1
|
Bonomini A, Mercorelli B, Loregian A. Antiviral strategies against influenza virus: an update on approved and innovative therapeutic approaches. Cell Mol Life Sci 2025; 82:75. [PMID: 39945883 PMCID: PMC11825441 DOI: 10.1007/s00018-025-05611-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 01/27/2025] [Accepted: 02/02/2025] [Indexed: 02/16/2025]
Abstract
Influenza viruses still represent a great concern for Public Health by causing yearly seasonal epidemics and occasionally worldwide pandemics. Moreover, spillover events at the animal-human interface are becoming more frequent nowadays, also involving animal species not previously found as reservoirs. To restrict the effects of influenza virus epidemics, especially in at-risk population, and to prepare a drug arsenal for possible future pandemics, researchers worldwide have been working on the development of antiviral strategies since the 80's of the last century. One of the main obstacles is the considerable genomic variability of influenza viruses, which constantly poses the issues of drug-resistance emergence and immune evasion. This review summarizes the approved therapeutics for clinical management of influenza, promising new anti-flu compounds and monoclonal antibodies currently undergoing clinical evaluation, and molecules with efficacy against influenza virus in preclinical studies. Moreover, we discuss some innovative anti-influenza therapeutic approaches such as combination therapies and targeted protein degradation. Given the limited number of drugs approved for influenza treatment, there is a still strong need for novel potent anti-influenza drugs endowed with a high barrier to drug resistance and broad-spectrum activity against influenza viruses of animal origin that may be responsible of future large outbreaks and pandemics.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | | | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
- Microbiology and Virology Unit, Padua University Hospital, Padua, Italy.
| |
Collapse
|
2
|
Bonomini A, Felicetti T, Pacetti M, Bertagnin C, Coletti A, Giammarino F, De Angelis M, Poggialini F, Macchiarulo A, Sabatini S, Mercorelli B, Nencioni L, Vicenti I, Dreassi E, Cecchetti V, Tabarrini O, Loregian A, Massari S. Optimization of potent, broad-spectrum, and specific anti-influenza compounds targeting RNA polymerase PA-PB1 heterodimerization. Eur J Med Chem 2024; 277:116737. [PMID: 39153334 DOI: 10.1016/j.ejmech.2024.116737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/19/2024]
Abstract
Influenza viruses (IV) are single-stranded RNA viruses with a negative-sense genome and have the potential to cause pandemics. While vaccines exist for influenza, their protection is only partial. Additionally, there is only a limited number of approved anti-IV drugs, which are associated to emergence of drug resistance. To address these issues, for years we have focused on the development of small-molecules that can interfere with the heterodimerization of PA and PB1 subunits of the IV RNA-dependent RNA polymerase (RdRP). In this study, starting from a cycloheptathiophene-3-carboxamide compound that we recently identified, we performed iterative cycles of medicinal chemistry optimization that led to the identification of compounds 43 and 45 with activity in the nanomolar range against circulating A and B strains of IV. Mechanistic studies demonstrated the ability of 43 and 45 to interfere with viral RdRP activity by disrupting PA-PB1 subunits heterodimerization and to bind to the PA C-terminal domain through biophysical assays. Most important, ADME studies of 45 also showed an improvement in the pharmacokinetic profile with respect to the starting hit.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Martina Pacetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Alice Coletti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Federica Giammarino
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Marta De Angelis
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185, Rome, Italy
| | - Federica Poggialini
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Antonio Macchiarulo
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | | | - Lucia Nencioni
- Department of Public Health and Infectious Diseases, Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University of Rome, 00185, Rome, Italy
| | - Ilaria Vicenti
- Department of Medical Biotechnologies, University of Siena, 53100, Siena, Italy
| | - Elena Dreassi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100, Siena, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy.
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy.
| |
Collapse
|
3
|
Bonomini A, Zhang J, Ju H, Zago A, Pacetti M, Tabarrini O, Massari S, Liu X, Mercorelli B, Zhan P, Loregian A. Synergistic activity of an RNA polymerase PA-PB1 interaction inhibitor with oseltamivir against human and avian influenza viruses in cell culture and in ovo. Antiviral Res 2024; 230:105980. [PMID: 39117284 DOI: 10.1016/j.antiviral.2024.105980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 08/10/2024]
Abstract
In search of novel therapeutic options to treat influenza virus (IV) infections, we previously identified a series of inhibitors that act by disrupting the interactions between the PA and PB1 subunits of the viral RNA polymerase. These compounds showed broad-spectrum antiviral activity against human influenza A and B viruses and a high barrier to the induction of drug resistance in vitro. In this short communication, we investigated the effects of combinations of the PA-PB1 interaction inhibitor 54 with oseltamivir carboxylate (OSC), zanamivir (ZA), favipiravir (FPV), and baloxavir marboxil (BXM) on the inhibition of influenza A and B virus replication in vitro. We observed a synergistic effect of the 54/OSC and 54/ZA combinations and an antagonistic effect when 54 was combined with either FPV or BXM. Moreover, we demonstrated the efficacy of 54 against highly pathogenic avian influenza viruses (HPAIVs) both in cell culture and in the embryonated chicken eggs model. Finally, we observed that 54 enhances OSC protective effect against HPAIV replication in the embryonated eggs model. Our findings represent an advance in the development of alternative therapeutic strategies against both human and avian IV infections.
Collapse
Affiliation(s)
- Anna Bonomini
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China
| | - Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China
| | - Alessia Zago
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Martina Pacetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China.
| | | | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Shandong University, 250012, Jinan, Shandong, PR China.
