as a promising drug candidate warranting further investigation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
10. Psychedelic innovations and the crisis of psychopharmacology.
- Author
-
Langlitz, Nicolas
- Subjects
- *
PSYCHOPHARMACOLOGY , *MENTAL illness treatment , *PSYCHOTHERAPY , *HALLUCINOGENIC drugs , *DRUG development , *PHARMACOGENOMICS - Abstract
In the 2010s, psychopharmacological research and development experienced a crisis: since no genuinely new drugs for the treatment of mental illness had been successfully developed for decades, major pharmaceutical corporations decided to disinvest their neuropsychopharmacology departments. At the same time, however, one branch of psychopharmacology began to boom. The FDA declared psychedelic-assisted psychotherapy a breakthrough therapy and hundreds of start-up companies began to compete for this potentially emerging health care market. The article looks at the case of psychedelic research to examine three different responses to the innovation crisis in psychopharmacology: (1) the resumption of pharmacopsychotherapy as a half-century old but previously marginalized and discontinued practice; (2) the continuation of self-experimentation as a simultaneously repressed and revitalized method of drug development; (3) computational drug design as a cutting-edge approach currently used to create non-psychedelic psychedelics that reduce psychiatric symptoms without any mind-altering effects. These responses point to conflicting imaginaries of innovation that envisage the future of psychopharmacology and thereby provide different diagnoses of its current predicament. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
11. Innovations in Antifungal Drug Discovery among Cell Envelope Synthesis Enzymes through Structural Insights.
- Author
-
Zhou, Yue and Reynolds, Todd B.
- Subjects
- *
DRUG discovery , *DRUG design , *ANTIFUNGAL agents , *FUNGAL cell walls , *ENZYMES , *CELL membranes , *DRUG resistance , *MYCOSES - Abstract
Life-threatening systemic fungal infections occur in immunocompromised patients at an alarming rate. Current antifungal therapies face challenges like drug resistance and patient toxicity, emphasizing the need for new treatments. Membrane-bound enzymes account for a large proportion of current and potential antifungal targets, especially ones that contribute to cell wall and cell membrane biosynthesis. Moreover, structural biology has led to a better understanding of the mechanisms by which these enzymes synthesize their products, as well as the mechanism of action for some antifungals. This review summarizes the structures of several current and potential membrane-bound antifungal targets involved in cell wall and cell membrane biosynthesis and their interactions with known inhibitors or drugs. The proposed mechanisms of action for some molecules, gleaned from detailed inhibitor–protein studeis, are also described, which aids in further rational drug design. Furthermore, some potential membrane-bound antifungal targets with known inhibitors that lack solved structures are discussed, as these might be good enzymes for future structure interrogation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
12. Targeting disease with benzoxazoles: a comprehensive review of recent developments.
- Author
-
Abdullahi, Abdulrahman and Yeong, Keng Yoon
- Abstract
Benzoxazole is an attractive scaffold in medicinal chemistry due to its diverse biological applications. From 2016 to 2023, a plethora of benzoxazole derivatives have been synthesized and evaluated for their pharmacological activities but a review on this topic was found lacking. This review thus aims to fill the gap and discusses the pharmacological activities of the synthesized derivatives, emphasizing their interactions with key biological targets implicated in diseases such as cancer, diabetes, pain, inflammation, and cardiovascular disorders. Relevant works were selected mainly from databases such as PubMed and Google Scholar to offer a comprehensive overview of derivatives with promising bioactivities. The limitations pertinent to some derivatives, such as poor in vitro or in vivo activities, were highlighted, while their prospects in drug discovery was discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
13. In-silico guided chemical exploration of KDM4A fragments hits
- Author
-
Jessica Lombino, Rosario Vallone, Maura Cimino, Maria Rita Gulotta, Giada De Simone, Maria Agnese Morando, Raffaele Sabbatella, Simona Di Martino, Mario Fogazza, Federica Sarno, Claudia Coronnello, Maria De Rosa, Chiara Cipollina, Lucia Altucci, Ugo Perricone, and Caterina Alfano
- Subjects
Epigenetic regulation ,Lysine demethylases ,KDM4 inhibitors ,Fragment-based drug discovery (FBDD) ,KDM4A ,Rational drug design ,Medicine ,Genetics ,QH426-470 - Abstract
Abstract Background Lysine demethylase enzymes (KDMs) are an emerging class of therapeutic targets, that catalyse the removal of methyl marks from histone lysine residues regulating chromatin structure and gene expression. KDM4A isoform plays an important role in the epigenetic dysregulation in various cancers and is linked to aggressive disease and poor clinical outcomes. Despite several efforts, the KDM4 family lacks successful specific molecular inhibitors. Results Herein, starting from a structure-based fragments virtual screening campaign we developed a synergic framework as a guide to rationally design efficient KDM4A inhibitors. Commercial libraries were used to create a fragments collection and perform a virtual screening campaign combining docking and pharmacophore approaches. The most promising compounds were tested in-vitro by a Homogeneous Time-Resolved Fluorescence-based assay developed for identifying selective substrate-competitive inhibitors by means of inhibition of H3K9me3 peptide demethylation. 2-(methylcarbamoyl)isonicotinic acid was identified as a preliminary active fragment, displaying inhibition of KDM4A enzymatic activity. Its chemical exploration was deeply investigated by computational and experimental approaches which allowed a rational fragment growing process. The in-silico studies guided the development of derivatives designed as expansion of the primary fragment hit and provided further knowledge on the structure–activity relationship. Conclusions Our study describes useful insights into key ligand-KDM4A protein interaction and provides structural features for the development of successful selective KDM4A inhibitors.
- Published
- 2023
- Full Text
- View/download PDF
14. Development of Novel ROCK Inhibitors via 3D-QSAR and Molecular Docking Studies: A Framework for Multi-Target Drug Design
- Author
-
Milan Beljkas, Milos Petkovic, Ana Vuletic, Ana Djuric, Juan Francisco Santibanez, Tatjana Srdic-Rajic, Katarina Nikolic, and Slavica Oljacic
- Subjects
Rho-associated protein kinases ,multitarget-directed ligands ,rational drug design ,molecular modeling ,3D-QSAR ,antineoplastic agents ,Pharmacy and materia medica ,RS1-441 - Abstract
Background/Objectives: Alterations in the actin cytoskeleton correlates to tumor progression and affect critical cellular processes such as adhesion, migration and invasion. Rho-associated coiled-coil-containing protein kinases (ROCK1 and ROCK2), important regulators of the actin cytoskeleton, are frequently overexpressed in various malignancies. The aim of this study was therefore to identify the key structural features of ROCK1/ROCK2 inhibitors using computer-aided drug design (CADD) approaches. In addition, new developed ROCK inhibitors provided a significant framework for the development of multitarget therapeutics—ROCK/HDAC (histone deacetylases) multitarget inhibitors. Methods: 3D-QSAR (Quantitative structure-activity relationship study) and molecular docking study were employed in order to identify key structural features that positively correlate with ROCK inhibition. MDA-MB-231, HCC1937, Panc-1 and Mia PaCa-2 cells were used for evaluation of anticancer properties of synthesized compounds. Results: C-19 showed potent anti-cancer properties, especially enhancement of apoptosis and cell cycle modulation in pancreatic cancer cell lines. In addition, C-19 and C-22 showed potent anti-migratory and anti-invasive effects comparable to the well-known ROCK inhibitor fasudil. Conclusions: In light of the results of this study, we propose a novel multi-target approach focusing on developing dual HDAC/ROCK inhibitors based on the structure of both C-19 and C-22, exploiting the synergistic potential of these two signaling pathways to improve therapeutic efficacy in metastatic tumors. Our results emphasize the potential of multi-target ROCK inhibitors as a basis for future cancer therapies.
