1
|
Yang M, Lee MK, Gao S, Song L, Jang HY, Jo I, Yang CC, Sylvester K, Ko C, Wang S, Ye B, Tang K, Li J, Gu M, Müller CE, Sträter N, Liu X, Kim M, Zhan P. Miniaturized Modular Click Chemistry-enabled Rapid Discovery of Unique SARS-CoV-2 M pro Inhibitors With Robust Potency and Drug-like Profile. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2404884. [PMID: 39319611 DOI: 10.1002/advs.202404884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/30/2024] [Indexed: 09/26/2024]
Abstract
The COVID-19 pandemic has required an expeditious advancement of innovative antiviral drugs. In this study, focused compound libraries are synthesized in 96- well plates utilizing modular click chemistry to rapidly discover potent inhibitors targeting the main protease (Mpro) of SARS-CoV-2. Subsequent direct biological screening identifies novel 1,2,3-triazole derivatives as robust Mpro inhibitors with high anti-SARS-CoV-2 activity. Notably, C5N17B demonstrates sub-micromolar Mpro inhibitory potency (IC50 = 0.12 µM) and excellent antiviral activity in Calu-3 cells determined in an immunofluorescence-based antiviral assay (EC50 = 0.078 µM, no cytotoxicity: CC50 > 100 µM). C5N17B shows superior potency to nirmatrelvir (EC50 = 1.95 µM) and similar efficacy to ensitrelvir (EC50 = 0.11 µM). Importantly, this compound displays high antiviral activities against several SARS-CoV-2 variants (Gamma, Delta, and Omicron, EC50 = 0.13 - 0.26 µM) and HCoV-OC43, indicating its broad-spectrum antiviral activity. It is worthy that C5N17B retains antiviral activity against nirmatrelvir-resistant strains with T21I/E166V and L50F/E166V mutations in Mpro (EC50 = 0.26 and 0.15 µM, respectively). Furthermore, C5N17B displays favorable pharmacokinetic properties. Crystallography studies reveal a unique, non-covalent multi-site binding mode. In conclusion, these findings substantiate the potential of C5N17B as an up-and-coming drug candidate targeting SARS-CoV-2 Mpro for clinical therapy.
Collapse
Affiliation(s)
- Mianling Yang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Myoung Kyu Lee
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Shenghua Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Letian Song
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Hye-Yeon Jang
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Inseong Jo
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Chun-Chiao Yang
- Institute of Bioanalytical Chemistry, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
| | - Katharina Sylvester
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany
| | - Chunkyu Ko
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Shuo Wang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Bing Ye
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Kai Tang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Junyi Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Manyu Gu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Christa E Müller
- PharmaCenter Bonn & Pharmaceutical Institute, Department of Pharmaceutical & Medicinal Chemistry, University of Bonn, An der Immenburg 4, 53113, Bonn, Germany
| | - Norbert Sträter
- Institute of Bioanalytical Chemistry, Leipzig University, Deutscher Platz 5, 04103, Leipzig, Germany
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| | - Meehyein Kim
- Infectious Diseases Therapeutic Research Center, Korea Research Institute of Chemical Technology (KRICT), Daejeon, 34114, Republic of Korea
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology, Ministry of Education, School of Pharmaceutical Sciences, Shandong University, Ji'nan, 250012, China
| |
Collapse
|
2
|
Cui S, Wang F, Yang W, Yu Y, Li Y. Protein-Templated Click Ligation Reaction Triggered by Protein-Split Aptamer Interactions. Anal Chem 2024. [PMID: 39264850 DOI: 10.1021/acs.analchem.4c03316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/14/2024]
Abstract
DNA-templated reactions have found wide applications in sensing and drug discovery. However, this strategy has been limited to the use of nucleic acids as templating elements to direct the proximity effect. Herein, we describe a versatile protein-templated split aptamer click ligation reaction (PT-SpA-CLR) in which the protein template-induced covalent proximity ligation of split aptamer elements enables translating protein/aptamer binding events into the output of ligated DNA products. A ligation yield of >80% is observed for three model protein templates, including VEGF165, PDGF-BB, and SARS-CoV-2 S1. The ligation reaction compensates for the weakness of reduced binding affinity resulting from splitting the aptamer, as evidenced by an approximately 2-fold lower dissociation constant than the non-ligated system. This newly developed PT-SpA-CLR strategy is further integrated with colorimetric or fluorescent reporting mechanisms to achieve easy-to-use and low-cost biosensors utilizing ligation to produce a fully active G-quadruplex or an RNA-cleaving DNAzyme to report protein binding. Both assays can achieve specific detection of an intended protein target with a limit of detection at the picomolar level even when challenged in biological samples. The combined PT-SpA-CLR and versatile sensing strategies offer attractive universal platforms for efficient detection of protein biomarkers.
Collapse
Affiliation(s)
- Susu Cui
- State Key Laboratory of Space Power-Sources, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Fan Wang
- State Key Laboratory of Space Power-Sources, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
| | - Weiwei Yang
- State Key Laboratory of Space Power-Sources, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, Harbin Institute of Technology, Harbin 150001, China
| | - Yongsheng Yu
- State Key Laboratory of Space Power-Sources, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin 150001, China
- MIIT Key Laboratory of Critical Materials Technology for New Energy Conversion and Storage, Harbin Institute of Technology, Harbin 150001, China
| | - Yingfu Li
- Department of Biochemistry and Biomedical Sciences, McMaster University, 1280 Main Street West, Hamilton, Ontario L8S 4K1, Canada
| |
Collapse
|
3
|
Ambala S, Thumma V, Mallikanti V, Bathini V, K J, Pochampally J. Synthesis of New Chroman-4-one Based 1,2,3-Triazole Analogues as Antioxidant and Anti-Inflammatory Agents. Chem Biodivers 2024; 21:e202400587. [PMID: 38718104 DOI: 10.1002/cbdv.202400587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/08/2024] [Indexed: 06/19/2024]
Abstract
A library of new chroman-4-one based 1,2,3-triazole analogues were synthesized involving a series of condensation, cyclization, Suzuki coupling and copper catalysed click chemistry protocols. The newly synthesized compounds 8a-l were screened for their invitro antioxidant and anti-inflammatory activities by employing Ascorbic acid and Diclofenac as reference drugs respectively. The compound without any substituent on benzyl ring (8a), compound with -Cl substituent in para position of benzyl ring (8i), and compound with ethoxy substituent in para position of benzyl ring (8k) exhibited potent antioxidant and anti-inflammatory activities with higher percentage of inhibition. To understand their binding affinities, molecular docking study of these three compounds performed against NADPH oxidase with presented outstanding docking scores and promising binding interactions like H-bond and hydrophobic.
Collapse
Affiliation(s)
- Shankaraiah Ambala
- Department of Chemistry, Osmania University, Hyderabad, 500007, Telangana, India
| | - Vishnu Thumma
- Department of Sciences and Humanities, Matrusri Engineering College, Hyderabad, 500059, Telangana, India
| | | | - Vineesha Bathini
- Department of Chemistry, Osmania University, Hyderabad, 500007, Telangana, India
| | - Jyothi K
- St. Marys College of Pharmacy, Secunderabad, Hyderabad, 500025, Telangana, India
| | | |
Collapse
|
4
|
Jyoti, Bhatt D, Kumar S, Maurya A, Pal A, Darokar MP, Bawankule DU, Tandon S. Cu-catalyzed click reaction in synthesis of eugenol derivatives as potent antimalarial agents. Nat Prod Res 2024:1-13. [PMID: 38828834 DOI: 10.1080/14786419.2024.2348670] [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: 10/16/2023] [Accepted: 04/20/2024] [Indexed: 06/05/2024]
Abstract
Eugenol(1), a terpenoid found in Ocimum, has various biological activities. The present study aims at extraction, isolation of the plant secondary metabolite eugenol (1), it's derivatisation and structure identification as bioactive molecules. Synthesis and antiplasmodial activity (in-vitro and in-vivo), of a series of fourteen novel eugenol-based 1,2,3-triazole derivatives was done in the present study. Derivatives 5a-5n showed good antimalarial activity against the strain Plasmodium falciparum NF54. Derivative 5 m, IC50 at 2.85 µM was found to be several times better than its precursor 1 (106.82 µM) whereas the derivative 5n showed three fold better activity than compound 1, in vitro. The structure-activity relationship of the synthesised compounds indicated that the presence of triazole ring in eugenol analogues is responsible for their good activity. Compound 5m, was further evaluated for in-vivo antimalarial activity which showed about 79% parasitemia suppression. It is the first report on antimalarial activity of triazole eugenol derivatives.
Collapse
Affiliation(s)
- Jyoti
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Divya Bhatt
- Bio-Prospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Saurabh Kumar
- Bio-Prospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Aransha Maurya
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Anirban Pal
- Bio-Prospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Mahendra P Darokar
- Bio-Prospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Dnyaneshwar U Bawankule
- Bio-Prospection and Product Development, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
| | - Sudeep Tandon
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants (CIMAP), Lucknow, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| |
Collapse
|
5
|
Zhou Y, Shen W, Gao Y, Peng J, Li Q, Wei X, Liu S, Lam FS, Mayol-Llinàs J, Zhao G, Li G, Li Y, Sun H, Cao Y, Li X. Protein-templated ligand discovery via the selection of DNA-encoded dynamic libraries. Nat Chem 2024; 16:543-555. [PMID: 38326646 DOI: 10.1038/s41557-024-01442-y] [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: 02/13/2023] [Accepted: 01/04/2024] [Indexed: 02/09/2024]
Abstract
DNA-encoded chemical libraries (DELs) have become a powerful technology platform in drug discovery. Dual-pharmacophore DELs display two sets of small molecules at the termini of DNA duplexes, thereby enabling the identification of synergistic binders against biological targets, and have been successfully applied in fragment-based ligand discovery and affinity maturation of known ligands. However, dual-pharmacophore DELs identify separate binders that require subsequent linking to obtain the full ligands, which is often challenging. Here we report a protein-templated DEL selection approach that can identify full ligand/inhibitor structures from DNA-encoded dynamic libraries (DEDLs) without the need for subsequent fragment linking. Our approach is based on dynamic DNA hybridization and target-templated in situ ligand synthesis, and it incorporates and encodes the linker structures in the library, along with the building blocks, to be sampled by the target protein. To demonstrate the performance of this method, 4.35-million- and 3.00-million-member DEDLs with different library architectures were prepared, and hit selection was achieved against four therapeutically relevant target proteins.
Collapse
Grants
- AoE/P-705/16, 17301118, 17111319, 17303220, 17300321, 17318322, C7005-20G, C7016-22G, and 2122-7S04 Research Grants Council, University Grants Committee (RGC, UGC)
- 21877093, 22222702, and 91953119 National Science Foundation of China | National Natural Science Foundation of China-Yunnan Joint Fund (NSFC-Yunnan Joint Fund)
- Health@InnoHK Innovation and Technology Commission (ITF)
Collapse
Affiliation(s)
- Yu Zhou
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Health@InnoHK, Innovation and Technology Commission, Hong Kong SAR, China
| | - Wenyin Shen
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Ying Gao
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Jianzhao Peng
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Qingrong Li
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Xueying Wei
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Shihao Liu
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Fong Sang Lam
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China
| | - Joan Mayol-Llinàs
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Health@InnoHK, Innovation and Technology Commission, Hong Kong SAR, China
| | - Guixian Zhao
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Gang Li
- Institute of Systems and Physical Biology, Shenzhen Bay Laboratory, Shenzhen, China
| | - Yizhou Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, School of Pharmaceutical Sciences; Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China
| | - Hongzhe Sun
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China.
| | - Yan Cao
- School of Pharmacy, Naval Medical University, Shanghai, China.
| | - Xiaoyu Li
- Department of Chemistry and State Key Laboratory of Synthetic Chemistry, The University of Hong Kong, Hong Kong SAR, China.