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
4
|
Campbell JA, Do P, Li Z, Malik F, Mead C, Miller N, Pisiechko C, Powers K, Li Z. Synthesis and biological studies of 2-aminothiophene derivatives as positive allosteric modulators of glucagon-like peptide 1 receptor. Bioorg Med Chem 2024; 111:117864. [PMID: 39116711 DOI: 10.1016/j.bmc.2024.117864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/24/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024]
Abstract
As a step toward the development of novel small-molecule positive allosteric modulators (PAMs) of glucagon-like peptide 1 receptor (GLP-1R) for the treatment of type 2 diabetes, obesity, and heart diseases, we discovered a novel 2-amino-thiophene (2-AT) based lead compound bearing an ethyl 3-carboxylate appendage. In this work, we report the syntheses and biological studies of more than forty 2-AT analogs, that have revealed a 2-aminothiophene-3-arylketone analogue 7 (MW 299) showing approximately a 2-fold increase in insulin secretion at 5 μM when combined with the GLP-1 peptide at 10 nM. In vivo studies using CD1 mice at a dose of 10 mg/kg, clearly demonstrated that the blood plasma glucose level was lowered by 50% after 60 min. Co-treatment of 7 with sitagliptin, an inhibitor of GLP-1 degrading enzyme Dipeptidyl Peptidase IV, further confirmed 7 to be an effective PAM of GLP-1R. The small molecular weight and demonstrated allosteric modulating properties of these compound series, show the potential of these scaffolds for future drug development.
Collapse
Affiliation(s)
- Jeffrey A Campbell
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Phu Do
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Zhiyu Li
- Department of Pharmaceutical Sciences, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Faisal Malik
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Christopher Mead
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Nick Miller
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Christopher Pisiechko
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Kimberly Powers
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA
| | - Zhijun Li
- Department of Chemistry and Biochemistry, Saint Joseph's University, Philadelphia, PA 19104, USA.
| |
Collapse
|
5
|
Giacchello I, Cianciusi A, Bertagnin C, Bonomini A, Francesconi V, Mori M, Carbone A, Musumeci F, Loregian A, Schenone S. Exploring a New Generation of Pyrimidine and Pyridine Derivatives as Anti-Influenza Agents Targeting the Polymerase PA-PB1 Subunits Interaction. Pharmaceutics 2024; 16:954. [PMID: 39065650 PMCID: PMC11279468 DOI: 10.3390/pharmaceutics16070954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/08/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
The limited range of available flu treatments due to virus mutations and drug resistance have prompted the search for new therapies. RNA-dependent RNA polymerase (RdRp) is a heterotrimeric complex of three subunits, i.e., polymerase acidic protein (PA) and polymerase basic proteins 1 and 2 (PB1 and PB2). It is widely recognized as one of the most promising anti-flu targets because of its critical role in influenza infection and high amino acid conservation. In particular, the disruption of RdRp complex assembly through protein-protein interaction (PPI) inhibition has emerged as a valuable strategy for discovering a new therapy. Our group previously identified the 3-cyano-4,6-diphenyl-pyridine core as a privileged scaffold for developing PA-PB1 PPI inhibitors. Encouraged by these findings, we synthesized a small library of pyridine and pyrimidine derivatives decorated with a thio-N-(m-tolyl)acetamide side chain (compounds 2a-n) or several amino acid groups (compounds 3a-n) at the C2 position. Interestingly, derivative 2d, characterized by a pyrimidine core and a phenyl and 4-chloro phenyl ring at the C4 and C6 positions, respectively, showed an IC50 value of 90.1 μM in PA-PB1 ELISA, an EC50 value of 2.8 μM in PRA, and a favorable cytotoxic profile, emerging as a significant breakthrough in the pursuit of new PPI inhibitors. A molecular modeling study was also completed as part of this project, allowing us to clarify the biological profile of these compounds.
Collapse
Affiliation(s)
- Ilaria Giacchello
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (I.G.); (A.C.); (V.F.); (S.S.)
| | - Annarita Cianciusi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (I.G.); (A.C.); (V.F.); (S.S.)
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (C.B.); (A.B.); (A.L.)
| | - Anna Bonomini
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (C.B.); (A.B.); (A.L.)
| | - Valeria Francesconi
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (I.G.); (A.C.); (V.F.); (S.S.)
| | - Mattia Mori
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy;
| | - Anna Carbone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (I.G.); (A.C.); (V.F.); (S.S.)
| | - Francesca Musumeci
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (I.G.); (A.C.); (V.F.); (S.S.)
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Via A. Gabelli 63, 35121 Padua, Italy; (C.B.); (A.B.); (A.L.)
| | - Silvia Schenone
- Department of Pharmacy, University of Genoa, Viale Benedetto XV 3, 16132 Genoa, Italy; (I.G.); (A.C.); (V.F.); (S.S.)
| |
Collapse
|
6
|
Fatima A, Khanum G, Srivastava SK, Bhattacharya P, Ali A, Arora H, Siddiqui N, Javed S. Exploring quantum computational, molecular docking, and molecular dynamics simulation with MMGBSA studies of ethyl-2-amino-4-methyl thiophene-3-carboxylate. J Biomol Struct Dyn 2023; 41:10411-10429. [PMID: 37942665 DOI: 10.1080/07391102.2023.2180667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/10/2022] [Indexed: 02/24/2023]
Abstract
2-aminothiophenes derivative, Ethyl-2-amino-4-methyl thiophene-3-carboxylate (EAMC) has been synthesized, characterized, and investigated quantum chemically. It was experimentally investigated by different spectroscopic methods like- NMR (1H-NMR and 13C-NMR), FT-IR, and UV-Visible. B3LYP method and 6-311++G(d,p) basis set were employed for optimization of molecular structure and calculation of wave numbers of normal modes of vibrations and various other important parameters. Calculated bond lengths and angles were compared with the experimental bond lengths and Bond Angle Parameters. Optimized bond parameters and experimental bond parameters were found in good agreement. Complete potential energy distribution assignments were done successfully by VEDA. The HOMO/LUMO energy gap emphasizes adequate charge transfer happening within the molecule. A study of donor-acceptor interconnections was done via NBO analysis. MEP surface analysis was done to demonstrate charge distribution and reactive areas qualitatively in the molecule. The degree of relative localization of electrons was analyzed via ELF Diagram. The Fukui function analysis showed possible sites for attacks by different substituents. By using the TD-DFT method and PCM solvent model, the UV-Vis spectrum (gas, methanol, DMSO) and the maximum absorption wavelength was computed and compared with experimental data. 3D and 2D intermolecular interactions in the crystal were analyzed via Hirshfeld surface analysis and fingerprint plots reveal that the EAMC crystal was stabilized by H--H/H--H/C--H bond formation. The molecular docking was done with 7 different protein receptors on the molecule to find the best ligand-protein interactions. Molecular dynamic simulations and MMGBSA calculations were also carried out to find out the best binding of the ligand with the protein.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Aysha Fatima