- Published
- 2024
- Full Text
- View/download PDF
15. In-silico guided chemical exploration of KDM4A fragments hits.
- Author
-
Lombino, Jessica, Vallone, Rosario, Cimino, Maura, Gulotta, Maria Rita, De Simone, Giada, Morando, Maria Agnese, Sabbatella, Raffaele, Di Martino, Simona, Fogazza, Mario, Sarno, Federica, Coronnello, Claudia, De Rosa, Maria, Cipollina, Chiara, Altucci, Lucia, Perricone, Ugo, and Alfano, Caterina
- Subjects
- *
ISONICOTINIC acid , *PEPTIDES , *DRUG design , *STRUCTURE-activity relationships , *PROTEIN-protein interactions - Abstract
Background: Lysine demethylase enzymes (KDMs) are an emerging class of therapeutic targets, that catalyse the removal of methyl marks from histone lysine residues regulating chromatin structure and gene expression. KDM4A isoform plays an important role in the epigenetic dysregulation in various cancers and is linked to aggressive disease and poor clinical outcomes. Despite several efforts, the KDM4 family lacks successful specific molecular inhibitors. Results: Herein, starting from a structure-based fragments virtual screening campaign we developed a synergic framework as a guide to rationally design efficient KDM4A inhibitors. Commercial libraries were used to create a fragments collection and perform a virtual screening campaign combining docking and pharmacophore approaches. The most promising compounds were tested in-vitro by a Homogeneous Time-Resolved Fluorescence-based assay developed for identifying selective substrate-competitive inhibitors by means of inhibition of H3K9me3 peptide demethylation. 2-(methylcarbamoyl)isonicotinic acid was identified as a preliminary active fragment, displaying inhibition of KDM4A enzymatic activity. Its chemical exploration was deeply investigated by computational and experimental approaches which allowed a rational fragment growing process. The in-silico studies guided the development of derivatives designed as expansion of the primary fragment hit and provided further knowledge on the structure–activity relationship. Conclusions: Our study describes useful insights into key ligand-KDM4A protein interaction and provides structural features for the development of successful selective KDM4A inhibitors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
16. Unlocking therapeutic potential of trigonelline through molecular docking as a promising approach for treating diverse neurological disorders.
- Author
-
Zia, Syeda Rehana, Wasim, Muhammad, and Ahmad, Saara
- Subjects
- *
MOLECULAR docking , *NEUROLOGICAL disorders , *DRUG design , *ALZHEIMER'S disease , *PROTEIN-ligand interactions - Abstract
Neurological disorders pose significant challenges in terms of treatment options, necessitating the exploration of novel therapeutic approaches. Trigonelline, a naturally occurring alkaloid found in various plants, has emerged as a potential treatment option. It has also been reported that trigonelline is involved in several pathways like; Oxidative Stress and Antioxidant, Inflammatory, Neuroprotection and Neurotrophic, Mitochondrial Function and Energy Metabolism. This study aims to investigate the therapeutic potential of trigonelline for diverse neurological disorders using a molecular docking approach. Molecular docking simulations were performed to predict the binding affinity and interaction between trigonelline and target proteins implicated in neurological disorders. The structural requirements for effective binding were also explored. The molecular docking results revealed strong binding interactions and favorable binding affinities between trigonelline and the target proteins involved in diverse neurological disorders like Alzheimer's disease, Parkinson's disease, epilepsy, and depression etc. The predicted binding modes provided insights into the key molecular interactions governing the ligand-protein complexes. The findings suggest that trigonelline holds promise as a therapeutic approach for several neurological disorders. The molecular docking approach employed in this study provides a valuable tool for rational drug design and optimization of trigonelline-based compounds. Further experimental validation and preclinical studies are warranted to confirm the efficacy and safety of trigonelline as a potential treatment option, paving the way for the development of more effective and targeted therapies for neurological disorders. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
17. Feature Selection Investigation in Machine Learning Docking Scoring Functions
- Author
-
Balboni, Maurício Dorneles Caldeira, Arrua, Oscar Emilio, Werhli, Adriano V., dos Santos Machado, Karina, Goos, Gerhard, Founding Editor, Hartmanis, Juris, Founding Editor, Bertino, Elisa, Editorial Board Member, Gao, Wen, Editorial Board Member, Steffen, Bernhard, Editorial Board Member, Yung, Moti, Editorial Board Member, Reis, Marcelo S., editor, and de Melo-Minardi, Raquel C., editor
- Published
- 2023
- Full Text
- View/download PDF
18. Computer‐aided design of muscarinic acetylcholine receptor M3 inhibitors: Promising compounds among trifluoromethyl containing hexahydropyrimidinones/thiones.
- Author
-
Nyporko, Alex, Tsymbalyuk, Olga, Voiteshenko, Ivan, Starosyla, Sergiy, Protopopov, Mykola, and Bdzhola, Volodymyr
- Subjects
MUSCARINIC acetylcholine receptors ,CHOLINERGIC receptors ,TRIFLUOROMETHYL compounds ,COMPUTER-aided design ,ADRENERGIC receptors ,THIONES - Abstract
The new high selective mAChRs M3 inhibitors with IC50 in nanomolecular ranges, which can be the prototypes for effective COPD and asthma treatment drugs, were discovered with computational approaches among trifluoromethyl containing hexahydropyrimidinones/thiones. Compounds [6‐(4‐ethoxy‐3‐methoxy‐phenyl)‐4‐hydroxy‐2‐thioxo‐4‐(trifluoromethyl)hexahydropyrimidin‐5‐yl]‐phenyl‐methanone (THPT‐1) and 5‐benzoyl‐6‐(3,4‐dimethoxyphenyl)‐4‐hydroxy‐4‐(trifluoromethyl)hexahydropyrimidin‐2‐one (THPO‐4) have been proved to be a highly effective (with IC50 values of 1.62 ⋅ 10−7 M and 3.09 ⋅ 10−9 M, respectively) at the same concentrations significantly competitive inhibit the signal conduction through mAChR3 in comparison with ipratropium bromide, without significant effect on mAChR2, nicotinic cholinergic and adrenergic receptors. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
19. Development of PI3Kγ selective inhibitors: the strategies and application
- Author
-
Gu, Dong-yan, Zhang, Meng-meng, Li, Jia, Zhou, Yu-bo, and Sheng, Rong
- Published
- 2024
- Full Text
- View/download PDF
20. Docking studies of some pyrazole containing compounds in the cyclooxygenase-2 active site
- Author
-
Savić Jelena, Antonijević Marija, Crevar Milkica, and Brborić Jasmina
- Subjects
cyclooxygenase-2 inhibitors ,molecular interactions ,rational drug design ,protein-ligand interactions ,b-hydroxy-b-arylpropanoic acids ,Pharmacy and materia medica ,RS1-441 - Abstract
Whereas nonselective nonsteroidal anti-inflammatory drugs, such as aspirin, ibuprofen and diclofenac, inhibit both cyclooxygenase-1 and cyclooxigenase-2 enzymes, selective inhibitors target cyclooxygenase-2, which is overexpressed in inflammation, but also in cancer, atherosclerosis, Alzheimer's disease, and Parkinson's disease. Potential cardiovascular and hepatic side effects of cyclooxygenase-2 inhibitors have limited their use. The development of selective and safe cyclooxygenase-2 inhibitors remains a high priority in drug discovery. Based on the structure of previously investigated newly synthesized b-hydroxy-b-arylpropanoic acids, two groups of compounds were designed: analogs in which one of the benzene rings was replaced by a pyrazole, while the carboxyl group was retained, and amides of b-hydroxy-b-arylpropanoic acids with pyrazole. The compounds were docked into the 3D structure of the catalytic site of the enzyme cyclooxygenase-2 using AutoDock Vina 1.2.0. and the obtained interactions were compared with the interactions of celecoxib, a selective inhibitor. The amides had lower binding energies than the designed acids, which makes them attractive target compounds for synthesis and further examination.
- Published
- 2023
21. SARS-CoV-2 Main Protease Inhibitors: Structure-Based Enhancement to Anti-Viral Pre-Clinical GC376 Encourages Further Development.