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Health@InnoHK, Innovation and Technology Commission, Hong Kong SAR, China.
| |
Collapse
|
6
|
Zhao R, Zhu J, Jiang X, Bai R. Click chemistry-aided drug discovery: A retrospective and prospective outlook. Eur J Med Chem 2024; 264:116037. [PMID: 38101038 DOI: 10.1016/j.ejmech.2023.116037] [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: 10/22/2023] [Revised: 11/20/2023] [Accepted: 12/08/2023] [Indexed: 12/17/2023]
Abstract
Click chemistry has emerged as a valuable tool for rapid compound synthesis, presenting notable advantages and convenience in the exploration of potential drug candidates. In particular, in situ click chemistry capitalizes on enzymes as reaction templates, leveraging their favorable conformation to selectively link individual building blocks and generate novel hits. This review comprehensively outlines and introduces the extensive use of click chemistry in compound library construction, and hit and lead discovery, supported by specific research examples. Additionally, it discusses the limitations and precautions associated with the application of click chemistry in drug discovery. Our intention for this review is to contribute to the development of a modular synthetic approach for the rapid identification of drug candidates.
Collapse
Affiliation(s)
- Rui Zhao
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Junlong Zhu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Xiaoying Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China
| | - Renren Bai
- School of Pharmacy, Hangzhou Normal University, Hangzhou, 311121, PR China; Key Laboratory of Elemene Class Anti-cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, 311121, PR China.
| |
Collapse
|
7
|
Kassu M, Parvatkar PT, Milanes J, Monaghan NP, Kim C, Dowgiallo M, Zhao Y, Asakawa AH, Huang L, Wagner A, Miller B, Carter K, Barrett KF, Tillery LM, Barrett LK, Phan IQ, Subramanian S, Myler PJ, Van Voorhis WC, Leahy JW, Rice CA, Kyle DE, Morris J, Manetsch R. Shotgun Kinetic Target-Guided Synthesis Approach Enables the Discovery of Small-Molecule Inhibitors against Pathogenic Free-Living Amoeba Glucokinases. ACS Infect Dis 2023; 9:2190-2201. [PMID: 37820055 PMCID: PMC10644346 DOI: 10.1021/acsinfecdis.3c00284] [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: 06/20/2023] [Indexed: 10/13/2023]
Abstract
Pathogenic free-living amoebae (pFLA) can cause life-threatening central nervous system (CNS) infections and warrant the investigation of new chemical agents to combat the rise of infection from these pathogens. Naegleria fowleri glucokinase (NfGlck), a key metabolic enzyme involved in generating glucose-6-phosphate, was previously identified as a potential target due to its limited sequence similarity with human Glck (HsGlck). Herein, we used our previously demonstrated multifragment kinetic target-guided synthesis (KTGS) screening strategy to identify inhibitors against pFLA glucokinases. Unlike the majority of previous KTGS reports, our current study implements a "shotgun" approach, where fragments were not biased by predetermined binding potentials. The study resulted in the identification of 12 inhibitors against 3 pFLA glucokinase enzymes─NfGlck, Balamuthia mandrillaris Glck (BmGlck), and Acanthamoeba castellanii Glck (AcGlck). This work demonstrates the utility of KTGS to identify small-molecule binders for biological targets where resolved X-ray crystal structures are not readily accessible.
Collapse
Affiliation(s)
- Mintesinot Kassu
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Prakash T. Parvatkar
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Jillian Milanes
- Eukaryotic
Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Neil P. Monaghan
- Eukaryotic
Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Chungsik Kim
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Matthew Dowgiallo
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Yingzhao Zhao
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Ami H. Asakawa
- Department
of Pharmaceutical Sciences, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Lili Huang
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Alicia Wagner
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Brandon Miller
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Karissa Carter
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Kayleigh F. Barrett
- Center
for Emerging and Re-emerging Infectious Diseases (CERID), Division
of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109, United States
| | - Logan M. Tillery
- Center
for Emerging and Re-emerging Infectious Diseases (CERID), Division
of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109, United States
| | - Lynn K. Barrett
- Center
for Emerging and Re-emerging Infectious Diseases (CERID), Division
of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109, United States
| | - Isabelle Q. Phan
- Center for Global Infectious Diseases Research, Seattle Children’s Research Center, Seattle, Washington 98109, United States
| | - Sandhya Subramanian
- Center for Global Infectious Diseases Research, Seattle Children’s Research Center, Seattle, Washington 98109, United States
| | - Peter J. Myler
- Center for Global Infectious Diseases Research, Seattle Children’s Research Center, Seattle, Washington 98109, United States
| | - Wesley C. Van Voorhis
- Center
for Emerging and Re-emerging Infectious Diseases (CERID), Division
of Allergy and Infectious Diseases, Department of Medicine, University of Washington School of Medicine, Seattle, Washington 98109, United States
| | - James W. Leahy
- Department of Chemistry, University
of
South Florida, Tampa, Florida 33620, United States
| | - Christopher A. Rice
- Department
of Comparative Pathobiology, College of Veterinary Medicine, Purdue University, West Lafayette, Indiana 47907, United States
- Purdue
Institute for Drug Discovery (PIDD), Purdue
University, West Lafayette, Indiana 47907, United States
- Purdue Institute
of Inflammation, Immunology and Infectious Disease (PI4D), Purdue University, West Lafayette, Indiana 47907, United States
- Department
of Cellular Biology, University of Georgia, Athens, Georgia 30602, United States
| | - Dennis E. Kyle
- Department
of Cellular Biology, University of Georgia, Athens, Georgia 30602, United States
| | - James Morris
- Eukaryotic
Pathogens Innovation Center, Department of Genetics and Biochemistry, Clemson University, Clemson, South Carolina 29634, United States
| | - Roman Manetsch
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
- Department
of Pharmaceutical Sciences, Northeastern
University, Boston, Massachusetts 02115, United States
- Center
for Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
- Barnett
Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Liu P, Jiang Y, Jiao L, Luo Y, Wang X, Yang T. Strategies for the Discovery of Oxazolidinone Antibacterial Agents: Development and Future Perspectives. J Med Chem 2023; 66:13860-13873. [PMID: 37807849 DOI: 10.1021/acs.jmedchem.3c01040] [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: 10/10/2023]
Abstract
Oxazolidinones represent a significant class of synthetic bacterial protein synthesis inhibitors that are primarily effective against Gram-positive bacteria. The commercial success of linezolid, the first FDA-approved oxazolidinone antibiotic, has motivated researchers to develop more potent oxazolidinones by employing various drug development strategies to fight against antimicrobial resistance, some of which have shown promising results. Thus, this Perspective aims to discuss the strategies employed in constructing oxazolidinone-based antibacterial agents and summarize recent advances in discovering oxazolidinone antibiotics to provide valuable insights for potentially developing next-generation oxazolidinone antibacterial agents or other pharmaceuticals.
Collapse
Affiliation(s)
- Pingxian Liu
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yunhan Jiang
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Jiao
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Youfu Luo
- State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaodong Wang
- Department of Breast Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Tao Yang
- Center of Infectious Diseases and Laboratory of Human Diseases and Immunotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Institute of Immunology and Inflammation, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
9
|
Grenier D, Audebert S, Preto J, Guichou JF, Krimm I. Linkers in fragment-based drug design: an overview of the literature. Expert Opin Drug Discov 2023; 18:987-1009. [PMID: 37466331 DOI: 10.1080/17460441.2023.2234285] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/05/2023] [Indexed: 07/20/2023]
Abstract
INTRODUCTION In fragment-based drug design, fragment linking is a popular strategy where two fragments binding to different sub-pockets of a target are linked together. This attractive method remains challenging especially due to the design of ideal linkers. AREAS COVERED The authors review the types of linkers and chemical reactions commonly used to the synthesis of linkers, including those utilized in protein-templated fragment self-assembly, where fragments are directly linked in the presence of the protein. Finally, they detail computational workflows and software including generative models that have been developed for fragment linking. EXPERT OPINION The authors believe that fragment linking offers key advantages for compound design, particularly for the design of bivalent inhibitors linking two distinct pockets of the same or different subunits. On the other hand, more studies are needed to increase the potential of protein-templated approaches in FBDD. Important computational tools such as structure-based de novo software are emerging to select suitable linkers. Fragment linking will undoubtedly benefit from developments in computational approaches and machine learning models.
Collapse
Affiliation(s)
- Dylan Grenier
- Team Small Molecules for Biological Targets, Centre de Recherche En Cancérologie (CRCL) - INSERM 1052 - CNRS 5286 - Centre Léon Bérard - Université Claude Bernard Lyon 1, Institut Convergence Plascan, Lyon, France
| | - Solène Audebert
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Jordane Preto
- Team Small Molecules for Biological Targets, Centre de Recherche En Cancérologie (CRCL) - INSERM 1052 - CNRS 5286 - Centre Léon Bérard - Université Claude Bernard Lyon 1, Institut Convergence Plascan, Lyon, France
| | - Jean-François Guichou
- Centre de Biologie Structurale, CNRS, INSERM, Univ. Montpellier, Montpellier, France
| | - Isabelle Krimm
- Team Small Molecules for Biological Targets, Centre de Recherche En Cancérologie (CRCL) - INSERM 1052 - CNRS 5286 - Centre Léon Bérard - Université Claude Bernard Lyon 1, Institut Convergence Plascan, Lyon, France
| |
Collapse
|
10
|
Thongkum W, Klayprasert P, Semakul N, Jakmunee J, Kasinrerk W, Setshedi M, Sayed Y, Tayapiwatana C. Semi-quantification and Potency Verification of the HIV Protease Inhibitor Based on the Matrix-Capsid Protein Immobilized Nickel (II)/NTA-Tol/Graphene Oxide/SPCE Electrochemical Biosensor. ACS OMEGA 2023; 8:17932-17940. [PMID: 37251123 PMCID: PMC10210225 DOI: 10.1021/acsomega.3c01031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 05/04/2023] [Indexed: 05/31/2023]
Abstract
Human immunodeficiency virus (HIV) causing acquired immune deficiency syndrome (AIDS) is still a global issue. Long-term drug treatment and nonadherence to medication increase the spread of drug-resistant HIV strains. Therefore, the identification of new lead compounds is being investigated and is highly desirable. Nevertheless, a process generally necessitates a significant budget and human resources. In this study, a simple biosensor platform for semi-quantification and verification of the potency of HIV protease inhibitors (PIs) based on electrochemically detecting the cleavage activity of the HIV-1 subtype C-PR (C-SA HIV-1 PR) was proposed. An electrochemical biosensor was fabricated by immobilizing His6-matrix-capsid (H6MA-CA) on the electrode surface via the chelation to Ni2+-nitrilotriacetic acid (NTA) functionalized GO. The functional groups and the characteristics of modified screen-printed carbon electrodes (SPCE) were characterized by Fourier transform infrared (FTIR) spectroscopy, scanning electron microscopy (SEM), and energy-dispersive X-ray spectroscopy (EDS). C-SA HIV-1 PR activity and the effect of PIs were validated by recording changes in electrical current signals of the ferri/ferrocyanide redox probe. The detection of PIs, i.e., lopinavir (LPV) and indinavir (IDV), toward the HIV protease was confirmed by the decrease in the current signals in a dose-dependent manner. In addition, our developed biosensor demonstrates the ability to distinguish the potency of two PIs to inhibit C-SA HIV-1 PR activities. We anticipated that this low-cost electrochemical biosensor would increase the efficiency of the lead compound screening process and accelerate the discovery and development of new HIV drugs.