- S.O.S in Chemistry, Jiwaji University, Gwalior, India
- Department of Chemistry, CMP College, University of Allahabad, Prayagraj, India
| | | | | | | | - Akram Ali
- Department of Chemistry, CMP College, University of Allahabad, Prayagraj, India
| | - Himanshu Arora
- Department of Chemistry, University of Allahabad, Prayagraj, India
| | - Nazia Siddiqui
- Department of Chemistry, Dayalbagh Educational Institute, Agra, India
| | - Saleem Javed
- Department of Chemistry, University of Allahabad, Prayagraj, India
| |
Collapse
|
7
|
Liu X, Xu Z, Liang J, Xu T, Zou W, Zhu L, Wu Y, Dong C, Lan K, Wu S, Zhou HB. Rational design and optimization of acylthioureas as novel potent influenza virus non-nucleoside polymerase inhibitors. Eur J Med Chem 2023; 259:115678. [PMID: 37531746 DOI: 10.1016/j.ejmech.2023.115678] [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: 03/15/2023] [Revised: 07/12/2023] [Accepted: 07/23/2023] [Indexed: 08/04/2023]
Abstract
Evidence suggests that rapidly evolving virus subvariants risk rendering current vaccines and anti-influenza drugs ineffective. Hence, exploring novel scaffolds or new targets of anti-influenza drugs is of great urgency. Herein, we report the discovery of a series of acylthiourea derivatives produced via a scaffold-hopping strategy as potent antiviral agents against influenza A and B subtypes. The most effective compound 10m displayed subnanomolar activity against H1N1 proliferation (EC50 = 0.8 nM) and exhibited inhibitory activity toward other influenza strains, including influenza B virus and H1N1 variant (H1N1, H274Y). Additionally, druggability evaluation revealed that 10m exhibited favorable pharmacokinetic properties and was metabolically stable in liver microsome preparations from three different species as well as in human plasma. In vitro and in vivo toxicity studies confirmed that 10m demonstrated a high safety profile. Furthermore, 10m exhibited satisfactory antiviral activity in a lethal influenza virus mouse model. Moreover, mechanistic studies indicated that these acylthiourea derivatives inhibited influenza virus proliferation by targeting influenza virus RNA-dependent RNA polymerase. Thus, 10m is a potential lead compound for the further exploration of treatment options for influenza.
Collapse
Affiliation(s)
- Xinjin Liu
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Zhichao Xu
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Jinsen Liang
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ting Xu
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Wenting Zou
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Lijun Zhu
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Yihe Wu
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Chune Dong
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| | - Ke Lan
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Shuwen Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China.
| | - Hai-Bing Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China; Frontier Science Center for Immunology and Metabolism, State Key Laboratory of Virology, Provincial Key Laboratory of Developmentally Originated Disease, Key Laboratory of Combinatorial Biosynthesis and Drug Discovery (MOE) and Hubei Province Engineering and Technology Research Center for Fluorinated Pharmaceuticals, Wuhan University, Wuhan, 430071, China.
| |
Collapse
|
8
|
Bertagnin C, Messa L, Pavan M, Celegato M, Sturlese M, Mercorelli B, Moro S, Loregian A. A small molecule targeting the interaction between human papillomavirus E7 oncoprotein and cellular phosphatase PTPN14 exerts antitumoral activity in cervical cancer cells. Cancer Lett 2023; 571:216331. [PMID: 37532093 DOI: 10.1016/j.canlet.2023.216331] [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: 04/27/2023] [Revised: 07/21/2023] [Accepted: 07/30/2023] [Indexed: 08/04/2023]
Abstract
Human papillomavirus (HPV)-induced cancers still represent a major health issue for worldwide population and lack specific therapeutic regimens. Despite substantial advancements in anti-HPV vaccination, the incidence of HPV-related cancers remains high, thus there is an urgent need for specific anti-HPV drugs. The HPV E7 oncoprotein is a major driver of carcinogenesis that acts by inducing the degradation of several host factors. A target is represented by the cellular phosphatase PTPN14 and its E7-mediated degradation was shown to be crucial in HPV oncogenesis. Here, by exploiting the crystal structure of E7 bound to PTPN14, we performed an in silico screening of small-molecule compounds targeting the C-terminal CR3 domain of E7 involved in the interaction with PTPN14. We discovered a compound able to inhibit the E7/PTPN14 interaction in vitro and to rescue PTPN14 levels in cells, leading to a reduction in viability, proliferation, migration, and cancer-stem cell potential of HPV-positive cervical cancer cells. Mechanistically, as a consequence of PTPN14 rescue, treatment of cancer cells with this compound altered the Yes-associated protein (YAP) nuclear-cytoplasmic shuttling and downstream signaling. Notably, this compound was active against cervical cancer cells transformed by different high-risk (HR)-HPV genotypes indicating a potential broad-spectrum activity. Overall, our study reports the first-in-class inhibitor of E7/PTPN14 interaction and provides the proof-of-principle that pharmacological inhibition of this interaction by small-molecule compounds could be a feasible therapeutic strategy for the development of novel antitumoral drugs specific for HPV-associated cancers.
Collapse
Affiliation(s)
- Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Lorenzo Messa
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Matteo Pavan
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Marta Celegato
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Mattia Sturlese
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | | | - Stefano Moro
- Molecular Modeling Section (MMS), Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
9
|
Duvauchelle V, Meffre P, Benfodda Z. Green methodologies for the synthesis of 2-aminothiophene. ENVIRONMENTAL CHEMISTRY LETTERS 2023; 21:597-621. [PMID: 36060495 PMCID: PMC9421116 DOI: 10.1007/s10311-022-01482-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 06/29/2022] [Indexed: 05/16/2023]
Abstract
Pollution and the rising energy demand have prompted the design of new synthetic reactions that meet the principles of green chemistry. In particular, alternative synthesis of 2-aminothiophene have recently focused interest because 2-aminothiophene is a unique 5-membered S-heterocycle and a pharmacophore providing antiprotozoal, antiproliferative, antiviral, antibacterial or antifungal properties. Here, we review new synthetic routes to 2-aminothiophenes, including multicomponent reactions, homogeneously- or heterogeneously-catalyzed reactions, with focus on green pathways.