- Author
-
Perry, Elliot D, Chapman, Simon, and Xu, Yao-Zhong
- Subjects
- *
PROTEASE inhibitors , *SARS-CoV-2 , *BINDING energy , *DRUG design , *VIRAL replication - Abstract
SARS-CoV-2 Main protease (Mpro) is pivotal in viral replication and transcription. Mpro mediates proteolysis of translated products of replicase genes ORF1a and ORF1ab. Surveying pre-clinical trial Mpro inhibitors suggests potential enhanced efficacy for some moieties. Concordant with promising in vitro and in silico data, the protease inhibitor GC376 was chosen as a lead. Modification of GC376 analogues yielded a series of promising Mpro inhibitors. Design optimization identified compound G59i as lead candidate, displaying a binding energy of − 10.54 kcal/mol for the complex. Robust interactivity was noted between G59i and Mpro. With commendable ADMET characteristics and enhanced potency, further G59i analysis may be advantageous; moreover, identified key Mpro residues could contribute to the design of neotenic inhibitors. Serial modifications to the SARS-2-CoV main protease (Mpro) inhibitor GC376 yielded promising analogues from which compound G59i was progressed. A more exothermic binding energy was predicted for Mpro ligation compared to GC376. With commendable ADMET characteristics and enhanced potency, further G59i analysis may be advantageous. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
22. NFX1‐123: A potential therapeutic target in cervical cancer.
- Author
-
Chintala, Sreenivasulu, Dankoski, Maura A., Anbarasu, Anand, Ramaiah, Sudha, Miryala, Sravan Kumar, and Katzenellenbogen, Rachel A.
- Subjects
CERVICAL cancer ,CELL migration inhibition ,HEAD & neck cancer ,CELL growth ,STRUCTURAL frame models ,DRUG design - Abstract
NFX1‐123 is a splice variant isoform of the NFX1 gene. It is highly expressed in cervical cancers caused by HPV, and NFX1‐123 is a protein partner with the HPV oncoprotein E6. Together, NFX1‐123 and E6 affect cellular growth, longevity, and differentiation. The expression status of NFX1‐123 in cancers beyond cervical and head and neck cancers, and its potential as therapeutic target, have not been investigated. TSVdb of TCGA was used to quantify NFX1‐123 expression in 24 cancers compared with normal tissues. The NFX1‐123 protein structure was predicted and then submitted to retrieve suitable drug molecules. The top four compounds, found to bind in silico to NFX1‐123, were tested experimentally to determine their effects on NFX1‐123‐related cellular growth, survival, and migration. 46% of cancers (11 of 24 had significant differences in NFX1‐123 expression, with nine having had greater NFX1‐123 expression, when compared with adjacent normal tissues. Bioinformatics and proteomic predictive analysis modeled the three‐dimensional structure of NFX1‐123, and drug libraries were screened for high‐binding affinity compounds using this modeled structure. Seventeen drugs with binding energies ranging from −1.3 to −10 Kcal/mol were identified. The top four compounds were used to treat HPV‐ and HPV+ cervical cancer cell lines, three of which (Ropitoin, R428 and Ketoconazole) reduced NFX1‐123 protein levels, inhibited cellular growth, survival, and migration, and enhanced the cytotoxicity of Cisplatin. These findings highlight cancers expressing high levels of NFX1‐123, and drugs that target it, may reduce cellular growth, survival, and migration, making NFX1‐123 a potential novel therapeutic target. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
23. Validation of catalytic site residues of Ubiquitin Specific Protease 2 (USP2) by molecular dynamic simulation and novel kinetics assay for rational drug design.
- Author
-
Ullah, Shafi, Junaid, Muhammad, Liu, Yunqi, Chen, Shiyao, Zhao, Yaxue, and Wadood, Abdul
- Abstract
Post-translational modifications of proteins such as protein ubiquitination are crucial for regulating conformation, stability and localization of the modified protein. Ubiquitin-specific protease 2 (USP2), a multifunctional cysteine protease is reported to be a key regulator of ubiquitylation events in numerous oncogenic proteins e.g., fatty acid synthetase, Mdm2, EGFR, cyclin A1, and cyclin-D1, etc. Thus targeting USP2 is a promising strategy for cancer therapy. USP2 is characterized by a catalytic triad comprising of cysteine, histidine and aspartic acid residues. Five residues including three from the catalytic triad and two from outside of the catalytic triad have been reported as a catalytic site of USP2 that catalyze hydrolysis and stabilizes the oxyanion formed in the intermediate step of catalysis. Here, we report two more novel residues (L269 and Y558) on USP2 involved in the catalysis of Ubiquitin using computational alanine scanning (CAS) followed by molecular dynamic simulation studies. The results obtained from CAS were further validated by a highly reliable, time- and cost-effective SDS-PAGE-based kinetics assay using UBA52 which is a natural substrate of USP2. Our results showed that mutating L269 and Y558 significantly compromised the catalytic efficiency of USP2 in hydrolyzing UBA52 which can further be extended to rational drug design of USP2 selective inhibitors and to explore the catalytic sites of other USPs. Two novel residues take part in catalytic activity of USP2 which were depicted by MD Simulations and were further validated by novel SDS-PAGE-based reliable time- and cost-effective kinetics assay. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
24. Differential chemoproteomic analysis of RRS-1 candidate molecule and molecules of several nonsteroidal anti-inflammatory drugs
- Author
-
P. A. Galenko-Yaroshevsky, I. Yu. Torshin, A. N. Gromov, O. A. Gromova, K. F. Suzdalev, R. A. Murashko, A. V. Zelenskaya, A. V. Zadorozhniy, T. R. Glechyan, G. V. Simavonyan, and E. M.I. Muhammad
- Subjects
inflammation ,rational drug design ,rrs-1 ,machine learning ,Therapeutics. Pharmacology ,RM1-950 ,Economics as a science ,HB71-74 - Abstract
Background. To plan effective and safe pharmacotherapy for inflammation and pain, it is important to evaluate the mechanisms and spectrum of action of nonsteroidal anti-inflammatory drugs (NSAIDs), including their effects on human proteome.Objective: to identify and evaluate the most significant specific differences of candidate molecule RRS-1 (N-{(Z)-2-(1-methyl-1H-indol-3-yl)-1-[(propylamino)carbonyl]vinyl}benzamide) from other NSAIDs through differential chemoreactome analysis.Materials and methods. Chemoproteomic modeling of pharmacological effects of RRS-1 molecule and a number of well-known NSAIDs (diclofenac, nimesulide, ketorolac) on human proteome was carried out on the basis of numerical prediction algorithms over the space of heterogeneous feature descriptions, developed in the topological approach to recognition by Yu.I. Zhuravlev and K.V. Rudakov scientific school.Results. Significant differences in the effects of the studied molecules were found for 1232 proteins of human proteome. The features of assessing interactions of the studied molecules with 47 target proteins, which most distinguished the effects of RRS-1 molecule from all others were identified. RRS-1 could activate adenosine and dopamine receptors, cannabinoid receptor 2 and GABAA receptor to a greater extent than other molecules. Activation of these receptors corresponded to anti-inflammatory, anti-nociceptive and neuroprotective effects. RRS-1 could preferably inhibit a number of pro-inflammatory proteins, receptor bradykinin 1, metabotropic glutamate receptor 5, matrix metalloproteinases 8, 9, 12, and blood coagulation factor X. Additionally, RRS-1 molecule showed preferable inhibition of a number of kinases targeted in antitumor and anti-inflammatory therapy. RRS-1, less than other studied molecules, interacted with the receptors of vitamin D3, thyroid hormone, acetylcholine, cannabinoids and opioids, orexin, and various metabolic enzymes, which is important in assessment of the safety of using drugs based on this molecule. RRS-1 characteristically exhibited a moderate profile of antivitamin action: the total score of vitamin and mineral loss (7.4±3.7) was significantly less in comparison to diclofenac (11.7±4.5) and was actually on the same level as nimesulide (6.9±3.7) and ketorolac (6.7±3.6).Conclusion. Chemoreactomic and chemoproteomic profiling of RRS-1 candidate molecule provided pre-experimental assessments of its efficacy and safety through modeling interactions with the human proteome.