Collapse
Affiliation(s)
- Weeraya Thongkum
- Division
of Clinical Immunology, Department of Medical Technology, Faculty
of Associated Medical Sciences, Chiang Mai
University, Chiang
Mai 50200, Thailand
- Center
of Innovative Immunodiagnostic Development, Department of Medical
Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang
Mai 50200, Thailand
- Center
of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical
Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Puttaporn Klayprasert
- Research
Laboratory for Analytical Instrument and Electrochemistry Innovation,
Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang
Mai 50200, Thailand
| | - Natthawat Semakul
- Department
of Chemistry, Faculty of Science, Chiang
Mai University, Chiang Mai 50200, Thailand
| | - Jaroon Jakmunee
- Research
Laboratory for Analytical Instrument and Electrochemistry Innovation,
Department of Chemistry, Faculty of Science, Chiang Mai University, Chiang
Mai 50200, Thailand
- Department
of Chemistry, Faculty of Science, Chiang
Mai University, Chiang Mai 50200, Thailand
- Center
of
Excellence for Innovation in Chemistry, Faculty of Science, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Watchara Kasinrerk
- Division
of Clinical Immunology, Department of Medical Technology, Faculty
of Associated Medical Sciences, Chiang Mai
University, Chiang
Mai 50200, Thailand
- Center
of Innovative Immunodiagnostic Development, Department of Medical
Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang
Mai 50200, Thailand
| | - Mpho Setshedi
- Protein
Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Wits 2050, South Africa
| | - Yasien Sayed
- Protein
Structure-Function Research Unit, School of Molecular and Cell Biology, University of the Witwatersrand, Wits 2050, South Africa
| | - Chatchai Tayapiwatana
- Division
of Clinical Immunology, Department of Medical Technology, Faculty
of Associated Medical Sciences, Chiang Mai
University, Chiang
Mai 50200, Thailand
- Center
of Innovative Immunodiagnostic Development, Department of Medical
Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang
Mai 50200, Thailand
- Center
of Biomolecular Therapy and Diagnostic, Faculty of Associated Medical
Sciences, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
11
|
Said AI, Gajdács M, Zupkó I, Haukka M, Palkó M. Angular Regioselective Synthesis of Varied Functionalized Hexahydro-1,2,4-triazolo[4,3- a]quinazolin-9-ones and Their Antiproliferative Action. Molecules 2023; 28:molecules28093718. [PMID: 37175130 PMCID: PMC10180492 DOI: 10.3390/molecules28093718] [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: 04/12/2023] [Revised: 04/19/2023] [Accepted: 04/19/2023] [Indexed: 05/15/2023] Open
Abstract
New 2-thioxopyrimidin-4-ones capable of participating in regioselective reactions with functionally diverse hydrazonoyl chlorides towards angular regioisomers, rather than linear ones, were designed and synthesized to form stereoisomeric cis- and trans-hexahydro [1,2,4]triazolo[4,3-a]quinazolin-9-ones to be tested as antitumor candidates. The angular regiochemistry of the products was verified through crystallographic experiments and NMR studies. In addition, the regioselectivity of the reaction was found to be independent of the stereochemistry of the used 2-thioxopyrimidin-4-one. Only compound 4c demonstrated satisfactory growth inhibition against all the cancer cells used among all the produced drugs.
Collapse
Affiliation(s)
- Awad I Said
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
- Chemistry Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Márió Gajdács
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - István Zupkó
- Institute of Pharmacodynamics and Biopharmacy, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| | - Matti Haukka
- Department of Chemistry, University of Jyväskulä, FIN-40014 Jyväskulä, Finland
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry, University of Szeged, Eötvös u. 6, H-6720 Szeged, Hungary
| |
Collapse
|
12
|
Nacheva K, Kulkarni SS, Kassu M, Flanigan D, Monastyrskyi A, Iyamu ID, Doi K, Barber M, Namelikonda N, Tipton JD, Parvatkar P, Wang HG, Manetsch R. Going beyond Binary: Rapid Identification of Protein-Protein Interaction Modulators Using a Multifragment Kinetic Target-Guided Synthesis Approach. J Med Chem 2023; 66:5196-5207. [PMID: 37000900 PMCID: PMC10620989 DOI: 10.1021/acs.jmedchem.3c00108] [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: 01/18/2023] [Indexed: 04/03/2023]
Abstract
Kinetic target-guided synthesis (KTGS) is a powerful screening approach that enables identification of small molecule modulators for biomolecules. While many KTGS variants have emerged, a majority of the examples suffer from limited throughput and a poor signal/noise ratio, hampering reliable hit detection. Herein, we present our optimized multifragment KTGS screening strategy that tackles these limitations. This approach utilizes selected reaction monitoring liquid chromatography tandem mass spectrometry for hit detection, enabling the incubation of 190 fragment combinations per screening well. Consequentially, our fragment library was expanded from 81 possible combinations to 1710, representing the largest KTGS screening library assembled to date. The expanded library was screened against Mcl-1, leading to the discovery of 24 inhibitors. This work unveils the true potential of KTGS with respect to the rapid and reliable identification of hits, further highlighting its utility as a complement to the existing repertoire of screening methods used in drug discovery.
Collapse
Affiliation(s)
- Katya Nacheva
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Sameer S. Kulkarni
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Mintesinot Kassu
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - David Flanigan
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
- Department
of Sciences, Hillsborough Community College, Tampa, Florida 33619, United States
| | - Andrii Monastyrskyi
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Iredia D. Iyamu
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Kenichiro Doi
- Department
of Pediatrics, Division of Pediatric Hematology and Oncology, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Megan Barber
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Niranjan Namelikonda
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
| | - Jeremiah D. Tipton
- Proteomics
and Mass Spectrometry Core Facility, University
of South Florida, Tampa, Florida 33620, United States
| | - Prakash Parvatkar
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
| | - Hong-Gang Wang
- Department
of Pediatrics, Division of Pediatric Hematology and Oncology, Penn State College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Roman Manetsch
- Department
of Chemistry, University of South Florida, Tampa, Florida 33620, United States
- Department
of Chemistry and Chemical Biology, Northeastern
University, Boston, Massachusetts 02115, United States
- Department
of Pharmaceutical Sciences, Northeastern
University, Boston, Massachusetts 02115, United States
- Center for
Drug Discovery, Northeastern University, Boston, Massachusetts 02115, United States
- Barnett
Institute of Chemical and Biological Analysis, Northeastern University, Boston, Massachusetts 02115, United States
| |
Collapse
|
13
|
Dong R, Yang X, Wang B, Ji X. Mutual leveraging of proximity effects and click chemistry in chemical biology. Med Res Rev 2023; 43:319-342. [PMID: 36177531 DOI: 10.1002/med.21927] [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: 11/30/2021] [Revised: 08/14/2022] [Accepted: 09/11/2022] [Indexed: 02/05/2023]
Abstract
Nature has the remarkable ability to realize reactions under physiological conditions that normally would require high temperature and other forcing conditions. In doing so, often proximity effects such as simultaneous binding of two reactants in the same pocket and/or strategic positioning of catalytic functional groups are used as ways to achieve otherwise kinetically challenging reactions. Though true biomimicry is challenging, there have been many beautiful examples of how to leverage proximity effects in realizing reactions that otherwise would not readily happen under near-physiological conditions. Along this line, click chemistry is often used to endow proximity effects, and proximity effects are also used to further leverage the facile and bioorthogonal nature of click chemistry. This review brings otherwise seemingly unrelated topics in chemical biology and drug discovery under one unifying theme of mutual leveraging of proximity effects and click chemistry and aims to critically analyze the biomimicry use of such leveraging effects as powerful approaches in chemical biology and drug discovery. We hope that this review demonstrates the power of employing mutual leveraging proximity effects and click chemistry and inspires the development of new strategies that will address unmet needs in chemistry and biology.
Collapse
Affiliation(s)
- Ru Dong
- Department of Medicinal Chemistry, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| | - Xiaoxiao Yang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Binghe Wang
- Department of Chemistry and Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA
| | - Xingyue Ji
- Department of Medicinal Chemistry, College of Pharmaceutical Science, Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
14
|
Gül EY, Karataş EA, Doğan HA, Karataş ÖF, Çoşut B, Eçik ET. Erlotinib-Modified BODIPY Photosensitizers for Targeted Photodynamic Therapy. ChemMedChem 2023; 18:e202200439. [PMID: 36317417 DOI: 10.1002/cmdc.202200439] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/28/2022] [Indexed: 11/06/2022]
Abstract
Photodynamic therapy (PDT) is an innovative, non-invasive and highly selective therapeutic modality for tumours and non-malignant diseases. BODIPY based molecules can function as new generation photosensitizers (PSs) in various PDT applications. Despite numerous conjugated PS systems are available, BODIPYs containing erlotinib lagged behind other photosensitizer units. In this study, smart photosensitizers containing BODIPY, erlotinib and hydrophilic units were prepared for the first time, their physicochemical properties and PDT effects were investigated. Compared with non-halogenated compound, halogenated derivatives possessed much lower fluorescence profile as well as the good ROS generation ability under red light. In vitro PDT studies were performed on both healthy (PNT1a) and prostate cancerous cells (PC3) to determine the selectivity of the compounds on cancerous cells and their effects under light. The halogenated conjugates, exposed to low dose of light illumination exhibited potent activity on cancer cell viability and the calculated IC50 values proved the high phototoxicity of the photosensitizers. It was also determined that the PSs have very low dark toxicity and that the light illumination and ROS formation are required for the initiation of the cell death mechanism. As a result, erlotinib modified BODIPYs could serve as promising agents in anticancer photodynamic therapy.
Collapse
Affiliation(s)
- Elif Yıldız Gül
- Department of Chemistry, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey
| | - Elanur Aydın Karataş
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25050, Erzurum, Turkey.,High Technology Application and Research Center, Erzurum Technical University, 25050, Erzurum, Turkey
| | - Hatice Aydın Doğan
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25050, Erzurum, Turkey.,High Technology Application and Research Center, Erzurum Technical University, 25050, Erzurum, Turkey
| | - Ömer Faruk Karataş
- Department of Molecular Biology and Genetics, Faculty of Science, Erzurum Technical University, 25050, Erzurum, Turkey.,High Technology Application and Research Center, Erzurum Technical University, 25050, Erzurum, Turkey
| | - Bünyemin Çoşut
- Department of Chemistry, Gebze Technical University, 41400, Kocaeli, Turkey
| | - Esra Tanrıverdi Eçik
- Department of Chemistry, Faculty of Science, Atatürk University, 25240, Erzurum, Turkey
| |
Collapse
|
15
|
Synthesis of Benzothiazole Linked Triazole Conjugates and Their Evaluation Against Cholinesterase Enzymes. ChemistrySelect 2022. [DOI: 10.1002/slct.202203060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
16
|
Bhattacherjee D, Kovalev IS, Kopchuk DS, Rahman M, Santra S, Zyryanov GV, Das P, Purohit R, Rusinov VL, Chupakhin ON. Mechanochemical Approach towards Multi-Functionalized 1,2,3-Triazoles and Anti-Seizure Drug Rufinamide Analogs Using Copper Beads. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27227784. [PMID: 36431885 PMCID: PMC9693609 DOI: 10.3390/molecules27227784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/04/2022] [Accepted: 11/07/2022] [Indexed: 11/16/2022]
Abstract
Highly regiospecific, copper-salt-free and neat conditions have been demonstrated for the 1,3-dipolar azide-alkyne cycloaddition (AAC) reactions under mechanochemical conditions. A group of structurally challenging alkynes and heterocyclic derivatives was efficiently implemented to achieve highly functionalized 1,4-disubstituted-1,2,3-triazoles in good to excellent yield by using the Cu beads without generation of unwanted byproducts. Furthermore, the high-speed ball milling (HSBM) strategy has also been extended to the synthesis of the commercially available pharmaceutical agent, Rufinamide, an antiepileptic drug (AED) and its analogues. The same strategy was also applied for the synthesis of the Cl-derivative of Rufinamide. Analysis of the single crystal XRD data of the triazole was also performed for the final structural confirmation. The Cu beads are easily recoverable from the reaction mixture and used for the further reactions without any special treatment.