Collapse
Affiliation(s)
- Valentin Duvauchelle
- CHROME Laboratory, University of Nîmes, Rue du Dr. G. Salan, 30021 Nîmes Cedex 1, France
| | - Patrick Meffre
- CHROME Laboratory, University of Nîmes, Rue du Dr. G. Salan, 30021 Nîmes Cedex 1, France
| | - Zohra Benfodda
- CHROME Laboratory, University of Nîmes, Rue du Dr. G. Salan, 30021 Nîmes Cedex 1, France
| |
Collapse
|
10
|
Radilová K, Zima V, Kráľ M, Machara A, Majer P, Hodek J, Weber J, Brynda J, Strmeň T, Konvalinka J, Kožíšek M. Thermodynamic and structural characterization of an optimized peptide-based inhibitor of the influenza polymerase PA-PB1 subunit interaction. Antiviral Res 2022; 208:105449. [DOI: 10.1016/j.antiviral.2022.105449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/10/2022] [Accepted: 10/13/2022] [Indexed: 11/02/2022]
|
11
|
Astolfi A, Milano F, Palazzotti D, Brea J, Pismataro MC, Morlando M, Tabarrini O, Loza MI, Massari S, Martelli MP, Barreca ML. From Serendipity to Rational Identification of the 5,6,7,8-Tetrahydrobenzo[4,5]thieno[2,3- d]pyrimidin-4(3 H)-one Core as a New Chemotype of AKT1 Inhibitors for Acute Myeloid Leukemia. Pharmaceutics 2022; 14:2295. [PMID: 36365115 PMCID: PMC9698716 DOI: 10.3390/pharmaceutics14112295] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/20/2022] [Accepted: 10/22/2022] [Indexed: 07/30/2023] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous hematopoietic malignancy whose prognosis is globally poor. In more than 60% of AML patients, the PI3K/AKTs/mTOR signaling pathway is aberrantly activated because of oncogenic driver alterations and further enhanced by chemotherapy as a mechanism of drug resistance. Against this backdrop, very recently we have started a multidisciplinary research project focused on AKT1 as a pharmacological target to identify novel anti-AML agents. Indeed, the serendipitous finding of the in-house compound T187 as an AKT1 inhibitor has paved the way to the rational identification of new active small molecules, among which T126 has emerged as the most interesting compound with IC50 = 1.99 ± 0.11 μM, ligand efficiency of 0.35, and a clear effect at low micromolar concentrations on growth inhibition and induction of apoptosis in AML cells. The collected results together with preliminary SAR data strongly indicate that the 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidin-4(3H)-one derivative T126 is worthy of future biological experiments and medicinal chemistry efforts aimed at developing a novel chemical class of AKT1 inhibitors as anti-AML agents.
Collapse
Affiliation(s)
- Andrea Astolfi
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| | - Francesca Milano
- Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Deborah Palazzotti
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| | - Jose Brea
- CIMUS Research Center, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Maria Chiara Pismataro
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| | - Mariangela Morlando
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| | - Maria Isabel Loza
- CIMUS Research Center, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Serena Massari
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| | - Maria Paola Martelli
- Hematology and Clinical Immunology, Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Maria Letizia Barreca
- Department of Pharmaceutical Sciences, “Department of Excellence 2018–2022”, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
12
|
Jia R, Zhang J, Zhang J, Bertagnin C, Bonomini A, Guizzo L, Gao Z, Ji X, Li Z, Liu C, Ju H, Ma X, Loregian A, Huang B, Zhan P, Liu X. Discovery of Novel Boron-Containing N-Substituted Oseltamivir Derivatives as Anti-Influenza A Virus Agents for Overcoming N1-H274Y Oseltamivir-Resistant. Molecules 2022; 27:molecules27196426. [PMID: 36234966 PMCID: PMC9571049 DOI: 10.3390/molecules27196426] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/23/2022] Open
Abstract
To address drug resistance to influenza virus neuraminidase inhibitors (NAIs), a series of novel boron-containing N-substituted oseltamivir derivatives were designed and synthesized to target the 150-cavity of neuraminidase (NA). In NA inhibitory assays, it was found that most of the new compounds exhibited moderate inhibitory potency against the wild-type NAs. Among them, compound 2c bearing 4-(3-boronic acid benzyloxy)benzyl group displayed weaker or slightly improved activities against group-1 NAs (H1N1, H5N1, H5N8 and H5N1-H274Y) compared to that of oseltamivir carboxylate (OSC). Encouragingly, 2c showed 4.6 times greater activity than OSC toward H5N1-H274Y NA. Moreover, 2c exerted equivalent or more potent antiviral activities than OSC against H1N1, H5N1 and H5N8. Additionally, 2c demonstrated low cytotoxicity in vitro and no acute toxicity at the dose of 1000 mg/kg in mice. Molecular docking of 2c was employed to provide a possible explanation for the improved anti-H274Y NA activity, which may be due to the formation of key additional hydrogen bonds with surrounding amino acid residues, such as Arg152, Gln136 and Val149. Taken together, 2c appeared to be a promising lead compound for further optimization.
Collapse
Affiliation(s)
- Ruifang Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Jian Zhang
- Institute of Medical Sciences, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250033, China
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Anna Bonomini
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Laura Guizzo
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xiangkai Ji
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Zhuo Li
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Chuanfeng Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
| | - Xiuli Ma
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 1 Jiaoxiao Road, Jinan 250023, China
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Bing Huang
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 1 Jiaoxiao Road, Jinan 250023, China
- Correspondence: (B.H.); (P.Z.); (X.L.)
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, Jinan 250012, China
- Correspondence: (B.H.); (P.Z.); (X.L.)