- Published
- 2023
- Full Text
- View/download PDF
25. Innovations in Antifungal Drug Discovery among Cell Envelope Synthesis Enzymes through Structural Insights
- Author
-
Yue Zhou and Todd B. Reynolds
- Subjects
cryo-EM ,structure biology ,membrane-bound enzymes ,antifungal development ,drug resistance ,rational drug design ,Biology (General) ,QH301-705.5 - Abstract
Life-threatening systemic fungal infections occur in immunocompromised patients at an alarming rate. Current antifungal therapies face challenges like drug resistance and patient toxicity, emphasizing the need for new treatments. Membrane-bound enzymes account for a large proportion of current and potential antifungal targets, especially ones that contribute to cell wall and cell membrane biosynthesis. Moreover, structural biology has led to a better understanding of the mechanisms by which these enzymes synthesize their products, as well as the mechanism of action for some antifungals. This review summarizes the structures of several current and potential membrane-bound antifungal targets involved in cell wall and cell membrane biosynthesis and their interactions with known inhibitors or drugs. The proposed mechanisms of action for some molecules, gleaned from detailed inhibitor–protein studeis, are also described, which aids in further rational drug design. Furthermore, some potential membrane-bound antifungal targets with known inhibitors that lack solved structures are discussed, as these might be good enzymes for future structure interrogation.
- Published
- 2024
- Full Text
- View/download PDF
26. Computational Methods for Peptide Macrocycle Drug Design
- Author
-
Mulligan, Vikram Khipple, Perrie, Yvonne, Series Editor, and Jois, Seetharama D., editor
- Published
- 2022
- Full Text
- View/download PDF
27. Design, synthesis, and in silico study of hybrid oxoazetidine conjugated thiazoles as anti‐EGFR with cytotoxicity activity.
- Author
-
Pathak, Prateek, Gupta, Santosh, Grishina, Maria, Khalilullah, Habibullah, and Verma, Amita
- Subjects
EPIDERMAL growth factor receptors ,THIAZOLES - Abstract
We report a series of hybrid oxoazetidine conjugated thiazoles as epidermal growth factor receptor (EGFR) inhibitors, which were synthesized and tested using a variety of in silico and in vitro studies. The compounds were found to be active against breast and hepatic cancer cell lines, with Compounds 7a, 7b, and 7e being the most potent ones. The derivatives were also evaluated for molecular docking and complementarity studies to explicate fundamental substituent groups essential for their bioactivity. Moreover, the structural activity relationship of the analogues was performed for future compound optimization. These studies advocated that the analogues have a high affinity towards EGFR with favorable anticancer potential. The study advised that the derivatives have potency against breast and hepatic cancer and can assist as an initial scaffold for further development of anti‐EGFR compounds. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
28. Drug repurposing for viral cancers: A paradigm of machine learning, deep learning, and virtual screening‐based approaches.
- Author
-
Ahmed, Faheem, Kang, In Suk, Kim, Kyung Hwan, Asif, Arun, Rahim, Chethikkattuveli Salih Abdul, Samantasinghar, Anupama, Memon, Fida Hussain, and Choi, Kyung Hyun
- Subjects
DRUG repositioning ,DEEP learning ,MACHINE learning ,DRUG discovery ,DRUG design ,SIMULATED patients - Abstract
Cancer management is major concern of health organizations and viral cancers account for approximately 15.4% of all known human cancers. Due to large number of patients, efficient treatments for viral cancers are needed. De novo drug discovery is time consuming and expensive process with high failure rate in clinical stages. To address this problem and provide treatments to patients suffering from viral cancers faster, drug repurposing emerges as an effective alternative which aims to find the other indications of the Food and Drug Administration approved drugs. Applied to viral cancers, drug repurposing studies following the niche have tried to find if already existing drugs could be used to treat viral cancers. Multiple drug repurposing approaches till date have been introduced with successful results in viral cancers and many drugs have been successfully repurposed various viral cancers. Here in this study, a critical review of viral cancer related databases, tools, and different machine learning, deep learning and virtual screening‐based drug repurposing studies focusing on viral cancers is provided. Additionally, the mechanism of viral cancers is presented along with drug repurposing case study specific to each viral cancer. Finally, the limitations and challenges of various approaches along with possible solutions are provided. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
29. Lessons Learnt from COVID-19: Computational Strategies for Facing Present and Future Pandemics.
- Author
-
Pavan, Matteo and Moro, Stefano
- Subjects
- *
PANDEMICS , *COVID-19 , *MOLECULAR dynamics , *DRUG discovery , *SARS-CoV-2 - Abstract
Since its outbreak in December 2019, the COVID-19 pandemic has caused the death of more than 6.5 million people around the world. The high transmissibility of its causative agent, the SARS-CoV-2 virus, coupled with its potentially lethal outcome, provoked a profound global economic and social crisis. The urgency of finding suitable pharmacological tools to tame the pandemic shed light on the ever-increasing importance of computer simulations in rationalizing and speeding up the design of new drugs, further stressing the need for developing quick and reliable methods to identify novel active molecules and characterize their mechanism of action. In the present work, we aim at providing the reader with a general overview of the COVID-19 pandemic, discussing the hallmarks in its management, from the initial attempts at drug repurposing to the commercialization of Paxlovid, the first orally available COVID-19 drug. Furthermore, we analyze and discuss the role of computer-aided drug discovery (CADD) techniques, especially those that fall in the structure-based drug design (SBDD) category, in facing present and future pandemics, by showcasing several successful examples of drug discovery campaigns where commonly used methods such as docking and molecular dynamics have been employed in the rational design of effective therapeutic entities against COVID-19. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
30. Discovery of novel fused-heterocycle-bearing diarypyrimidine derivatives as HIV-1 potent NNRTIs targeting tolerant region I for enhanced antiviral activity and resistance profile.
- Author
-
Dai, Jiaojiao, Jiang, Xiangyi, Gao, Heng, Huang, Boshi, De Clercq, Erik, Pannecouque, Christophe, Du, Shaoqing, Liu, Xinyong, and Zhan, Peng
- Subjects
- *
NON-nucleoside reverse transcriptase inhibitors , *DRUG design , *MOLECULAR dynamics , *ANTI-HIV agents , *ENZYME-linked immunosorbent assay , *ANTIVIRAL agents - Abstract
As an important part of anti-AIDS therapy, HIV-1 non-nucleoside reverse transcriptase inhibitors are plagued by resistance and toxicity issues. Taking our reported XJ-18b1 as lead compound, we designed a series of novel diarypyrimidine derivatives by employing a scaffold hopping strategy to discover potent NNRTIs with improved anti-resistance properties and drug-like profiles. The most active compound 3k exhibited prominent inhibitory activity against wild-type HIV-1 (EC 50 = 0.0019 μM) and common mutant strains including K103 N (EC 50 = 0.0019 μM), L100I (EC 50 = 0.0087 μM), E138K (EC 50 = 0.011 μM), along with low cytotoxicity and high selectivity index (CC 50 = 21.95 μM, SI = 11478). Additionally, compound 3k demonstrated antiviral activity against HIV-2 with EC 50 value of 6.14 μM. The enzyme-linked immunosorbent assay validated that 3k could significantly inhibit the activity of HIV-1 reverse transcriptase (IC 50 = 0.025 μM). Furthermore, molecular dynamics simulation studies were performed to illustrate the potential binding mode and binding free energy of the RT- 3k complex, and in silico prediction revealed that 3k possessed favorable drug-like profiles. Collectively, 3k proved to be a promising lead compound for further optimization to obtain anti-HIV drug candidates. [Display omitted] • A series of novel diarypyrimidine derivatives were synthesized targeting the tolerant region I of the NNIBP using scaffold hopping strategy. • Pyridinotriazole compound 3k exhibited promising activity against the wild-type and mutant strains of HIV-1. • The molecular dynamics simulation studies revealed the binding mode of 3k in the binding pocket. • The preliminary structure-activity relationships were summarized in detail. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
31. Exploring the landscape of post-translational modification in drug discovery.
- Author
-
Cao, Yuhao, Yu, Tianyi, Zhu, Ziang, Zhang, Yuanjiao, Sun, Shanliang, Li, Nianguang, Gu, Chunyan, and Yang, Ye
- Subjects
- *
DRUG design , *SMALL molecules , *POST-translational modification , *DRUG discovery , *BIOCHEMICAL substrates - Abstract
Post-translational modifications (PTMs) play a crucial role in regulating protein function, and their dysregulation is frequently associated with various diseases. The emergence of epigenetic drugs targeting factors such as histone deacetylases (HDACs) and histone methyltransferase enhancers of zeste homolog 2 (EZH2) has led to a significant shift towards precision medicine, offering new possibilities to overcome the limitations of traditional therapeutics. In this review, we aim to systematically explore how small molecules modulate PTMs. We discuss the direct targeting of enzymes involved in PTM pathways, the modulation of substrate proteins, and the disruption of protein-enzyme interactions that govern PTM processes. Additionally, we delve into the emerging strategy of employing multifunctional molecules to precisely regulate the modification levels of proteins of interest (POIs). Furthermore, we examine the specific characteristics of these molecules, evaluating their therapeutic benefits and potential drawbacks. The goal of this review is to provide a comprehensive understanding of PTM-targeting strategies and their potential for personalized medicine, offering a forward-looking perspective on the evolution of precision therapeutics. [ABSTRACT FROM AUTHOR]
- Published
- 2025
- Full Text
- View/download PDF
32. Identification of novel diarylpyrimidine derivatives as potent HIV-1 non-nucleoside reverse transcriptase inhibitors against wild-type and K103N mutant viruses.