Collapse
Affiliation(s)
- Dhananjay Bhattacherjee
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Igor S. Kovalev
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Dmitry S. Kopchuk
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| | - Matiur Rahman
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
| | - Sougata Santra
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- Correspondence:
| | - Grigory V. Zyryanov
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| | - Pralay Das
- Chemical Technology Division, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur 176061, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Rituraj Purohit
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Structural Bioinformatics Lab, CSIR-Institute of Himalayan Bioresource Technology (CSIR-IHBT), Palampur 176061, India
| | - Vladimir L. Rusinov
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| | - Oleg N. Chupakhin
- Department of Organic and Biomolecular Chemistry, Ural Federal University, 19 Mira Street, 620002 Yekaterinburg, Russia
- I. Ya. Postovsky Institute of Organic Synthesis, Ural Branch of the Russian Academy of Sciences, 22 S. Kovalevskoi Street, 620219 Yekaterinburg, Russia
| |
Collapse
|
17
|
Gupta P, Kumar P, Syal B, Shamim T. Synthesis of 1,4-disubstituted-1,2,3-triazoles using starch-functionalized copper (II) acetate as a recyclable heterogeneous catalyst in water. RESEARCH ON CHEMICAL INTERMEDIATES 2022. [DOI: 10.1007/s11164-022-04829-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
18
|
Sykam K, Donempudi S, Basak P. 1,2,
3‐Triazole
rich polymers for flame retardant application: A review. J Appl Polym Sci 2022. [DOI: 10.1002/app.52771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Kesavarao Sykam
- Polymers & Functional Materials Division CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovation Research (AcSIR) Ghaziabad India
| | - Shailaja Donempudi
- Polymers & Functional Materials Division CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovation Research (AcSIR) Ghaziabad India
| | - Pratyay Basak
- Polymers & Functional Materials Division CSIR‐Indian Institute of Chemical Technology Hyderabad India
- Academy of Scientific and Innovation Research (AcSIR) Ghaziabad India
| |
Collapse
|
19
|
Verma N, Bera S, Gonnade R, Mondal D. Regioselective synthesis of 1,4,5‐Trisubstituted‐1,2,3‐Triazole Derivatives from α,β‐Unsaturated Carbonyls. European J Org Chem 2022. [DOI: 10.1002/ejoc.202200317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Naimish Verma
- Central University of Gujarat School of Chemical Sciences Sector-30 382030 Gandhinagar INDIA
| | - Smritilekha Bera
- Central University of Gujarat School of Chemical Sciences 30 sector 382030 Gandhinagar INDIA
| | - Rajesh Gonnade
- CSIR-National Chemical Laboratory: National Chemical Laboratory CSIR Physical and Materials Chemistry Division 411008 Pune INDIA
| | - Dhananjoy Mondal
- Central University of Gujarat School of Chemical Sciences Sector-30 382030 Gandhinagar INDIA
| |
Collapse
|
20
|
Moussaoui O, Bhadane R, Sghyar R, Ilaš J, El Hadrami EM, Chakroune S, Salo‐Ahen OMH. Design, Synthesis, in vitro and in silico Characterization of 2-Quinolone-L-alaninate-1,2,3-triazoles as Antimicrobial Agents. ChemMedChem 2022; 17:e202100714. [PMID: 34978160 PMCID: PMC9305408 DOI: 10.1002/cmdc.202100714] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 12/16/2021] [Indexed: 11/07/2022]
Abstract
Due to the ever-increasing antimicrobial resistance there is an urgent need to continuously design and develop novel antimicrobial agents. Inspired by the broad antibacterial activities of various heterocyclic compounds such as 2-quinolone derivatives, we designed and synthesized new methyl-(2-oxo-1,2-dihydroquinolin-4-yl)-L-alaninate-1,2,3-triazole derivatives via 1,3-dipolar cycloaddition reaction of 1-propargyl-2-quinolone-L-alaninate with appropriate azide groups. The synthesized compounds were obtained in good yield ranging from 75 to 80 %. The chemical structures of these novel hybrid molecules were determined by spectroscopic methods and the antimicrobial activity of the compounds was investigated against both bacterial and fungal strains. The tested compounds showed significant antimicrobial activity and weak to moderate antifungal activity. Despite the evident similarity of the quinolone moiety of our compounds with fluoroquinolones, our compounds do not function by inhibiting DNA gyrase. Computational characterization of the compounds shows that they have attractive physicochemical and pharmacokinetic properties and could serve as templates for developing potential antimicrobial agents for clinical use.
Collapse
Affiliation(s)
- Oussama Moussaoui
- Laboratory of Applied Organic ChemistrySidi Mohamed Ben Abdellah University30000FezMorocco
| | - Rajendra Bhadane
- Structural Bioinformatics Laboratory, BiochemistryÅbo Akademi University20520TurkuFinland
- Pharmaceutical Sciences Laboratory, PharmacyÅbo Akademi University20520TurkuFinland
| | - Riham Sghyar
- Laboratory of Applied Organic ChemistrySidi Mohamed Ben Abdellah University30000FezMorocco
| | - Janez Ilaš
- Faculty of PharmacyUniversity of Ljubljana1000LjubljanaSlovenia
| | - El Mestafa El Hadrami
- Laboratory of Applied Organic ChemistrySidi Mohamed Ben Abdellah University30000FezMorocco
| | - Said Chakroune
- Laboratory of Applied Organic ChemistrySidi Mohamed Ben Abdellah University30000FezMorocco
| | - Outi M. H. Salo‐Ahen
- Structural Bioinformatics Laboratory, BiochemistryÅbo Akademi University20520TurkuFinland
- Pharmaceutical Sciences Laboratory, PharmacyÅbo Akademi University20520TurkuFinland
| |
Collapse
|
21
|
Novel Copper Tagged Supported Ionic Liquid Phase Catalyst for the Synthesis of 1,4‑Disubstituted 1,2,3‑Triazoles via Cu-catalyzed Azide–Alkyne Cycloaddition Reactions in Water. Catal Letters 2022. [DOI: 10.1007/s10562-021-03898-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
22
|
Caiana RRA, Santos CS, de Oliveira RN, Freitas JCR. Scientific and Technological Prospecting of 1H-1,2,3-Triazoles. CURR ORG CHEM 2022. [DOI: 10.2174/1385272826666220126153429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Abstract:
The use of 1H-1,2,3-triazoles has become an important scaffold for applications in different technological sectors. Therefore, we sought to carry out a technological monitoring to understand the international scenario involving 1H-1,2,3-triazoles from the patents filed, in addition to evaluating the relationship between the growth in the number of patents and the improvement of strategies for obtaining of these compounds via a metal-catalyzed azide-alkyne cycloaddition reaction. Technological monitoring was performed with the support of the PatentInspiration® platform, using the keywords "1,2,3-triazol", "1,2,3-triazole", and "1,2,3-triazolyl". A total of 960 registered patents were found, most for the years 2014 and 2019. The main filers were prestigious multinational companies such as Syngenta, Merck, Sandoz, Pfizer, and Bayer. The United States, China, Japan, and Germany lead patent registrations, mainly addressing innovations in chemistry and metallurgy, human needs, and new technologies. These results help to understand the state of innovation for this topic, pointing out the characteristics of the main discoveries concerning 1H-1,2,3-triazole derivatives.
Collapse
Affiliation(s)
| | - Cosme Silva Santos
- Department of Chemistry, Federal Rural University of Pernambuco, 52171-900, Recife-PE, Brazil
| | | | | |
Collapse
|
23
|
Oubella A, Bimoussa A, N’ait Oussidi A, Fawzi M, Auhmani A, Morjani H, Riahi A, Esseffar M, Parish C, Ait Itto MY. New 1,2,3-Triazoles from (R)-Carvone: Synthesis, DFT Mechanistic Study and In Vitro Cytotoxic Evaluation. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030769. [PMID: 35164037 PMCID: PMC8839216 DOI: 10.3390/molecules27030769] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/14/2021] [Accepted: 10/18/2021] [Indexed: 12/11/2022]
Abstract
Aseries of novel 1,4-disubstituted 1,2,3-triazoles were synthesized from an (R)-carvone terminal alkyne derivative via a Cu (I)-catalyzed azide–alkyne cycloaddition reaction using CuSO4,5H2O as the copper (II) source and sodium ascorbate as a reducing agent which reduces Cu (II) into Cu (I). All the newly synthesized 1,2,3-triazoles 9a–h were fully identified on the basis of their HRMS and NMR spectral data and then evaluated for their cell growth inhibition potential by MTS assay against HT-1080 fibrosarcoma, A-549 lung carcinoma, and two breast adenocarcinoma (MCF-7 and MDA-MB-231) cell lines. Compound 9d showed notable cytotoxic effects against the HT-1080 and MCF-7 cells with IC50 values of 25.77 and 27.89 µM, respectively, while compound 9c displayed significant activity against MCF-7 cells with an IC50 value of 25.03 µM. Density functional calculations at the B3LYP/6-31G* level of theory were used to confirm the high reactivity of the terminal alkyne as a dipolarophile. Quantum calculations were also used to investigate the mechanism of both the uncatalyzed and copper (I)-catalyzed azide–alkyne cycloaddition reaction (CuAAC). The catalyzed reaction gives complete regioselectivity via a stepwise mechanism streamlining experimental observations. The calculated free-energy barriers 4.33 kcal/mol and 29.35 kcal/mol for the 1,4- and 1,5-regioisomers, respectively, explain the marked regioselectivity of the CuAAC reaction.
Collapse
Affiliation(s)
- Ali Oubella
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
| | - Abdoullah Bimoussa
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
| | - Abdellah N’ait Oussidi
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
| | - Mourad Fawzi
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
| | - Aziz Auhmani
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
| | - Hamid Morjani
- BioSpectroscopie Translationnelle, BioSpecT—EA7506, UFR de Pharmacie, Université de Reims Champagne-Ardenne, 51 Rue Cognacq Jay, CEDEX, 51096 Reims, France;
| | - Abdelkhalek Riahi
- Equipe MSO, CNRS UMR 7312 Institut de Chimie Moléculaire, Université de Reims Champagne-Ardenne, Bat. Europol’Agro-Moulin de La Housse UFR Sciences B.P., 1039, CEDEX 2, 51687 Reims, France;
| | - M’hamed Esseffar
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
- Correspondence: (M.E.); (C.P.); (M.Y.A.)
| | - Carol Parish
- Gottwald Science Center, 28Westhampton Way, University of Richmond, Richmond, VA 23173, USA
- Correspondence: (M.E.); (C.P.); (M.Y.A.)
| | - Moulay Youssef Ait Itto
- Laboratoire de Chimie Moléculaire, Département de Chimie, Faculté des Sciences, Semlalia B.P 2390, Marrakech 40001, Morocco; (A.O.); (A.B.); (A.N.O.); (M.F.); (A.A.)