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, Jinan 250012, China
- China-Belgium Collaborative Research Center for Innovative Antiviral Drugs of Shandong Province, 44 West Culture Road, Jinan 250012, China
- Correspondence: (B.H.); (P.Z.); (X.L.)
| |
Collapse
|
13
|
Ju H, Hou L, Zhao F, Zhang Y, Jia R, Guizzo L, Bonomini A, Zhang J, Gao Z, Liang R, Bertagnin C, Kong X, Ma X, Kang D, Loregian A, Huang B, Liu X, Zhan P. Iterative Optimization and Structure-Activity Relationship Studies of Oseltamivir Amino Derivatives as Potent and Selective Neuraminidase Inhibitors via Targeting 150-Cavity. J Med Chem 2022; 65:11550-11573. [PMID: 35939763 DOI: 10.1021/acs.jmedchem.1c01970] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With our continuous endeavors in seeking neuraminidase (NA) inhibitors, we reported herein three series of novel oseltamivir amino derivatives with the goal of exploring the druggable chemical space inside the 150-cavity of influenza virus NAs. Among them, around half of the compounds in series C were demonstrated to be better inhibitors against both wild-type and oseltamivir-resistant group-1 NAs than oseltamivir carboxylate (OSC). Notably, compounds 12d, 12e, 15e, and 15i showed more potent or equipotent antiviral activity against H1N1, H5N1, and H5N8 viruses compared to OSC in cellular assays. Furthermore, compounds 12e and 15e exhibited high metabolic stability in human liver microsomes (HLMs) and low inhibitory effect on main cytochrome P450 (CYP) enzymes, as well as low acute/subacute toxicity and certain antiviral efficacy in vivo. Also, pharmacokinetic (PK) and molecular docking studies were performed. Overall, 12e and 15e possess great potential to serve as anti-influenza candidates and are worthy of further investigation.
Collapse
Affiliation(s)
- Han Ju
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Lingxin Hou
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Fabao Zhao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ying Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ruifang Jia
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Laura Guizzo
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Anna Bonomini
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Jiwei Zhang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Zhen Gao
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Ruipeng Liang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Xiujie Kong
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Xiuli Ma
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 202 North Gongye Road, 250100 Jinan, Shandong, P. R. China
| | - Dongwei Kang
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padova, Via Gabelli 63, 35121 Padova, Italy
| | - Bing Huang
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, 202 North Gongye Road, 250100 Jinan, Shandong, P. R. China
| | - Xinyong Liu
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| | - Peng Zhan
- Department of Medicinal Chemistry, Key Laboratory of Chemical Biology (Ministry of Education), School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, 44 West Culture Road, 250012 Jinan, Shandong, P. R. China
| |
Collapse
|
14
|
Discovery of novel SARS-CoV-2 inhibitors targeting the main protease M pro by virtual screenings and hit optimization. Antiviral Res 2022; 204:105350. [PMID: 35688349 PMCID: PMC9172283 DOI: 10.1016/j.antiviral.2022.105350] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/25/2022] [Accepted: 05/30/2022] [Indexed: 11/21/2022]
Abstract
Two years after its emergence, SARS-CoV-2 still represents a serious and global threat to human health. Antiviral drug development usually takes a long time and, to increase the chances of success, chemical variability of hit compounds represents a valuable source for the discovery of new antivirals. In this work, we applied a platform of variably oriented virtual screening campaigns to seek for novel chemical scaffolds for SARS-CoV-2 main protease (Mpro) inhibitors. The study on the resulting 30 best hits led to the identification of a series of structurally unrelated Mpro inhibitors. Some of them exhibited antiviral activity in the low micromolar range against SARS-CoV-2 and other human coronaviruses (HCoVs) in different cell lines. Time-of-addition experiments demonstrated an antiviral effect during the viral replication cycle at a time frame consistent with the inhibition of SARS-CoV-2 Mpro activity. As a proof-of-concept, to validate the pharmaceutical potential of the selected hits against SARS-CoV-2, we rationally optimized one of the hit compounds and obtained two potent SARS-CoV-2 inhibitors with increased activity against Mpro both in vitro and in a cellular context, as well as against SARS-CoV-2 replication in infected cells. This study significantly contributes to the expansion of the chemical variability of SARS-CoV-2 Mpro inhibitors and provides new scaffolds to be exploited for pan-coronavirus antiviral drug development.
Collapse
|
15
|
Zhang C, Tang YS, Meng CR, Xu J, Zhang DL, Wang J, Huang EF, Shaw PC, Hu C. Design, Synthesis, Molecular Docking Analysis and Biological Evaluations of 4-[(Quinolin-4-yl)amino]benzamide Derivatives as Novel Anti-Influenza Virus Agents. Int J Mol Sci 2022; 23:ijms23116307. [PMID: 35682986 PMCID: PMC9181126 DOI: 10.3390/ijms23116307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 05/23/2022] [Accepted: 06/01/2022] [Indexed: 12/04/2022] Open
Abstract
In this study, a series of 4-[(quinolin-4-yl)amino]benzamide derivatives as the novel anti-influenza agents were designed and synthesized. Cytotoxicity assay, cytopathic effect assay and plaque inhibition assay were performed to evaluate the anti-influenza virus A/WSN/33 (H1N1) activity of the target compounds. The target compound G07 demonstrated significant anti-influenza virus A/WSN/33 (H1N1) activity both in cytopathic effect assay (EC50 = 11.38 ± 1.89 µM) and plaque inhibition assay (IC50 = 0.23 ± 0.15 µM). G07 also exhibited significant anti-influenza virus activities against other three different influenza virus strains A/PR/8 (H1N1), A/HK/68 (H3N2) and influenza B virus. According to the result of ribonucleoprotein reconstitution assay, G07 could interact well with ribonucleoprotein with an inhibition rate of 80.65% at 100 µM. Furthermore, G07 exhibited significant activity target PA−PB1 subunit of RNA polymerase according to the PA−PB1 inhibitory activity prediction by the best pharmacophore Hypo1. In addition, G07 was well drug-likeness based on the results of Lipinski’s rule and ADMET prediction. All the results proved that 4-[(quinolin-4-yl)amino]benzamide derivatives could generate potential candidates in discovery of anti-influenza virus agents.