- Author
-
Jiang, Xiangyi, Zalloum, Waleed A., Gao, Zhen, Dai, Jiaojiao, Ji, Xiangkai, Xie, Minghui, Dong, Guanyu, De Clercq, Erik, Huang, Boshi, Pannecouque, Christophe, Zhan, Peng, and Liu, Xinyong
- Subjects
- *
NON-nucleoside reverse transcriptase inhibitors , *DRUG design , *ANTI-HIV agents , *MOLECULAR dynamics , *NUCLEOSIDE derivatives - Abstract
HIV-1 non-nucleoside reverse transcriptase inhibitors (NNRTIs) play a crucial role in combination antiretroviral therapy (cART). To further enhance their antiviral activity and anti-resistance properties, we developed a series of novel NNRTIs, by specifically targeting tolerant region I of the NNRTI binding pocket. Among them, compound 9t-2 displayed excellent anti-HIV-1 potency against wild-type and prevalent mutant strains with EC 50 values between 0.0019 and 0.012 μM. This outperformed the positive drugs ETR, NVP and RPV. Aslo, ELISA results confirmed that these compounds can effectively inhibit the activity of HIV-1 RT. Molecular dynamics (MD) simulation studies indicated that the thiomorpholine-1,1-dioxide moiety of 9t-2 is capable of establishing additional interactions with residues P225, F227 and P236 in the tolerant region I, which contributed to its enhanced activity. Compound 9t-2 possessed negligible inhibitory effect on the five main CYP isoenzymes (IC 50 > 10 μM), indicating a low potential for inducing CYP-mediated drug-drug interactions. In conclusion, compound 9t-2 , with its enhanced anti-resistance properties, stands out as a promising lead compound for further optimization towards discovering the new generation of anti-HIV agents. A series of novel diarylpyrimidine derivatives were identified as potent HIV-1 NNRTIs with improved anti-resistance properties by exploiting the tolerant region I of NNIBP. [Display omitted] • Identification of novel potent HIV-1 NNRTIs by exploring solvent-exposed region. • 9t-2 showed significant enhanced anti-HIV-1 activities compared to etravirine. • Molecular dynamics simulation revealed the binding mode between 9t-2 and HIV-1 RT. • The likelihood of 9t-2 causing CYP-mediated drug interactions is low. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
33. Exploring the anticancer properties of Azadirachta indica: In silico and in vitro study of its phytochemicals against hepatocellular carcinoma.
- Author
-
Khalid, Hina, Shityakov, Sergey, Förster, Carola Y., and Song, Yuanda
- Subjects
- *
DRUG design , *MOLECULAR dynamics , *HEPATOCELLULAR carcinoma , *CYTOTOXINS , *PROTEIN kinases - Abstract
• Azadirachta indica's methanol extract demonstrates significant cytotoxicity against HepG2 cells, with an IC₅₀ value of 81.17 µg/ml. • In silico analysis identifies two potent Akt1 inhibitors, 2′,3′-dehydrosalannol and quercetin, showcasing improved binding affinity and hepatoprotective effects. • Molecular dynamics simulations, particularly the MM-GBSA model, provide detailed insights into the inhibitory potential of docked bioconstituents, aiding in rational drug design for hepatocellular carcinoma (HCC). • The study proposes an innovative in silico pipeline for early-stage identification of lead candidates against HCC, offering a promising approach for future drug development and clinical trials. The medicinal plant Azadirachta indica (Neem) interferes with carcinogenesis and acts as a tumor suppressor. This study evaluated the anticancer effects of a methanol extract of Azadirachta Indica on a human hepatocellular carcinoma cell line (HepG2) using an in vitro MTT assay. The results demonstrated that the methanol extract of Azadirachta Indica exhibited significant cytotoxicity against HepG2 cells, with an IC₅₀ value of 81.17 µg/ml. Furthermore, an in silico approach was used to investigate the interaction between Azadirachta indica and Akt1, a protein kinase implicated in the pathogenesis of hepatocellular carcinoma (HCC) via the PI3K/Akt/mTOR pathway. This study identified two promising Akt1 inhibitors, 2′,3′-dehydrosalannol and quercetin, with improved binding affinity (ΔGbind) and a safe pharmacological profile. These compounds also showed hepatoprotective effects. Molecular dynamics simulations, specifically the MM-GBSA model, were used to further scrutinize the inhibitory potential of the docked bioconstituents. The results suggest that the proposed in silico pipeline can be useful for identifying lead candidates for HCC through early-stage rational drug design. These candidates can then be verified in vitro and in vivo before being tested in clinical trials as mainstream targets against HCC. [Display omitted] [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
34. Molecular Modeling Insights into the Structure and Behavior of Integrins: A Review.
- Author
-
Tvaroška, Igor, Kozmon, Stanislav, and Kóňa, Juraj
- Subjects
- *
INTEGRINS , *DRUG design , *SYNAPTOGENESIS , *GLYCOPROTEINS - Abstract
Integrins are heterodimeric glycoproteins crucial to the physiology and pathology of many biological functions. As adhesion molecules, they mediate immune cell trafficking, migration, and immunological synapse formation during inflammation and cancer. The recognition of the vital roles of integrins in various diseases revealed their therapeutic potential. Despite the great effort in the last thirty years, up to now, only seven integrin-based drugs have entered the market. Recent progress in deciphering integrin functions, signaling, and interactions with ligands, along with advancement in rational drug design strategies, provide an opportunity to exploit their therapeutic potential and discover novel agents. This review will discuss the molecular modeling methods used in determining integrins' dynamic properties and in providing information toward understanding their properties and function at the atomic level. Then, we will survey the relevant contributions and the current understanding of integrin structure, activation, the binding of essential ligands, and the role of molecular modeling methods in the rational design of antagonists. We will emphasize the role played by molecular modeling methods in progress in these areas and the designing of integrin antagonists. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