- Correspondence: (M.E.); (C.P.); (M.Y.A.)
| |
Collapse
|
24
|
Khatua M, Goswami B, Kamal, Samanta S. Azide-Alkyne "Click" Reaction in Water Using Parts-Per-Million Amine-Functionalized Azoaromatic Cu(I) Complex as Catalyst: Effect of the Amine Side Arm. Inorg Chem 2021; 60:17537-17554. [PMID: 34806366 DOI: 10.1021/acs.inorgchem.1c02115] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
A series of Cu(II) complexes, 1-4 and 6, were synthesized through a reaction of amine-functionalized pincer-like ligands, HL1,2, La,b, and a bidentate ligand L1 with CuCl2·2H2O. The chemical reduction of complex 1 using 1 equiv of sodium l-ascorbate resulted in a dimeric Cu(I) complex 5 in excellent yield. All of the complexes, 1-6, were thoroughly characterized using various physicochemical characterization techniques, single-crystal X-ray structure determination, and density functional theory calculations. Ligands HL1,2 and La,b behaved as tridentated donors by the coordination of the amine side arm in their respective Cu(II) complexes, and the amine side arm remained as a pendant in Cu(I) complexes. All of these complexes (1-6) were explored for copper(I)-catalyzed 1,3-dipolar azide-alkyne cycloaddition (CuAAC) reaction at room temperature in water under air. Complex 5 directly served as an active catalyst; however, complexes 1-4 and 6 required 1 equiv of sodium l-ascorbate to generate their corresponding active Cu(I) catalyst. It has been observed that azo-based ligand-containing Cu(I)-complexes are air-stable and were highly efficient for the CuAAC reaction. The amine side arm in the ligand backbone has a dramatic role in accelerating the reaction rate. Mechanistic investigations showed that the alkyne C-H deprotonation was the rate-limiting step and the pendant amine side arm intramolecularly served as a base for Cu-coordinated alkyne deprotonation, leading to the azide-alkyne 2 + 3 cycloaddition reaction. Thus, variation of the amine side arm in complexes 1-4 and use of the most basic diisopropyl amine moiety in complex 4 has resulted in an unique amine-functionalized azoaromatic Cu(I) system for CuAAC reaction upon sodium l-ascorbate reduction. The complex 4 has shown excellent catalysis at its low parts-per-million level loading in water. The catalytic protocol was versatile and exhibited very good functional group tolerance. It was also employed efficiently to synthesize a number of useful functional triazoles having medicinal, catalytic, and targeting properties.
Collapse
Affiliation(s)
- Manas Khatua
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India 741246
| | - Bappaditya Goswami
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, West Bengal, India 741246
| | - Kamal
- Department of Chemistry, Indian Institute of Technology Jammu, Jagti, Jammu, India 181221
| | - Subhas Samanta
- Department of Chemistry, Indian Institute of Technology Jammu, Jagti, Jammu, India 181221
| |
Collapse
|
25
|
Recent advances in the chemistry of 1,2,4-triazoles: Synthesis, reactivity and biological activities. Tetrahedron Lett 2021. [DOI: 10.1016/j.tetlet.2021.153518] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
26
|
Sheoran A, Kaur J, Agarwal J, Singhal S. Ring Opening of Epoxides and Aziridines with Benzotriazoles Using Magnetically Retrievable Graphene Based (CoFe@rGO) Nanohybrid. ChemistrySelect 2021. [DOI: 10.1002/slct.202101268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Ankush Sheoran
- Department of Chemistry & Centre of Advanced Studies in Chemistry Panjab University Chandigarh India- 160014
| | - Jaspreet Kaur
- Energy Research Centre Panjab University Chandigarh India- 160014
| | - Jyoti Agarwal
- Department of Chemistry & Centre of Advanced Studies in Chemistry Panjab University Chandigarh India- 160014
| | - Sonal Singhal
- Department of Chemistry & Centre of Advanced Studies in Chemistry Panjab University Chandigarh India- 160014
| |
Collapse
|
27
|
Beukeaw D, Yotphan S. Copper‐Catalyzed Decarboxylative Cycloaddition of Alkynyl Carboxylic Acids and Sodium Azide with Epoxides and Ethers. ChemistrySelect 2021. [DOI: 10.1002/slct.202102494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Danupat Beukeaw
- Department of Chemistry and Center of Excellence for Innovation in Chemistry Faculty of Science Mahidol University Rama VI Road Bangkok 10400 Thailand
| | - Sirilata Yotphan
- Department of Chemistry and Center of Excellence for Innovation in Chemistry Faculty of Science Mahidol University Rama VI Road Bangkok 10400 Thailand
| |
Collapse
|
28
|
Tauber C, Wamser R, Arkona C, Tügend M, Abdul Aziz UB, Pach S, Schulz R, Jochmans D, Wolber G, Neyts J, Rademann J. Chemische Evolution antiviraler Wirkstoffe gegen Enterovirus D68 durch Proteintemplat‐gesteuerte Knoevenagelreaktionen. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202102074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Carolin Tauber
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Rebekka Wamser
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Christoph Arkona
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Marisa Tügend
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Umer Bin Abdul Aziz
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Szymon Pach
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Robert Schulz
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Dirk Jochmans
- Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgien
| | - Gerhard Wolber
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| | - Johan Neyts
- Department of Microbiology, Immunology and Transplantation Rega Institute KU Leuven Leuven Belgien
| | - Jörg Rademann
- Fachbereich Biologie, Chemie and Pharmazie Institut für Pharmazie Medizinische Chemie Freie Universität Berlin Königin-Luise-Str. 2+4 14195 Berlin Deutschland
| |
Collapse
|
29
|
Tauber C, Wamser R, Arkona C, Tügend M, Abdul Aziz UB, Pach S, Schulz R, Jochmans D, Wolber G, Neyts J, Rademann J. Chemical Evolution of Antivirals Against Enterovirus D68 through Protein-Templated Knoevenagel Reactions. Angew Chem Int Ed Engl 2021; 60:13294-13301. [PMID: 33749121 PMCID: PMC8252737 DOI: 10.1002/anie.202102074] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Indexed: 02/06/2023]
Abstract
The generation of bioactive molecules from inactive precursors is a crucial step in the chemical evolution of life, however, mechanistic insights into this aspect of abiogenesis are scarce. Here, we investigate the protein-catalyzed formation of antivirals by the 3C-protease of enterovirus D68. The enzyme induces aldol condensations yielding inhibitors with antiviral activity in cells. Kinetic and thermodynamic analyses reveal that the bioactivity emerges from a dynamic reaction system including inhibitor formation, alkylation of the protein target by the inhibitors, and competitive addition of non-protein nucleophiles to the inhibitors. The most active antivirals are slowly reversible inhibitors with elongated target residence times. The study reveals first examples for the chemical evolution of bio-actives through protein-catalyzed, non-enzymatic C-C couplings. The discovered mechanism works under physiological conditions and might constitute a native process of drug development.
Collapse
Affiliation(s)
- Carolin Tauber
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Rebekka Wamser
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Christoph Arkona
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Marisa Tügend
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Umer Bin Abdul Aziz
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Szymon Pach
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Robert Schulz
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Dirk Jochmans
- Department of Microbiology, Immunology and TransplantationRega InstituteKU LeuvenLeuvenBelgium
| | - Gerhard Wolber
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| | - Johan Neyts
- Department of Microbiology, Immunology and TransplantationRega InstituteKU LeuvenLeuvenBelgium
| | - Jörg Rademann
- Department of Biology, Chemistry and PharmacyInstitute of PharmacyMedicinal ChemistryFreie Universität BerlinKönigin-Luise-Str. 2+414195BerlinGermany
| |
Collapse
|
30
|
Morais PAB, Francisco CS, de Paula H, Ribeiro R, Eloy MA, Javarini CL, Neto ÁC, Júnior VL. Semisynthetic Triazoles as an Approach in the Discovery of Novel Lead Compounds. CURR ORG CHEM 2021. [DOI: 10.2174/1385272825666210126100227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Historically, medicinal chemistry has been concerned with the approach of organic
chemistry for new drug synthesis. Considering the fruitful collections of new molecular entities,
the dedicated efforts for medicinal chemistry are rewarding. Planning and search for new
and applicable pharmacologic therapies involve the altruistic nature of the scientists. Since
the 19th century, notoriously applying isolated and characterized plant-derived compounds in
modern drug discovery and various stages of clinical development highlight its viability and
significance. Natural products influence a broad range of biological processes, covering transcription,
translation, and post-translational modification, being effective modulators of most
basic cellular processes. The research of new chemical entities through “click chemistry”
continuously opens up a map for the remarkable exploration of chemical space towards leading
natural products optimization by structure-activity relationship. Finally, in this review, we expect to gather a
broad knowledge involving triazolic natural product derivatives, synthetic routes, structures, and their biological activities.
Collapse
Affiliation(s)
- Pedro Alves Bezerra Morais
- Centro de Ciencias Exatas, Naturais e da Saude, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Carla Santana Francisco
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Heberth de Paula
- Centro de Ciencias Exatas, Naturais e da Saude, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Rayssa Ribeiro
- Programa de Pos- Graduacao em Agroquimica, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Mariana Alves Eloy
- Programa de Pos- Graduacao em Agroquimica, Universidade Federal do Espirito Santo, 29500000, Alegre, ES, Brazil
| | - Clara Lirian Javarini
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Álvaro Cunha Neto
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| | - Valdemar Lacerda Júnior
- Programa de Pos-Graduacao em Quimica, Universidade Federal do Espirito Santo, 29075910, Vitória, ES, Brazil
| |
Collapse
|
31
|
De Rosa L, Capasso D, Diana D, Stefania R, Di Stasi R, Fattorusso R, D'Andrea LD. Metabolic and conformational stabilization of a VEGF-mimetic beta-hairpin peptide by click-chemistry. Eur J Med Chem 2021; 222:113575. [PMID: 34130005 DOI: 10.1016/j.ejmech.2021.113575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 04/30/2021] [Accepted: 05/19/2021] [Indexed: 01/09/2023]
Abstract
HPLW is a Vascular Endothelial Growth Factor (VEGF)-mimicking beta-hairpin peptide endowed of proangiogenic effect and showing valuable biomedical application in the proangiogenic therapy. However, the translational potential of HPLW is limited by its low metabolic stability, which would shorten the in vivo efficacy of the molecule. Here, we developed a peptide analog of HPLW, named HPLW2, that retains the structural and biological properties of the original peptide but features an impressive resistance to degradation by human serum proteases. HPLW2 was obtained by covalently modifying the chemical structure of the peptide with molecular tools known to impart protease resistance. Notably, the peptide was cyclized by installing an interstrand triazole bridge through Cu(I)-catalyzed azide-alkyne 1,3-dipolar cycloaddition (CuAAC) reaction. HPLW2 appears as a novel and promising drug candidate with potential biomedical application in the proangiogenic therapy as a low molecular weight drug, alternative to the use of VEGF. Our work points out the utility of the interstrand triazole bridge as effective chemical platform for the conformational and metabolic stabilization of beta-hairpin bioactive peptides.