Collapse
Affiliation(s)
- Chao Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
| | - Yun-Sang Tang
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China;
| | - Chu-Ren Meng
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
| | - Jing Xu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
| | - De-Liang Zhang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
| | - Jian Wang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
| | - Er-Fang Huang
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
| | - Pang-Chui Shaw
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong 999077, China;
- Correspondence: (P.-C.S.); (C.H.); Tel.: +86-24-43520246 (C.H.)
| | - Chun Hu
- Key Laboratory of Structure-Based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China; (C.Z.); (C.-R.M.); (J.X.); (D.-L.Z.); (J.W.); (E.-F.H.)
- Correspondence: (P.-C.S.); (C.H.); Tel.: +86-24-43520246 (C.H.)
| |
Collapse
|
16
|
Redij T, McKee JA, Do P, Campbell JA, Ma J, Li Z, Miller N, Srikanlaya C, Zhang D, Hua X, Li Z. 2-Aminothiophene derivatives as a new class of positive allosteric modulators of glucagon-like peptide 1 receptor. Chem Biol Drug Des 2022; 99:857-867. [PMID: 35313084 DOI: 10.1111/cbdd.14039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 02/08/2022] [Accepted: 03/05/2022] [Indexed: 11/29/2022]
Abstract
We report the discovery of two new 2-aminothiophene based small molecule positive allosteric modulators (PAMs) of glucagon-like peptide 1 receptor (GLP-1R) for the treatment of type 2 diabetes. One of the chemotypes, (S-1), has a molecular weight of 239 g/mol, the smallest molecule among all reported GLP-1R PAMs. When combined with GLP-1 peptide, S-1 increased the GLP-1R activity in a dose-dependent manner in a cell-based assay. When combined with the peptide agonist of vasoactive intestinal polypeptide receptor 1 (VIPR1), S-1 showed no specific activity on VIPR1, another class B GPCR present in the same HEK293-CREB cell line. Insulin secretion studies found S-1 combined with GLP-1 increased insulin secretion by 1.5-fold at 5 μM. In a mechanistic study, evidence is provided that the synergistic effect of S-1 with GLP-1 may be partly due to the enhanced impact on CREB based phosphorylation. Given the favorable profile of these chemotypes, the work reported herein suggests that 2-aminothiophene derivatives are a new and promising class of GLP-1R PAMs.
Collapse
Affiliation(s)
- Tejashree Redij
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - James A McKee
- Department of Chemistry & Biochemistry, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - Phu Do
- Department of Chemistry & Biochemistry, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jeffrey A Campbell
- Department of Chemistry & Biochemistry, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - Jian Ma
- Department of Cancer Biology, Diabetes Research Center (DRC), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zhiyu Li
- Department of Pharmaceutical Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - Nicholas Miller
- Department of Chemistry & Biochemistry, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - Chananchida Srikanlaya
- Department of Chemistry & Biochemistry, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| | - Dianzheng Zhang
- Department of Bio-Medical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, Pennsylvania, USA
| | - Xianxin Hua
- Department of Cancer Biology, Diabetes Research Center (DRC), University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Zhijun Li
- Department of Biological Sciences, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA.,Department of Chemistry & Biochemistry, University of the Sciences in Philadelphia, Philadelphia, Pennsylvania, USA
| |
Collapse
|
17
|
Duvauchelle V, Meffre P, Benfodda Z. Recent contribution of medicinally active 2-aminothiophenes: A privileged scaffold for drug discovery. Eur J Med Chem 2022; 238:114502. [DOI: 10.1016/j.ejmech.2022.114502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 02/01/2023]
|
18
|
Hou L, Zhang Y, Ju H, Cherukupalli S, Jia R, Zhang J, Huang B, Loregian A, Liu X, Zhan P. Contemporary medicinal chemistry strategies for the discovery and optimization of influenza inhibitors targeting vRNP constituent proteins. Acta Pharm Sin B 2022; 12:1805-1824. [PMID: 35847499 PMCID: PMC9279641 DOI: 10.1016/j.apsb.2021.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Revised: 11/02/2021] [Accepted: 11/12/2021] [Indexed: 11/21/2022] Open
Abstract
Influenza is an acute respiratory infectious disease caused by the influenza virus, affecting people globally and causing significant social and economic losses. Due to the inevitable limitations of vaccines and approved drugs, there is an urgent need to discover new anti-influenza drugs with different mechanisms. The viral ribonucleoprotein complex (vRNP) plays an essential role in the life cycle of influenza viruses, representing an attractive target for drug design. In recent years, the functional area of constituent proteins in vRNP are widely used as targets for drug discovery, especially the PA endonuclease active site, the RNA-binding site of PB1, the cap-binding site of PB2 and the nuclear export signal of NP protein. Encouragingly, the PA inhibitor baloxavir has been marketed in Japan and the United States, and several drug candidates have also entered clinical trials, such as favipiravir. This article reviews the compositions and functions of the influenza virus vRNP and the research progress on vRNP inhibitors, and discusses the representative drug discovery and optimization strategies pursued.
Collapse
|
19
|
Zhang C, Xiang JJ, Zhao J, Meng YL, Zhang FR, Jin Z, Shaw PC, Liu XP, Hu C. Design, synthesis, and biological activity of a novel series of 2-ureidonicotinamide derivatives against influenza A virus. Curr Med Chem 2022; 29:4610-4627. [PMID: 35209813 DOI: 10.2174/0929867329666220224114627] [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: 11/17/2021] [Revised: 01/01/2022] [Accepted: 01/07/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Viral resistance to existing inhibitors and the time-dependent effectiveness of neuraminidase inhibitors have limited the number of antivirals that can be used for prophylaxis and therapeutic treatment of severe influenza infection. Thus, there is an urgent need to develop new drugs to prevent and treat influenza infection. OBJECTIVE The aim of this study was to design and synthesize a novel series of 2-ureidonicotinamide derivatives, and evaluate their anti-IAV activities. Furthermore, we predicted the abilities of these compounds inhibiting PA-PB1 subunit and forecasted the docking poses of these compounds with RNA polymerase protein (PDB ID 3CM8). METHOD The novel designed compounds were synthesized using classical methods of organic chemistry and tested in vitro for their abilities inhibiting RNP and against influenza A virus. In addition, the 23 synthesized molecules were subjected to the generated pharmacophore Hypo1 to forecast the activity target PA-PB1 subunit of RNA polymerase. The ADMET pharmacokinetic parameters were calculated by the ADMET modules in Discovery Studio 2016. The docking results helped us to demonstrate the possible interactions between these compounds with 3CM8. RESULTS The synthesized 2-ureidonicotinamide derivatives were characterized as potent anti-influenza inhibitors. The target compounds 7b and 7c demonstrated significant antiviral activities, and could be considered as novel lead compounds of antiviral inhibitors. In addition, compound 7b revealed suitable ADME properties expressed, and might be a significant RNA polymerase inhibitor targeting PA-PB1 subunit based on the predictable results and the docking results. CONCLUSION This study revealed a novel series of compounds that might be useful in the search for an effective drug against influenza virus.