35. Molecular Recognition of FDA-Approved Small Molecule Protein Kinase Drugs in Protein Kinases.
- Author
-
Zhu, Yan and Hu, Xiche
- Subjects
- *
PROTEIN kinases , *MOLECULAR recognition , *SMALL molecules , *PROTEIN drugs , *PROTEIN kinase inhibitors , *DRUG discovery , *ADENOSINE triphosphate , *ARAMID fibers - Abstract
Protein kinases are key enzymes that catalyze the covalent phosphorylation of substrates via the transfer of the γ-phosphate of ATP, playing a crucial role in cellular proliferation, differentiation, and various cell regulatory processes. Due to their pivotal cellular role, the aberrant function of kinases has been associated with cancers and many other diseases. Consequently, competitive inhibition of the ATP binding site of protein kinases has emerged as an effective means of curing these diseases. Decades of intense development of protein kinase inhibitors (PKIs) resulted in 71 FDA-approved PKI drugs that target dozens of protein kinases for the treatment of various diseases. How do FDA-approved protein kinase inhibitor PKI drugs compete with ATP in their own binding pocket? This is the central question we attempt to address in this work. Based on modes of non-bonded interactions and their calculated interaction strengths by means of the advanced double hybrid DFT method B2PLYP, the molecular recognition of PKI drugs in the ATP-binding pockets was systematically analyzed. It was found that (1) all the FDA-approved PKI drugs studied here form one or more hydrogen bond(s) with the backbone amide N, O atoms in the hinge region of the ATP binding site, mimicking the adenine base; (2) all the FDA-approved PKI drugs feature two or more aromatic rings. The latter reach far and deep into the hydrophobic regions I and II, forming multiple CH-π interactions with aliphatic residues L(3), V(11), A(15), V(36), G(51), L(77) and π-π stacking interactions with aromatic residues F(47) and F(82), but ATP itself does not utilize these regions extensively; (3) all FDA-approved PKI drugs studied here have one thing in common, i.e., they frequently formed non-bonded interactions with a total of 12 residues L(3),V(11), A(15), K(17), E(24),V(36),T(45), F(47), G(51), L(77), D(81) and F(82) in the ATP binding. Many of those 12 commonly involved residues are highly conserved residues with important structural and catalytic functional roles. K(17) and E(24) are the two highly conserved residues crucial for the catalytic function of kinases. D(81) and F(82) belong to the DFG motif; T(45) was dubbed the gate keeper residue. F(47) is located on the hinge region and G(51) sits on the linker that connects the hinge to the αD-helix. It is this targeting of highly conserved residues in protein kinases that led to promiscuous PKI drugs that lack selectivity. Although the formation of hydrogen bond(s) with the backbone of the hinge gives PKI drugs the added binding affinity and the much-needed directionality, selectivity is sacrificed. That is why so many FDA-approved PKI drugs are known to have multiple targets. Moreover, off-target-mediated toxicity caused by a lack of selectivity was one of the major challenges facing the PKI drug discovery community. This work suggests a road map for future PKI drug design, i.e., targeting non-conserved residues in the ATP binding pocket to gain better selectivity so as to avoid off-target-mediated toxicity. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
36. Rational Design of a Novel Tubulin Inhibitor with a Unique Mechanism of Action.
- Author
-
Mühlethaler, Tobias, Milanos, Lampros, Ortega, Jose Antonio, Blum, Thorsten B., Gioia, Dario, Roy, Bibhas, Prota, Andrea E., Cavalli, Andrea, and Steinmetz, Michel O.
- Abstract
In this study, we capitalized on our previously performed crystallographic fragment screen and developed the antitubulin small molecule Todalam with only two rounds of straightforward chemical synthesis. Todalam binds to a novel tubulin site, disrupts microtubule networks in cells, arrests cells in G2/M, induces cell death, and synergizes with vinblastine. The compound destabilizes microtubules by acting as a molecular plug that sterically inhibits the curved‐to‐straight conformational switch in the α‐tubulin subunit, and by sequestering tubulin dimers into assembly incompetent oligomers. Our results describe for the first time the generation of a fully rationally designed small molecule tubulin inhibitor from a fragment, which displays a unique molecular mechanism of action. They thus demonstrate the usefulness of tubulin‐binding fragments as valuable starting points for innovative antitubulin drug and chemical probe discovery campaigns. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
37. Applications of density functional theory in COVID-19 drug modeling.
- Author
-
Ye, Naike, Yang, Zekai, and Liu, Yuchen
- Subjects
- *
SARS-CoV-2 , *DENSITY functional theory , *COVID-19 - Abstract
• Rapidly evolving COVID-19 pandemic demands the discovery of effective antiviral drugs. • DFT is extensively applied in molecular modeling studies of COVID-19 pharmaceuticals. • DFT is used as a stand-alone method for electronic structure calculations of drug compounds. • Molecular mechanics techniques and DFT are applied in synergy to examine drug-target interactions. • Hybrid QM/MM approach accurately elucidates catalytic/inhibitory reaction mechanisms. The rapidly evolving Coronavirus 2019 (COVID-19) pandemic has led to millions of deaths around the world, highlighting the pressing need to develop effective antiviral pharmaceuticals. Recent efforts with computer-aided rational drug discovery have allowed detailed examination of drug–macromolecule interactions primarily by molecular mechanics (MM) techniques. Less widely applied in COVID-19 drug modeling is density functional theory (DFT), a quantum mechanics (QM) method that enables electronic structure calculations and elucidations of reaction mechanisms. Here, we review recent advances in applying DFT in molecular modeling studies of COVID-19 pharmaceuticals. We start by providing an overview of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) drugs and targets, followed by a brief introduction to DFT. We then provide a discussion of different approaches by which DFT has been applied. Finally, we discuss essential factors to consider when incorporating DFT in future drug modeling research. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
38. Novel colchicine derivative CR42-24 demonstrates potent anti-tumor activity in urothelial carcinoma.
- Author
-
Bell, Clayton J., Potts, Kyle G., Hitt, Mary M., Pink, Desmond, Tuszynski, Jack A., and Lewis, John D.
- Subjects
- *
BLADDER cancer , *TRANSITIONAL cell carcinoma , *ANTINEOPLASTIC agents , *COLCHICINE , *DRUG design , *TUBULINS - Abstract
Bladder cancers, and specifically urothelial carcinoma, have few effective treatment options, and tumors typically develop resistance against standard of care chemotherapies leading to significant mortality. The development of alternative therapies with increased selectivity and improved tolerability would significantly impact this patient population. Here, we investigate a novel colchicine derivative, CR42-24, with increased selectivity for the βIII tubulin subtype as a treatment for urothelial carcinoma. βIII tubulin is a promising target due to its low expression in healthy tissues and its clinical association with poor prognosis. This study demonstrated that CR42-24 is selectively cytotoxic to several cancer cell lines at low nanomolar IC50, with high activity in bladder cancer cell lines both in vitro and in vivo. CR42-24 monotherapy in an aggressive urothelial carcinoma xenograft model results in effective control when treated early. We observed significant ablation of large tumors and patient-derived xenografts at low doses with excellent tolerability. CR42-24 was highly synergistic in combination with the standard of care chemotherapies gemcitabine and cisplatin, further increasing its therapeutic potential as a novel treatment for urothelial carcinoma. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
39. Mechanistic physicochemical insights into glycation and drug binding by serum albumin: Implications in diabetic conditions.