Collapse
Affiliation(s)
- Lucia De Rosa
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Domenica Capasso
- CESTEV, Università di Napoli "Federico II", Via De Amicis 95, 80134, Napoli, Italy; CIRPeB Università di Napoli "Federico II" Via Mezzocannone 16, 80134, Napoli, Italy
| | - Donatella Diana
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Rachele Stefania
- Dipartimento di Biotecnologie Molecolari e Scienze per La Salute, Università di Torino, Via Nizza 52, 10126, Torino, Italy
| | - Rossella Di Stasi
- Istituto di Biostrutture e Bioimmagini, CNR, Via Mezzocannone 16, 80134, Napoli, Italy
| | - Roberto Fattorusso
- CIRPeB Università di Napoli "Federico II" Via Mezzocannone 16, 80134, Napoli, Italy; Dipartimento di Scienze e Tecnologie Ambientali, Biologiche e Farmaceutiche, Università Della Campania "Luigi Vanvitelli", Via Vivaldi 43, 81100, Caserta, Italy
| | - Luca Domenico D'Andrea
- Istituto di Scienze e Tecnologie Chimiche "Giulio Natta", CNR, Via M. Bianco 9, 20131, Milano, Italy.
| |
Collapse
|
32
|
Seck I, Nguemo F. Triazole, imidazole, and thiazole-based compounds as potential agents against coronavirus. RESULTS IN CHEMISTRY 2021; 3:100132. [PMID: 33907666 PMCID: PMC8061185 DOI: 10.1016/j.rechem.2021.100132] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 04/19/2021] [Indexed: 02/08/2023] Open
Abstract
The expansion of the novel coronavirus known as SARS-CoV-2 (severe acute respiratory syndrome coronavirus 2), COVID-19 (coronavirus disease 2019), or 2019-nCoV (2019 novel coronavirus) is a global concern over its pandemic potential. The need for therapeutic alternatives to stop this new pandemic is urgent. Nowadays, no efficacious therapy is available, and vaccines and drugs are underdeveloped to cure or prevent SARS-CoV-2 infections in many countries. Some vaccines candidates have been approved; however, a number of people are still skeptical of this coronavirus vaccines. Probably because of issues related to the quantity of the vaccine and a possible long-term side effects which are still being studied. The previous pandemics of infections caused by coronavirus, such as SARS-CoV in 2003, the Middle East respiratory syndrome coronavirus (MERS-CoV) in 2012, HCoV-229E, and HCoV-OC43 were described in the 1960 s, -HCoV-NL63 isolated in 2004, and HCoV-HKU1identified in 2005 prompted researchers to characterize many compounds against these viruses. Most of them could be potentially active against the currently emerging novel coronavirus. Five membered nitrogen heterocycles with a triazole, imidazole, and thiazole moiety are often found in many bioactive molecules such as coronavirus inhibitors. This present work summarizes to review the biological and structural studies of these compound types as coronavirus inhibitors.
Collapse
Affiliation(s)
- Insa Seck
- Department of Chemistry, Faculty of Sciences and Technics, Cheikh Anta Diop University of Dakar, Dakar, Senegal
| | - Filomain Nguemo
- Institute for Neurophysiology, University of Cologne, Cologne, Germany
| |
Collapse
|
33
|
Shukla AK, Shrivash MK, Pandey A, Pandey J. Synthesis, in vitro and computational studies of novel glycosyl-1, 2, 3-1H-triazolyl methyl benzamide derivatives as potential α-glucosidase inhibitory activity. Bioorg Chem 2021; 109:104687. [PMID: 33601140 DOI: 10.1016/j.bioorg.2021.104687] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 12/22/2020] [Accepted: 01/21/2021] [Indexed: 10/22/2022]
Abstract
A series of novel glycosyl-1,2,3-1H-triazolyl methyl benzamide analogues were synthesized by the unambiguous strategy and evaluated for α-glucosidase inhibitory activity. Glycosyl benzamide exhibited a dose-dependent inhibition of α-glucosidase activity. The In-vitro α-glucosidase inhibition activity results indicated that all the synthesized triazolyl methyl benzamide compounds (IC50 values ranging from 25.3 ± 0.8 to 118.5 ± 5.3 μM) exhibited more inhibitory activity in comparison with the standard drug acarbose (IC50 = 750.0 ± 12.5 μM). Among all, the 3 deacetylated glycosyl methyl benzamide derivatives (4c, 4d and 4f) showed promising α-glucosidase enzyme inhibitory activities with IC50 value 25.3 ± 0.8, 26.1 ± 1.5 and 30.6 ± 2.1 respectively. Furthermore, these compounds were subjected to molecular docking and molecular dynamics simulation studies. The molecular docking studies were performed between (PDB ID: 3A4A) target protein and these synthesized molecules. The compounds displayed good docking energies in the range of -7.5 to -7.8 Kcal/mol. This work could be used as an initial approach in identifying potential novel molecules with the promising activity of type-2 diabetes mellitus.
Collapse
Affiliation(s)
- Akhilesh Kumar Shukla
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh 226025, India
| | - Manoj Kumar Shrivash
- Department of Applied Sciences, Indian Institute Information Technology Allahabad, India; Special Centre for Molecular Medicine, JNU, New Delhi 110067, India
| | - Anwesh Pandey
- Special Centre for Molecular Medicine, JNU, New Delhi 110067, India
| | - Jyoti Pandey
- Department of Chemistry, Babasaheb Bhimrao Ambedkar University, Lucknow, Uttar Pradesh 226025, India.
| |
Collapse
|
34
|
da S M Forezi L, Lima CGS, Amaral AAP, Ferreira PG, de Souza MCBV, Cunha AC, de C da Silva F, Ferreira VF. Bioactive 1,2,3-Triazoles: An Account on their Synthesis, Structural Diversity and Biological Applications. CHEM REC 2021; 21:2782-2807. [PMID: 33570242 DOI: 10.1002/tcr.202000185] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/14/2022]
Abstract
The triazole heterocycle is a privileged scaffold in medicinal chemistry, since its structure is present in a large number of biologically active molecules, including several drugs currently in the market. Due to their vast applications, a wide variety of methods are described for their preparation, such as the 1,3-dipolar cycloaddition and processes involving diazo compounds and diazo transfer reactions. Considering the significant number of contributions from our research group to this chemistry in recent decades, in this account we discuss both the development of new methods for the synthesis of 1,2,3-triazoles and the preparation of new triazole-functionalized biologically active molecules using classical approaches.
Collapse
Affiliation(s)
- Luana da S M Forezi
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-150, Niterói, RJ, Brazil
| | - Carolina G S Lima
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-150, Niterói, RJ, Brazil
| | - Adriane A P Amaral
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-150, Niterói, RJ, Brazil
| | - Patricia G Ferreira
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, R. Dr. Mario Vianna, 523 - Santa Rosa, 24241-000, Niterói, RJ, Brazil
| | - Maria Cecília B V de Souza
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-150, Niterói, RJ, Brazil
| | - Anna C Cunha
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-150, Niterói, RJ, Brazil
| | - Fernando de C da Silva
- Departamento de Química Orgânica, Instituto de Química, Universidade Federal Fluminense, Campus do Valonguinho, 24020-150, Niterói, RJ, Brazil
| | - Vitor F Ferreira
- Departamento de Tecnologia Farmacêutica, Faculdade de Farmácia, Universidade Federal Fluminense, R. Dr. Mario Vianna, 523 - Santa Rosa, 24241-000, Niterói, RJ, Brazil
| |
Collapse
|
35
|
Zhang X, Pan Y, Wang H, Liang C, Ma X, Jiao W, Shao H. Strategy to Construct 1,2,3‐Triazoles by K
2
CO
3
‐Mediated [4+1] Annulation Reactions of
N
‐Acetyl Hydrazones with Bifunctional Amino Reagents. Adv Synth Catal 2020. [DOI: 10.1002/adsc.202001375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Xiaoke Zhang
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
- Zunyi Medical University Zunyi Guizhou People's Republic of China
- University of Chinese Academy of Sciences People's Republic of China
| | - Yang Pan
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
- University of Chinese Academy of Sciences People's Republic of China
| | - Haibo Wang
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
- Zhejiang Hongyuan Pharmaceutical Co., Ltd. Chem & APIs. Industrial Zone, Linhai Taizhou Zhejiang People's Republic of China
- University of Chinese Academy of Sciences People's Republic of China
| | - Chong Liang
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
| | - Xiaofeng Ma
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
| | - Wei Jiao
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
| | - Huawu Shao
- Natural Products Research Centre,Chengdu Institute of Biology Chinese Academy of Sciences Chengdu People's Republic of China
| |
Collapse
|
36
|
Pomeislová A, Vrzal L, Kozák J, Dobiaš J, Hubálek M, Dvořáková H, Reyes‐Gutiérrez PE, Teplý F, Veverka V. Kinetic Target-Guided Synthesis of Small-Molecule G-Quadruplex Stabilizers. ChemistryOpen 2020; 9:1236-1250. [PMID: 33304739 PMCID: PMC7713561 DOI: 10.1002/open.202000261] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 11/02/2020] [Indexed: 12/25/2022] Open
Abstract
The formation of a G-quadruplex motif in the promoter region of the c-MYC protooncogene prevents its expression. Accordingly, G-quadruplex stabilization by a suitable ligand may be a viable approach for anticancer therapy. In our study, we used the 4-(4-methylpiperazin-1-yl)aniline molecule, previously identified as a fragment library screen hit, as a template for the SAR-guided design of a new small library of clickable fragments and subjected them to click reactions, including kinetic target-guided synthesis in the presence of a G-quadruplex forming oligonucleotide Pu24. We tested the clickable fragments and products of click reactions for their G-quadruplex stabilizing activity and determined their mode of binding to the c-MYC G-quadruplex by NMR spectroscopy. The enhanced stabilizing potency of click products in biology assays (FRET, Polymerase extension assay) matched the increased yields of in situ click reactions. In conclusion, we identified the newly synthesized click products of bis-amino derivatives of 4-(4-methylpiperazin-1-yl)aniline as potent stabilizers of c-MYC G-quadruplex, and their further evolution may lead to the development of an efficient tool for cancer treatment.
Collapse
Affiliation(s)
- Alice Pomeislová
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
- Department of Organic ChemistryCharles UniversityPragueCzech Republic
| | - Lukáš Vrzal
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
- NMR laboratoryUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Jaroslav Kozák
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
| | - Juraj Dobiaš
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
| | - Martin Hubálek
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
| | - Hana Dvořáková
- NMR laboratoryUniversity of Chemistry and TechnologyPragueCzech Republic
| | - Paul E. Reyes‐Gutiérrez
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
| | - Filip Teplý
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
| | - Václav Veverka
- Institute of Organic Chemistry and BiochemistryThe Czech Academy of Sciences Flemingovo nam. 2PragueCzech Republic
- Department of Cell BiologyCharles UniversityPragueCzech Republic
| |
Collapse
|
37
|
Mancini F, Unver MY, Elgaher WAM, Jumde VR, Alhayek A, Lukat P, Herrmann J, Witte MD, Köck M, Blankenfeldt W, Müller R, Hirsch AKH. Protein-Templated Hit Identification through an Ugi Four-Component Reaction*. Chemistry 2020; 26:14585-14593. [PMID: 32428268 PMCID: PMC7756422 DOI: 10.1002/chem.202002250] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Indexed: 12/21/2022]
Abstract
Kinetic target-guided synthesis represents an efficient hit-identification strategy, in which the protein assembles its own inhibitors from a pool of complementary building blocks via an irreversible reaction. Herein, we pioneered an in situ Ugi reaction for the identification of novel inhibitors of a model enzyme and binders for an important drug target, namely, the aspartic protease endothiapepsin and the bacterial β-sliding clamp DnaN, respectively. Highly sensitive mass-spectrometry methods enabled monitoring of the protein-templated reaction of four complementary reaction partners, which occurred in a background-free manner for endothiapepsin or with a clear amplification of two binders in the presence of DnaN. The Ugi products we identified show low micromolar activity on endothiapepsin or moderate affinity for the β-sliding clamp. We succeeded in expanding the portfolio of chemical reactions and biological targets and demonstrated the efficiency and sensitivity of this approach, which can find application on any drug target.