Collapse
Affiliation(s)
- Chao Zhang
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jun-Jie Xiang
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jing Zhao
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Yan-Li Meng
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Fu-Rong Zhang
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Zhe Jin
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Pang-Chui Shaw
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xiao-Ping Liu
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Chun Hu
- Key Laboratory of Structure-based Drug Design & Discovery, Ministry of Education, Shenyang Pharmaceutical University, Shenyang 110016, China
| |
Collapse
|
20
|
Synthesis and Evaluation of RNase L-binding 2-aminothiophenes as anticancer agents. Bioorg Med Chem 2022; 58:116653. [DOI: 10.1016/j.bmc.2022.116653] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/31/2022] [Accepted: 02/01/2022] [Indexed: 01/08/2023]
|
21
|
Duvauchelle V, Bénimélis D, Meffre P, Benfodda Z. Catalyst-Free Site Selective Hydroxyalkylation of 5-Phenylthiophen-2-amine with α-Trifluoromethyl Ketones through Electrophilic Aromatic Substitution. Molecules 2022; 27:molecules27030925. [PMID: 35164190 PMCID: PMC8839828 DOI: 10.3390/molecules27030925] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 01/26/2022] [Accepted: 01/27/2022] [Indexed: 12/10/2022] Open
Abstract
An original and effective approach for achieving trifluoromethyl hydroxyalkylation of 5-phenylthiophen-2-amine using α-trifluoromethyl ketones is described. In the last few years, reaction of Friedel-Crafts had been widely used to realize hydroxyalkylation on heterocycles such as indoles or thiophenes by means of Lewis acid as catalyst. Additionally, amine functions are rarely free when carbonyl reagents are used because of their tendency to form imines. This is the first time that a site-selective electrophilic aromatic substitution on C3 atom of an unprotected 5-phenylthiophen-2-amine moiety is reported. The liberty to allow reaction in neutral conditions between free amine is valuable in a synthesis pathway. The reaction proceeds smoothly using an atom-economical metal-and catalyst-free methodology in good to excellent yields. A mechanism similar to an electrophilic aromatic substitution has been proposed.
Collapse
|
22
|
Mizuta S, Otaki H, Ishikawa T, Makau JN, Yamaguchi T, Fujimoto T, Takakura N, Sakauchi N, Kitamura S, Nono H, Nishi R, Tanaka Y, Takeda K, Nishida N, Watanabe K. Lead Optimization of Influenza Virus RNA Polymerase Inhibitors Targeting PA-PB1 Interaction. J Med Chem 2021; 65:369-385. [PMID: 34905383 DOI: 10.1021/acs.jmedchem.1c01527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Influenza viruses are responsible for contagious respiratory illnesses in humans and cause seasonal epidemics and occasional pandemics worldwide. Previously, we identified a quinolinone derivative PA-49, which inhibited the influenza virus RNA-dependent RNA polymerase (RdRp) by targeting PA-PB1 interaction. This paper reports the structure optimization of PA-49, which resulted in the identification of 3-((dibenzylamino)methyl)quinolinone derivatives with more potent anti-influenza virus activity. During the optimization, the hit compound 89, which was more active than PA-49, was identified. Further optimization and scaffold hopping of 89 led to the most potent compounds 100 and a 1,8-naphthyridinone derivative 118, respectively. We conclusively determined that compounds 100 and 118 suppressed the replication of influenza virus and exhibited anti-influenza virus activity against both influenza virus types A and B in the range of 50% effective concentration (EC50) = 0.061-0.226 μM with low toxicity (50% cytotoxic concentration (CC50) >10 μM).
Collapse
Affiliation(s)
- Satoshi Mizuta
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo, Nagasaki 852-8521, Japan
| | - Hiroki Otaki
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo, Nagasaki 852-8521, Japan
| | - Takeshi Ishikawa
- Department of Chemistry, Biotechnology, and Chemical Engineering, Graduate School of Science and Engineering, Kagoshima University, 1-21-40 Korimoto, Kagoshima 890-0065, Japan
| | - Juliann Nzembi Makau
- Center for Virus Research, Kenya Medical Research Institute, 54840-00200 Nairobi, Kenya
| | - Tomoko Yamaguchi
- Center for Bioinformatics and Molecular Medicine, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo, Nagasaki 852-8521, Japan
| | - Takuya Fujimoto
- Chemistry, Discovery Science, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-0012, Japan
| | - Nobuyuki Takakura
- Chemistry, Discovery Science, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-0012, Japan
| | - Nobuki Sakauchi
- Chemistry, Discovery Science, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-0012, Japan
| | - Shuji Kitamura
- Chemistry, Discovery Science, Axcelead Drug Discovery Partners, Inc., 26-1, Muraoka-Higashi 2-chome, Fujisawa, Kanagawa 251-0012, Japan
| | - Hikaru Nono
- School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Ryota Nishi
- School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Kohsuke Takeda
- Department of Cell Regulation, Graduate School of Biomedical Sciences, Nagasaki University, 1-14 Bunkyo, Nagasaki 852-8521, Japan
| | - Noriyuki Nishida
- Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Ken Watanabe
- Department of Lifestyle Design, Faculty of Human Ecology, Yasuda Women's University, 6-13-1 Yasuhigashi, Asaminami Ward, Hiroshima 731-0153, Japan
| |
Collapse
|
23
|
Massari S, Desantis J, Nizi MG, Cecchetti V, Tabarrini O. Inhibition of Influenza Virus Polymerase by Interfering with Its Protein-Protein Interactions. ACS Infect Dis 2021; 7:1332-1350. [PMID: 33044059 PMCID: PMC8204303 DOI: 10.1021/acsinfecdis.0c00552] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Influenza (flu) virus is a serious threat to global health with the potential to generate devastating pandemics. The availability of broad spectrum antiviral drugs is an unequaled weapon during pandemic events, especially when a vaccine is still not available. One of the most promising targets for the development of new antiflu therapeutics is the viral RNA-dependent RNA polymerase (RdRP). The assembly of the flu RdRP heterotrimeric complex from the individual polymerase acidic protein (PA), polymerase basic protein 1 (PB1), and polymerase basic protein 2 (PB2) subunits is a prerequisite for RdRP functions, such as mRNA synthesis and genome replication. In this Review, we report the known protein-protein interactions (PPIs) occurring by RdRP that could be disrupted by small molecules and analyze their benefits and drawbacks as drug targets. An overview of small molecules able to interfere with flu RdRP functions exploiting the PPI inhibition approach is described. In particular, an update on the most recent inhibitors targeting the well-consolidated RdRP PA-PB1 subunit heterodimerization is mainly reported, together with pioneer inhibitors targeting other virus-virus or virus-host interactions involving RdRP subunits. As demonstrated by the PA-PB1 interaction inhibitors discussed herein, the inhibition of flu RdRP functions by PPI disrupters clearly represents a valid means to identify compounds endowed with a broad spectrum of action and a reduced propensity to develop drug resistance, which are the main issues of antiviral drugs.