- Author
-
Ghosh, Ritutama and Kishore, Nand
- Subjects
- *
SERUM albumin , *METFORMIN , *ADVANCED glycation end-products , *DRUG design , *RECEPTOR for advanced glycation end products (RAGE) , *MASS spectrometry - Abstract
The drug binding ability of serum albumin might get affected as a result of its glycation under diabetic conditions. It requires not only an understanding of the effect of glycation of the protein upon association with the drug, but also calls for an assessment of structure-property-energetics relationships. A combination of ultrasensitive calorimetric, spectroscopic and chromatographic approach has been employed to correlate thermodynamic signatures with recognition, conformation and mechanistic details of the processes involved. An important observation from this work is that 3-(dansylamino) phenyl boronic acid (DnsPBA) assay cannot always determine the extent of glycation as evidenced by MALDI-TOF mass spectra of glycated HSA due to its selectivity for 1,2 or 1,3 cis -diol structures which may be absent in certain AGEs. Protein gets modified post glycation with the formation of advanced glycation end products (AGEs), which are monitored to be targeted by the guanidine group present in anti-diabetic drugs. AGEs formed in the third and fourth week of glycation are significant in the recognition of anti-diabetic drugs. The results with metformin and aminoguanidine suggest that the extent of binding depends upon the number of guanidine group(s) in the drug molecule. Open chain molecules having guanidine group(s) exhibit stronger affinity towards glycated HSA than closed ring entities like naphthalene or pyridine moiety. The observation that the drug binding ability of HSA is not adversely affected, rather strengthened upon glycation, has implications in diabetic conditions. A rigorous structure-property-energetics correlation based on thermodynamic signatures and identification of functional groups on drugs for recognition by HSA are essential in deriving guidelines for rational drug design addressing diabetes. [Display omitted] • Open chain entities with guanidine groups target AGEs formed upon glycation of HSA. • Glycation strengthens drug transport ability of HSA having implications in diabetes. • Structure-property-energetics relations identified important functionality in binding. • Identification of functional groups has direct implications in rational drug design. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
40. Multiple Poses and Thermodynamics of Ligands Targeting Protein Surfaces: The Case of Furosemide Binding to mitoNEET in Aqueous Solution
- Author
-
Linh Gia Hoang, Jonas Goßen, Riccardo Capelli, Toan T. Nguyen, Zhaoxi Sun, Ke Zuo, Jörg B. Schulz, Giulia Rossetti, and Paolo Carloni
- Subjects
NEET proteins ,rational drug design ,localized volume-based metadynamics ,furosemide binding pose and affinity ,furosemide ,molecular dynamics ,Biology (General) ,QH301-705.5 - Abstract
Human NEET proteins, such as NAF-1 and mitoNEET, are homodimeric, redox iron-sulfur proteins characterized by triple cysteine and one histidine-coordinated [2Fe-2S] cluster. They exist in an oxidized and reduced state. Abnormal release of the cluster is implicated in a variety of diseases, including cancer and neurodegeneration. The computer-aided and structure-based design of ligands affecting cluster release is of paramount importance from a pharmaceutical perspective. Unfortunately, experimental structural information so far is limited to only one ligand/protein complex. This is the X-ray structure of furosemide bound to oxidized mitoNEET. Here we employ an enhanced sampling approach, Localized Volume-based Metadynamics, developed by some of us, to identify binding poses of furosemide to human mitoNEET protein in solution. The binding modes show a high variability within the same shallow binding pocket on the protein surface identified in the X-ray structure. Among the different binding conformations, one of them is in agreement with the crystal structure’s one. This conformation might have been overstabilized in the latter because of the presence of crystal packing interactions, absent in solution. The calculated binding affinity is compatible with experimental data. Our protocol can be used in a straightforward manner in drug design campaigns targeting this pharmaceutically important family of proteins.
- Published
- 2022
- Full Text
- View/download PDF
41. Allostery in Drug Development
- Author
-
Cheng, Xi, Jiang, Hualiang, LAMBRIS, JOHN D., Series Editor, REZAEI, NIMA, Series Editor, Zhang, Jian, editor, and Nussinov, Ruth, editor
- Published
- 2019
- Full Text
- View/download PDF
42. Isothermal Titration Calorimetry: A Powerful Tool for the Characterization of Molecular Interactions
- Author
-
Thanassoulas, Angelos, Nounesis, George, Demetzos, Costas, editor, and Pippa, Natassa, editor
- Published
- 2019
- Full Text
- View/download PDF
43. Modular and Flexible Tangible Molecular Interface for Interactive Molecular Simulation Based on Internet of Things Approach
- Author
-
Vincke, Bastien, Ghaoui, Mohamed Anis, Férey, Nicolas, Martinez, Xavier, Brochot, Loïc, Goos, Gerhard, Founding Editor, Hartmanis, Juris, Founding Editor, Bertino, Elisa, Editorial Board Member, Gao, Wen, Editorial Board Member, Steffen, Bernhard, Editorial Board Member, Woeginger, Gerhard, Editorial Board Member, Yung, Moti, Editorial Board Member, Bourdot, Patrick, editor, Interrante, Victoria, editor, Nedel, Luciana, editor, Magnenat-Thalmann, Nadia, editor, and Zachmann, Gabriel, editor
- Published
- 2019
- Full Text
- View/download PDF
44. Overcoming challenges of HERG potassium channel liability through rational design: Eag1 inhibitors for cancer treatment.
- Author
-
Toplak, Žan, Hendrickx, Louise A., Abdelaziz, Reham, Shi, Xiaoyi, Peigneur, Steve, Tomašič, Tihomir, Tytgat, Jan, Peterlin‐Mašič, Lucija, and Pardo, Luis A.
- Subjects
POTASSIUM channels ,DRUG design ,ION channels ,POTASSIUM antagonists ,CANCER treatment ,VOLTAGE-gated ion channels ,TUMOR growth - Abstract
Two decades of research have proven the relevance of ion channel expression for tumor progression in virtually every indication, and it has become clear that inhibition of specific ion channels will eventually become part of the oncology therapeutic arsenal. However, ion channels play relevant roles in all aspects of physiology, and specificity for the tumor tissue remains a challenge to avoid undesired effects. Eag1 (KV10.1) is a voltage‐gated potassium channel whose expression is very restricted in healthy tissues outside of the brain, while it is overexpressed in 70% of human tumors. Inhibition of Eag1 reduces tumor growth, but the search for potent inhibitors for tumor therapy suffers from the structural similarities with the cardiac HERG channel, a major off‐target. Existing inhibitors show low specificity between the two channels, and screenings for Eag1 binders are prone to enrichment in compounds that also bind HERG. Rational drug design requires knowledge of the structure of the target and the understanding of structure–function relationships. Recent studies have shown subtle structural differences between Eag1 and HERG channels with profound functional impact. Thus, although both targets' structure is likely too similar to identify leads that exclusively bind to one of the channels, the structural information combined with the new knowledge of the functional relevance of particular residues or areas suggests the possibility of selective targeting of Eag1 in cancer therapies. Further development of selective Eag1 inhibitors can lead to first‐in‐class compounds for the treatment of different cancers. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
45. Future direction of agrochemical development for plant disease in China.
- Author
-
Guo, Shengxin, He, Feng, Song, Baoan, and Wu, Jian
- Subjects
- *
DRUG design , *PLANT development , *ASYMMETRIC synthesis , *PESTICIDE resistance , *NATURAL products - Abstract
The growth of the global population poses a huge challenge to food security. The continuous application of crop protection products has many negative effects including increased pesticide resistance, accumulation of residues, and ecological damage. The challenge for the future is to develop agrochemicals with that are effective, with a high degree of selectivity, environmental friendliness, a low use rate, and cost‐effectiveness. Cutting‐edge developments in natural product chemistry, asymmetric synthesis, and rational drug design, as well as an increasing knowledge of the mechanisms that induce plant immunity, have provided potential solutions. In this review, we discuss the comprehensive use of emerging technologies that will help to tackle these current challenges and issues, highlighting the discovery of novel crop production products and their beneficial properties. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
46. The Fe-S cluster-containing NEET proteins mitoNEET and NAF-1 as chemotherapeutic targets in breast cancer
- Author
-
Bai, Fang, Morcos, Faruck, Sohn, Yang-Sung, Darash-Yahana, Merav, Rezende, Celso O, Lipper, Colin H, Paddock, Mark L, Song, Luhua, Luo, Yuting, Holt, Sarah H, Tamir, Sagi, Theodorakis, Emmanuel A, Jennings, Patricia A, Onuchic, José N, Mittler, Ron, and Nechushtai, Rachel
- Subjects
Cancer ,Breast Cancer ,5.1 Pharmaceuticals ,Development of treatments and therapeutic interventions ,Antineoplastic Agents ,Breast Neoplasms ,Cell Line ,Tumor ,Cell Proliferation ,Cell Survival ,Cluster Analysis ,Drug Design ,Female ,Humans ,Iron-Sulfur Proteins ,MCF-7 Cells ,Mitochondrial Proteins ,Molecular Conformation ,Molecular Docking Simulation ,Molecular Targeted Therapy ,Ribonucleoproteins ,Software ,Xanthones ,NEET proteins ,mitocan ,iron-sulfur proteins ,mitochondria ,rational drug design - Abstract
Identification of novel drug targets and chemotherapeutic agents is a high priority in the fight against cancer. Here, we report that MAD-28, a designed cluvenone (CLV) derivative, binds to and destabilizes two members of a unique class of mitochondrial and endoplasmic reticulum (ER) 2Fe-2S proteins, mitoNEET (mNT) and nutrient-deprivation autophagy factor-1 (NAF-1), recently implicated in cancer cell proliferation. Docking analysis of MAD-28 to mNT/NAF-1 revealed that in contrast to CLV, which formed a hydrogen bond network that stabilized the 2Fe-2S clusters of these proteins, MAD-28 broke the coordinative bond between the His ligand and the cluster's Fe of mNT/NAF-1. Analysis of MAD-28 performed with control (Michigan Cancer Foundation; MCF-10A) and malignant (M.D. Anderson-metastatic breast; MDA-MB-231 or MCF-7) human epithelial breast cells revealed that MAD-28 had a high specificity in the selective killing of cancer cells, without any apparent effects on normal breast cells. MAD-28 was found to target the mitochondria of cancer cells and displayed a surprising similarity in its effects to the effects of mNT/NAF-1 shRNA suppression in cancer cells, causing a decrease in respiration and mitochondrial membrane potential, as well as an increase in mitochondrial iron content and glycolysis. As expected, if the NEET proteins are targets of MAD-28, cancer cells with suppressed levels of NAF-1 or mNT were less susceptible to the drug. Taken together, our results suggest that NEET proteins are a novel class of drug targets in the chemotherapeutic treatment of breast cancer, and that MAD-28 can now be used as a template for rational drug design for NEET Fe-S cluster-destabilizing anticancer drugs.