Collapse
Affiliation(s)
- Federica Mancini
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - M. Yagiz Unver
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 79747AGGroningenThe Netherlands
| | - Walid A. M. Elgaher
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
| | - Varsha R. Jumde
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
| | - Alaa Alhayek
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
| | - Peer Lukat
- Department of Structure and Function of ProteinsHZI38124BraunschweigGermany
| | - Jennifer Herrmann
- Department of Microbial Natural ProductsHIPS–HZI66123SaarbrückenGermany
| | - Martin D. Witte
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 79747AGGroningenThe Netherlands
| | - Matthias Köck
- Department of Microbial Natural ProductsHIPS–HZI66123SaarbrückenGermany
| | - Wulf Blankenfeldt
- Department of Structure and Function of ProteinsHZI38124BraunschweigGermany
- Institute for Biochemistry, Biotechnology and BioinformaticsTechnische Universität BraunschweigSpielmannstr. 738106BraunschweigGermany
| | - Rolf Müller
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
- Department of Microbial Natural ProductsHIPS–HZI66123SaarbrückenGermany
| | - Anna K. H. Hirsch
- Department for Drug Design and OptimizationHelmholtz Institute for Pharmaceutical Research Saarland (HIPS)–, Helmholtz Centre for Infection Research (HZI)Campus Building E8.166123SaarbrückenGermany
- Department of PharmacySaarland UniversityCampus Building E8.166123SaarbrückenGermany
- Stratingh Institute for ChemistryUniversity of GroningenNijenborgh 79747AGGroningenThe Netherlands
| |
Collapse
|
38
|
Chowdhury R, Yu Z, Tong ML, Kohlhepp SV, Yin X, Mendoza A. Decarboxylative Alkyl Coupling Promoted by NADH and Blue Light. J Am Chem Soc 2020; 142:20143-20151. [PMID: 33125842 PMCID: PMC7705967 DOI: 10.1021/jacs.0c09678] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Photoexcited dihydronicotinamides like NADH and analogues have been found to generate alkyl radicals upon reductive decarboxylation of redox-active esters without auxiliary photocatalysts. This principle allowed aliphatic photocoupling between redox-active carboxylate derivatives and electron-poor olefins, displaying surprising water and air-tolerance and unusually high coupling rates in dilute conditions. The orthogonality of the reaction in the presence of other carboxylic acids and its utility in the functionalization of DNA is presented, notably using visible light in combination with NADH, the ubiquitous reductant of life.
Collapse
Affiliation(s)
- Rajdip Chowdhury
- Department of Organic Chemistry, Arrhenius laboratory, Stockholm University, 10691 Stockholm Sweden
| | - Zhunzhun Yu
- Department of Organic Chemistry, Arrhenius laboratory, Stockholm University, 10691 Stockholm Sweden
| | - My Linh Tong
- Department of Organic Chemistry, Arrhenius laboratory, Stockholm University, 10691 Stockholm Sweden
| | - Stefanie V Kohlhepp
- Department of Organic Chemistry, Arrhenius laboratory, Stockholm University, 10691 Stockholm Sweden
| | - Xiang Yin
- Department of Organic Chemistry, Arrhenius laboratory, Stockholm University, 10691 Stockholm Sweden
| | - Abraham Mendoza
- Department of Organic Chemistry, Arrhenius laboratory, Stockholm University, 10691 Stockholm Sweden
| |
Collapse
|
39
|
Assunção ELF, Carvalho DB, das Neves AR, Kawasoko Shiguemotto CY, Portapilla GB, de Albuquerque S, Baroni ACM. Synthesis and Antitrypanosomal Activity of 1,4-Disubstituted Triazole Compounds Based on a 2-Nitroimidazole Scaffold: a Structure-Activity Relationship Study. ChemMedChem 2020; 15:2019-2028. [PMID: 32729242 DOI: 10.1002/cmdc.202000460] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Indexed: 12/14/2022]
Abstract
Chagas disease affects 6-8 million people worldwide, remaining a public health concern. Toxicity, several adverse effects and inefficiency in the chronic stage of the disease are the major challenges regarding the available treatment protocols. This work involved the synthesis of twenty-two 1,4-disubstituted-1,2,3-triazole analogues of benznidazole (BZN), by using a click chemistry strategy. Analogues were obtained in moderate to good yields (40-97 %). Antitrypanosomal activity was evaluated against the amastigote forms of Trypanosoma cruzi. Compound 8 a (4-(2-nitro-1H-imidazol-1-yl)methyl)-1-phenyl-1H-1,2,3-triazole) without substituents on phenyl ring showed similar biological activity to BZN (IC50 =3.0 μM, SI>65.3), with an IC50 =3.1 μM and SI>64.5. Compound 8 o (3,4-di-OCH3 -Ph) with IC50 = 0.65 μM was five-fold more active than BZN, and showed an excellent selectivity index (SI>307.7). Compound 8 v (3-NO2 , 4-CH3 -Ph) with IC50 =1.2 μM and relevant SI>166.7, also exhibited higher activity than BZN. SAR analysis exhibited a pattern regarding antitrypanosomal activity relative to BZN, in compounds with electron-withdrawing groups (Hammett σ+) at position 3, and electron-donating groups (Hammett σ-) at position 4, as observed in 8 o and 8 v. Further research might explore in vivo antitrypanosomal activity of promising analogues 8 a, 8 o, and 8 v. Overall, this study indicates that approaches such as the bioisosteric replacement of amide group by 1,2,3-triazole ring, the use of click chemistry as a synthesis strategy, and design tools like Craig-plot and Topliss tree are promising alternatives to drug discovery.
Collapse
Affiliation(s)
- Elvis L F Assunção
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul - UFMS, Campo Grande, Mato Grosso do Sul CEP, 79051-470, Brazil
| | - Diego B Carvalho
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul - UFMS, Campo Grande, Mato Grosso do Sul CEP, 79051-470, Brazil
| | - Amarith R das Neves
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul - UFMS, Campo Grande, Mato Grosso do Sul CEP, 79051-470, Brazil
| | - Cristiane Y Kawasoko Shiguemotto
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul - UFMS, Campo Grande, Mato Grosso do Sul CEP, 79051-470, Brazil
| | - Gisele B Portapilla
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo,CEP, 14040-900, Brazil
| | - Sergio de Albuquerque
- Departamento de Análises Clínicas, Toxicológicas e Bromatológicas, Faculdade de Ciências Farmacêuticas de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, São Paulo,CEP, 14040-900, Brazil
| | - Adriano C M Baroni
- Laboratório de Síntese e Química Medicinal (LASQUIM), Faculdade de Ciências Farmacêuticas, Alimentos e Nutrição, Universidade Federal do Mato Grosso do Sul - UFMS, Campo Grande, Mato Grosso do Sul CEP, 79051-470, Brazil
| |
Collapse
|
40
|
Parthiban V, Yen PYM, Uruma Y, Lai PS. Designing Synthetic Glycosylated Photosensitizers for Photodynamic Therapy. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020. [DOI: 10.1246/bcsj.20200079] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- Venkatesan Parthiban
- Department of Chemistry, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung City 402, Taiwan (R.O.C.)
| | - Priscilla Yoong Mei Yen
- Department of Materials Science, National Institute of Technology, Yonago College, Yonago, Tottori 683-8502, Japan
| | - Yoshiyuki Uruma
- Department of Materials Science, National Institute of Technology, Yonago College, Yonago, Tottori 683-8502, Japan
| | - Ping-Shan Lai
- Department of Chemistry, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung City 402, Taiwan (R.O.C.)
| |
Collapse
|
41
|
Bächle F, Maichle-Mössmer C, Ziegler T. Helical Self-Assembly of Optically Active Glycoconjugated Phthalocyanine J-Aggregates. Chempluschem 2020; 84:1081-1093. [PMID: 31943966 DOI: 10.1002/cplu.201900381] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/18/2019] [Indexed: 01/26/2023]
Abstract
Four galactoconjugated zinc(II) phthalocyanines (Pcs) have been prepared and fully characterized. The carbohydrate-containing phthalonitrile precursors of the Pcs were synthesized through a copper-catalysed azide-alkyne cycloaddition (CuAAC). The Pcs show a remarkable aggregation behaviour in solution, depending on the nature of the solvent, the temperature and the substitution position on the phthalocyanine. Solvent-dependent CD-spectroscopy experiments show that these Pcs aggregate as chiral helices in solution. Crystal structure data of a phthalocyanine bearing two carbohydrate units substantiate the properties shown by CD spectroscopy. Furthermore, the 1,2,3-triazole moieties of the Pcs play a decisive role in the formation of supramolecular aggregates. The glycoconjugated zinc(II) phthalocyanines described here show molar extinction coefficients ϵmax >105 M-1 cm-1 and absorption maxima λmax >680 nm, which make them attractive photosensitizers for Photodynamic Therapy (PDT).
Collapse
Affiliation(s)
- Felix Bächle
- Institute of Organic Chemistry, University of Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany
| | - Cäcilia Maichle-Mössmer
- Institute of Inorganic Chemistry, University of Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany
| | - Thomas Ziegler
- Institute of Organic Chemistry, University of Tübingen, Auf der Morgenstelle 18, 72076, Tübingen, Germany
| |
Collapse
|
42
|
Paul R, Dutta D, Paul R, Dash J. Target-Directed Azide-Alkyne Cycloaddition for Assembling HIV-1 TAR RNA Binding Ligands. Angew Chem Int Ed Engl 2020; 59:12407-12411. [PMID: 32329147 PMCID: PMC7687225 DOI: 10.1002/anie.202003461] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Indexed: 01/05/2023]
Abstract
The highly conserved HIV-1 transactivation response element (TAR) binds to the trans-activator protein Tat and facilitates viral replication in its latent state. The inhibition of Tat-TAR interactions by selectively targeting TAR RNA has been used as a strategy to develop potent antiviral agents. Therefore, HIV-1 TAR RNA represents a paradigmatic system for therapeutic intervention. Herein, we have employed biotin-tagged TAR RNA to assemble its own ligands from a pool of reactive azide and alkyne building blocks. To identify the binding sites and selectivity of the ligands, the in situ cycloaddition has been further performed using control nucleotide (TAR DNA and TAR RNA without bulge) templates. The hit triazole-linked thiazole peptidomimetic products have been isolated from the biotin-tagged target templates using streptavidin beads. The major triazole lead generated by the TAR RNA presumably binds in the bulge region, shows specificity for TAR RNA over TAR DNA, and inhibits Tat-TAR interactions.
Collapse
Affiliation(s)
- Rakesh Paul
- School of Chemical SciencesIndian Association for the Cultivation of ScienceJadavpurKolkata700 032India
| | - Debasish Dutta
- School of Chemical SciencesIndian Association for the Cultivation of ScienceJadavpurKolkata700 032India
| | - Raj Paul
- School of Chemical SciencesIndian Association for the Cultivation of ScienceJadavpurKolkata700 032India
| | - Jyotirmayee Dash
- School of Chemical SciencesIndian Association for the Cultivation of ScienceJadavpurKolkata700 032India
| |
Collapse
|
43
|
Multicomponent click reactions catalysed by copper(I) oxide nanoparticles (Cu2ONPs) derived using Oryza sativa. J CHEM SCI 2020. [DOI: 10.1007/s12039-020-01774-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
44
|
Faeza Abdul Kareem Almashal, Al-Hujaj HH, Jassem AM, Al-Masoudi NA. A Click Synthesis, Molecular Docking, Cytotoxicity on Breast Cancer (MDA-MB 231) and Anti-HIV Activities of New 1,4-Disubstituted-1,2,3-Triazole Thymine Derivatives. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2020. [DOI: 10.1134/s1068162020030024] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
45
|
Hernández-López H, Leyva-Ramos S, Azael Gómez-Durán CF, Pedraza-Alvarez A, Rodríguez-Gutiérrez IR, Leyva-Peralta MA, Razo-Hernández RS. Synthesis of 1,4-Biphenyl-triazole Derivatives as Possible 17β-HSD1 Inhibitors: An in Silico Study. ACS OMEGA 2020; 5:14061-14068. [PMID: 32566872 PMCID: PMC7301541 DOI: 10.1021/acsomega.0c01519] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 05/15/2020] [Indexed: 05/12/2023]
Abstract
Triazoles occupy an important position in medicinal chemistry because of their various biological activities. The structural features of 1,2,3-triazoles enable them to act as a bioisostere of different functional groups such as amide, ester, carboxylic acid, and heterocycle, being capable of forming hydrogen bonds and π-π interactions or coordinate metal ions with biological targets. In this work, the synthesis of 1,2,3-triazole derivatives via copper(I)-catalyzed azide-alkyne cycloaddition (CuAAC) is reported. Overexpression of 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1) is often found in breast cancer cells. Molecular similarity and docking analysis were used to evaluate the potential inhibitory activity of 1,2,3-triazoles synthesized over 17β-HSD1 for the treatment of mammary tumors. Our in silico analysis shows that compounds 4c, 4d, 4f, 4g, and 4j are good molecular scaffold candidates as 17β-HSD1 inhibitors.
Collapse
Affiliation(s)
- Hiram Hernández-López
- Unidad
Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Carretera Zacatecas-Guadalajara Km 6, Ejido la
Escondida s/n, Zacatecas, Zacatecas 98160, México
| | - Socorro Leyva-Ramos
- Facultad
de Ciencias Químicas, Universidad
Autónoma de San Luis Potosí, Av. Manuel Nava 6, Zona Universitaria, San Luis Potosí, San Luis Potosí 78210, México
- . Phone: +52 444 826
2300 ext. 6476
| | - Cesar Fernando Azael Gómez-Durán
- Facultad
de Ciencias Químicas, Universidad
Autónoma de San Luis Potosí, Av. Manuel Nava 6, Zona Universitaria, San Luis Potosí, San Luis Potosí 78210, México
| | - Alberto Pedraza-Alvarez
- Facultad
de Ciencias Químicas, Universidad
Autónoma de San Luis Potosí, Av. Manuel Nava 6, Zona Universitaria, San Luis Potosí, San Luis Potosí 78210, México
| | - Irving Rubén Rodríguez-Gutiérrez
- Facultad
de Ciencias Químicas, Universidad
Autónoma de San Luis Potosí, Av. Manuel Nava 6, Zona Universitaria, San Luis Potosí, San Luis Potosí 78210, México
| | - Mario Alberto Leyva-Peralta
- Departamento
de Ciencias Químico Biológicas y Agropecuarias, Universidad de Sonora, URN Campus Caborca. Av. Universidad e Irigoyen
s/n. H. Caborca, Sonora 83621, México
| | - Rodrigo Said Razo-Hernández
- Centro
de Investigación en Dinámica Celular, Universidad Autónoma del Estado de Morelos. Av. Universidad 1001. Cuernavaca, Morelos 62209, México
| |
Collapse
|
46
|
Paul R, Dutta D, Paul R, Dash J. Target‐Directed Azide‐Alkyne Cycloaddition for Assembling HIV‐1 TAR RNA Binding Ligands. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202003461] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Rakesh Paul
- School of Chemical Sciences Indian Association for the Cultivation of Science Jadavpur Kolkata 700 032 India
| | - Debasish Dutta
- School of Chemical Sciences Indian Association for the Cultivation of Science Jadavpur Kolkata 700 032 India
| | - Raj Paul
- School of Chemical Sciences Indian Association for the Cultivation of Science Jadavpur Kolkata 700 032 India
| | - Jyotirmayee Dash
- School of Chemical Sciences Indian Association for the Cultivation of Science Jadavpur Kolkata 700 032 India
| |
Collapse
|
47
|
Novel amphiphilic dextran esters with antimicrobial activity. Int J Biol Macromol 2020; 150:746-755. [PMID: 32035962 DOI: 10.1016/j.ijbiomac.2020.02.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 11/21/2022]
Abstract
New amphiphilic dextran esters were obtained by polysaccharide functionalization with different substituted 1,2,3-triazoles-4-carboxylic acid via in situ activation with N, N'-carbonyldiimidazole. Nitrogen-containing heterocyclic derivatives were achieved by copper(I)-catalyzed cycloaddition reaction between organic azides and ethyl propiolate. Structural characteristics of the compounds were studied by elemental analysis, Fourier transform infrared and nuclear magnetic resonance spectroscopy (1H and 13C-NMR). Thermogravimetric analysis, differential scanning calorimetry and wide-angle X-ray diffraction were used for esters characterization. Properties of polymeric self-associates, formed in aqueous solution, were studied by dynamic light scattering and transmission electron microscopy. The critical aggregation concentration values for dextran esters, determined by fluorescence spectroscopy, were in the range of 4.1-9.5 mg/dL. Antimicrobial activity, investigated for some of the polymers by disc-diffusion method, pointed out that polysaccharide esters were active.
Collapse
|
48
|
Grob NM, Häussinger D, Deupi X, Schibli R, Behe M, Mindt TL. Triazolo-Peptidomimetics: Novel Radiolabeled Minigastrin Analogs for Improved Tumor Targeting. J Med Chem 2020; 63:4484-4495. [PMID: 32302139 DOI: 10.1021/acs.jmedchem.9b01936] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
MG11 is a truncated analog of minigastrin, a peptide with high affinity and specificity toward the cholecystokinin-2 receptor (CCK2R), which is overexpressed by different tumors. Thus, radiolabeled MG11 derivatives have great potential for use in cancer diagnosis and therapy. A drawback of MG11 is its fast degradation by proteases, leading to moderate tumor uptake in vivo. We introduced 1,4-disubstituted 1,2,3-triazoles as metabolically stable bioisosteres to replace labile amide bonds of the peptide. The "triazole scan" yielded peptidomimetics with improved resistance to enzymatic degradation and/or enhanced affinity toward the CCK2R. Remarkably, our lead compound achieved a 10-fold increase in receptor affinity, resulting in a 2.6-fold improved tumor uptake in vivo. Modeling of the ligand-CCK2R complex suggests that an additional cation-π interaction of the aromatic triazole moiety with the Arg356 residue of the receptor is accountable for these observations. We show for the first time that the amide-to-triazole substitution strategy offers new opportunities in drug development that go beyond the metabolic stabilization of bioactive peptides.
Collapse
Affiliation(s)
- Nathalie M Grob
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zürich, Switzerland
| | - Daniel Häussinger
- Department of Chemistry, University of Basel, 4056 Basel, Switzerland
| | - Xavier Deupi
- Condensed Matter Theory Group, Laboratory for Scientific Computing and Modelling, Paul Scherrer Institute, 5232 Villigen, Switzerland.,Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Roger Schibli
- Department of Chemistry and Applied Biosciences, ETH Zurich, 8093 Zürich, Switzerland.,Center for Radiopharmaceutical Sciences, Division of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Martin Behe
- Center for Radiopharmaceutical Sciences, Division of Biology and Chemistry, Paul Scherrer Institute, 5232 Villigen, Switzerland
| | - Thomas L Mindt
- Ludwig Boltzmann Institute Applied Diagnostics, General Hospital of Vienna, 1090 Vienna, Austria.,Department of Inorganic Chemistry, Faculty of Chemistry, University of Vienna, 1090 Vienna, Austria.,Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, 1090 Vienna, Austria
| |
Collapse
|
49
|
Rani A, Singh G, Singh A, Maqbool U, Kaur G, Singh J. CuAAC-ensembled 1,2,3-triazole-linked isosteres as pharmacophores in drug discovery: review. RSC Adv 2020; 10:5610-5635. [PMID: 35497465 PMCID: PMC9049420 DOI: 10.1039/c9ra09510a] [Citation(s) in RCA: 139] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 01/08/2020] [Indexed: 12/21/2022] Open
Abstract
The review lays emphasis on the significance of 1,2,3-triazoles synthesized via CuAAC reaction having potential to act as anti-microbial, anti-cancer, anti-viral, anti-inflammatory, anti-tuberculosis, anti-diabetic, and anti-Alzheimer drugs. The importance of click chemistry is due to its 'quicker' methodology that has the capability to create complex and efficient drugs with high yield and purity from simple and cheap starting materials. The activity of different triazolyl compounds was compiled considering MIC, IC50, and EC50 values against different species of microbes. In addition to this, the anti-oxidant property of triazolyl compounds have also been reviewed and discussed.
Collapse
Affiliation(s)
- Alisha Rani
- Department of Chemistry, Lovely Professional University Phagwara-144411 Punjab India +91 9815967272
| | - Gurjaspreet Singh
- Department of Chemistry, Centre of Advanced Studies in Chemistry, Panjab University Chandigarh-160014 India
| | - Akshpreet Singh
- Department of Chemistry, Centre of Advanced Studies in Chemistry, Panjab University Chandigarh-160014 India
| | - Ubair Maqbool
- Department of Chemistry, Lovely Professional University Phagwara-144411 Punjab India +91 9815967272
| | - Gurpreet Kaur
- Department of Chemistry, Gujranwala Guru Nanak Khalsa College Civil Lines Ludhiana-141001 India
| | - Jandeep Singh
- Department of Chemistry, Lovely Professional University Phagwara-144411 Punjab India +91 9815967272
| |
Collapse
|
50
|
Verma NK, Mondal D, Bera S. Pharmacological and Cellular Significance of Triazole-Surrogated Compounds. CURR ORG CHEM 2020. [DOI: 10.2174/1385272823666191021114906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
:
Heterocyclic compounds have been at the hierarchy position in academia, and
industrial arena, particularly the compounds containing triazole-core are found to be potent
with a broad range of biological activities. The resistance of triazole ring towards
chemical (acid and base) hydrolysis, oxidative and reductive reaction conditions, metabolic
degradation and its higher aromatic stabilization energy makes it a better heterocyclic
core as therapeutic agents. These triazole-linked compounds are used for clinical purposes
for antifungal, anti-mycobacterium, anticancer, anti-migraine and antidepressant
drugs. Triazole scaffolds are also found to act as a spacer for the sake of covalent attachment
of the high molecular weight bio-macromolecules with an experimental building
blocks to explore structure-function relationships. Herein, several methods and strategies
for the synthesis of compounds with 1,2,3-triazole moiety exploring Hüisgen, Meldal and Sharpless 1,3-dipolar
cycloaddition reaction between azide and alkyne derivatives have been deliberated for a series of representative
compounds. Moreover, this review article highlights in-depth applications of the [3+2]-cycloaddition reaction
for the advances of triazole-containing antibacterial as well as metabolic labelling agents for the in vitro and in
vivo studies on cellular level.
Collapse
Affiliation(s)
- Naimish Kumar Verma
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar-382030, India
| | - Dhananjoy Mondal
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar-382030, India
| | - Smritilekha Bera
- School of Chemical Sciences, Central University of Gujarat, Gandhinagar-382030, India
| |
Collapse
|