- Published
- 2015
47. Future direction of agrochemical development for plant disease in China
- Author
-
Shengxin Guo, Feng He, Baoan Song, and Jian Wu
- Subjects
asymmetric synthesis ,green agrochemicals ,natural products ,perspectives ,plant immune inducer ,rational drug design ,Agriculture ,Agriculture (General) ,S1-972 - Abstract
Abstract The growth of the global population poses a huge challenge to food security. The continuous application of crop protection products has many negative effects including increased pesticide resistance, accumulation of residues, and ecological damage. The challenge for the future is to develop agrochemicals with that are effective, with a high degree of selectivity, environmental friendliness, a low use rate, and cost‐effectiveness. Cutting‐edge developments in natural product chemistry, asymmetric synthesis, and rational drug design, as well as an increasing knowledge of the mechanisms that induce plant immunity, have provided potential solutions. In this review, we discuss the comprehensive use of emerging technologies that will help to tackle these current challenges and issues, highlighting the discovery of novel crop production products and their beneficial properties.
- Published
- 2021
- Full Text
- View/download PDF
48. Applications of artificial intelligence to drug design and discovery in the big data era: a comprehensive review.
- Author
-
Tripathi, Neetu, Goshisht, Manoj Kumar, Sahu, Sanat Kumar, and Arora, Charu
- Abstract
Artificial intelligence (AI) renders cutting-edge applications in diverse sectors of society. Due to substantial progress in high-performance computing, the development of superior algorithms, and the accumulation of huge biological and chemical data, computer-assisted drug design technology is playing a key role in drug discovery with its advantages of high efficiency, fast speed, and low cost. Over recent years, due to continuous progress in machine learning (ML) algorithms, AI has been extensively employed in various drug discovery stages. Very recently, drug design and discovery have entered the big data era. ML algorithms have progressively developed into a deep learning technique with potent generalization capability and more effectual big data handling, which further promotes the integration of AI technology and computer-assisted drug discovery technology, hence accelerating the design and discovery of the newest drugs. This review mainly summarizes the application progression of AI technology in the drug discovery process, and explores and compares its advantages over conventional methods. The challenges and limitations of AI in drug design and discovery have also been discussed. [ABSTRACT FROM AUTHOR]
- Published
- 2021
- Full Text
- View/download PDF
49. Structural insight into the recognition of S-adenosyl-L-homocysteine and sinefungin in SARS-CoV-2 Nsp16/Nsp10 RNA cap 2′-O-Methyltransferase
- Author
-
Panupong Mahalapbutr, Napat Kongtaworn, and Thanyada Rungrotmongkol
- Subjects
COVID-19 ,SARS-CoV-2 ,Nsp16/nsp10 ,Nucleoside analog ,MD simulations ,Rational drug design ,Biotechnology ,TP248.13-248.65 - Abstract
The recent ongoing coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) continues to rapidly spread across the world. To date, neither a specific antiviral drug nor a clinically effective vaccine is available. Among the 15 viral non-structural proteins (nsps), nsp16 methyltransferase has been considered as a potential target due to its crucial role in RNA cap 2′-O-methylation process, preventing the virus detection by cell innate immunity mechanisms. In the present study, molecular recognition between the two natural nucleoside analogs (S-adenosyl-l-homocysteine (SAH) and sinefungin (SFG)) and the SARS-CoV-2 nsp16/nsp10/m7GpppAC5 was studied using all-atom molecular dynamics simulations and free energy calculations based on MM/GBSA and WaterSwap approaches. The binding affinity and the number of hot-spot residues, atomic contacts, and H-bond formations of SFG/nsp16 complex were distinctly higher than those of SAH/nsp16 system, consistent with the lower water accessibility at the enzyme active site. Notably, only SFG could electrostatically interact with the 2′-OH and N3 of RNA’s adenosine moiety, mimicking the methyl transfer reaction of S-adenosyl-l-methionine substrate. The atomistic binding mechanism obtained from this work paves the way for further optimizations and designs of more specific SARS-CoV-2 nsp16 inhibitors in the fight against COVID-19.
- Published
- 2020
- Full Text
- View/download PDF
50. Molecular Recognition of FDA-Approved Small Molecule Protein Kinase Drugs in Protein Kinases
- Author
-
Yan Zhu and Xiche Hu
- Subjects
FDA-approved drugs ,molecular recognition ,quantum chemical calculations ,π-π stacking interactions ,CH-π interactions ,rational drug design ,Organic chemistry ,QD241-441 - Abstract
Protein kinases are key enzymes that catalyze the covalent phosphorylation of substrates via the transfer of the γ-phosphate of ATP, playing a crucial role in cellular proliferation, differentiation, and various cell regulatory processes. Due to their pivotal cellular role, the aberrant function of kinases has been associated with cancers and many other diseases. Consequently, competitive inhibition of the ATP binding site of protein kinases has emerged as an effective means of curing these diseases. Decades of intense development of protein kinase inhibitors (PKIs) resulted in 71 FDA-approved PKI drugs that target dozens of protein kinases for the treatment of various diseases. How do FDA-approved protein kinase inhibitor PKI drugs compete with ATP in their own binding pocket? This is the central question we attempt to address in this work. Based on modes of non-bonded interactions and their calculated interaction strengths by means of the advanced double hybrid DFT method B2PLYP, the molecular recognition of PKI drugs in the ATP-binding pockets was systematically analyzed. It was found that (1) all the FDA-approved PKI drugs studied here form one or more hydrogen bond(s) with the backbone amide N, O atoms in the hinge region of the ATP binding site, mimicking the adenine base; (2) all the FDA-approved PKI drugs feature two or more aromatic rings. The latter reach far and deep into the hydrophobic regions I and II, forming multiple CH-π interactions with aliphatic residues L(3), V(11), A(15), V(36), G(51), L(77) and π-π stacking interactions with aromatic residues F(47) and F(82), but ATP itself does not utilize these regions extensively; (3) all FDA-approved PKI drugs studied here have one thing in common, i.e., they frequently formed non-bonded interactions with a total of 12 residues L(3),V(11), A(15), K(17), E(24),V(36),T(45), F(47), G(51), L(77), D(81) and F(82) in the ATP binding. Many of those 12 commonly involved residues are highly conserved residues with important structural and catalytic functional roles. K(17) and E(24) are the two highly conserved residues crucial for the catalytic function of kinases. D(81) and F(82) belong to the DFG motif; T(45) was dubbed the gate keeper residue. F(47) is located on the hinge region and G(51) sits on the linker that connects the hinge to the αD-helix. It is this targeting of highly conserved residues in protein kinases that led to promiscuous PKI drugs that lack selectivity. Although the formation of hydrogen bond(s) with the backbone of the hinge gives PKI drugs the added binding affinity and the much-needed directionality, selectivity is sacrificed. That is why so many FDA-approved PKI drugs are known to have multiple targets. Moreover, off-target-mediated toxicity caused by a lack of selectivity was one of the major challenges facing the PKI drug discovery community. This work suggests a road map for future PKI drug design, i.e., targeting non-conserved residues in the ATP binding pocket to gain better selectivity so as to avoid off-target-mediated toxicity.
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.