Collapse
Affiliation(s)
- Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy
| | - Jenny Desantis
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy
| | - Maria Giulia Nizi
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123 Perugia, Italy
| |
Collapse
|
24
|
Asiri YI, Muhsinah AB, Alsayari A, Venkatesan K, Al-Ghorbani M, Mabkhot YN. Design, synthesis and antimicrobial activity of novel 2-aminothiophene containing cyclic and heterocyclic moieties. Bioorg Med Chem Lett 2021; 44:128117. [PMID: 34015500 DOI: 10.1016/j.bmcl.2021.128117] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/08/2021] [Accepted: 05/16/2021] [Indexed: 11/19/2022]
Abstract
One of the major challenges in the community and healthcare was an impedance of pathogenic bacteria to antibiotics. This work developed 2-aminothiophene derivatives as novel antimicrobial agents. Various 2-aminothiophene derivatives (3a-f, 5a-c, 6a, b, 7, 8a, b and 9) with cyclic and heterocyclic moieties at 5-position were synthesized, and characterized using NMR, IR, and mass spectroscopic techniques. The newly synthesized compounds were evaluated for their antimicrobial activity against bacteria S. pneumoniae, B. subtilis, P. aeruginosa, E. coli, and fungi A. fumigatus, S. mracemosum, G. candidum, C. albicans. Compound 3a with OH group at para position of phenyl ring exhibited significant antibacterial activity stronger than that of the drug standards Ampicillin and Gentamicin. Compound 6b possess pyrazole ring and compound 9 bearing pyridine ring showed promising antifungal activity compare to the standard drug Amphotericin B. The remaining compounds exhibited good to moderate inhibitory activities. In summary, the results suggest that the compounds from 2-aminothiophene derivatives can be used as antimicrobial agents.
Collapse
Affiliation(s)
- Yahya I Asiri
- Department of Pharmacology, College of Pharmacy, King Khalid University, P.O. Box 960, Abha 61421, Saudi Arabia.
| | - Abdullatif Bin Muhsinah
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Abdulrhman Alsayari
- Department of Pharmacognosy, College of Pharmacy, King Khalid University, Abha, Saudi Arabia.
| | - Kumar Venkatesan
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia.
| | - Mohammed Al-Ghorbani
- Department of Chemistry, College of Science and Arts, Ulla, Taibah University, Madina Monora, Saudi Arabia; Department of Chemistry, College of Education, Thamar University, Thamar, Yemen
| | - Yahia N Mabkhot
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Khalid University, Abha 61441, Saudi Arabia.
| |
Collapse
|
25
|
Terrier O, Slama-Schwok A. Anti-Influenza Drug Discovery and Development: Targeting the Virus and Its Host by All Possible Means. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1322:195-218. [PMID: 34258742 DOI: 10.1007/978-981-16-0267-2_8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Infections by influenza virus constitute a major and recurrent threat for human health. Together with vaccines, antiviral drugs play a key role in the prevention and treatment of influenza virus infection and disease. Today, the number of antiviral molecules approved for the treatment of influenza is relatively limited, and their use is threatened by the emergence of viral strains with resistance mutations. There is therefore a real need to expand the prophylactic and therapeutic arsenal. This chapter summarizes the state of the art in drug discovery and development for the treatment of influenza virus infections, with a focus on both virus-targeting and host cell-targeting strategies. Novel antiviral strategies targeting other viral proteins or targeting the host cell, some of which are based on drug repurposing, may be used in combination to strengthen our therapeutic arsenal against this major pathogen.
Collapse
Affiliation(s)
- Olivier Terrier
- CIRI, Centre International de Recherche en Infectiologie, (Team VirPath), Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Anny Slama-Schwok
- Sorbonne Université, Centre de Recherche Saint-Antoine, INSERM U938, Biologie et Thérapeutique du Cancer, Paris, France.
| |
Collapse
|
26
|
Almarhoon Z, Al Rasheed HH, El-Faham A. Ultrasonically Assisted N-Cyanoacylation and Synthesis of Alkyl(4-(3-cyano-4,6-dimethyl-2-oxopyridin-1(2 H)-yl)benzoyl)amino Acid Ester Derivatives. ACS OMEGA 2020; 5:30671-30678. [PMID: 33283115 PMCID: PMC7711942 DOI: 10.1021/acsomega.0c04730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Accepted: 11/04/2020] [Indexed: 06/12/2023]
Abstract
This work represents the use of N-3-(3,5-dimethyl-1H-pyrazol-1-yl)-3-oxopropanenitrile as a cyanoacetylating agent for the synthesis of cyanoacetamide benzoic acid and benzophenone derivatives by two different methods, namely, conventional heating and ultrasonication. The cyanoacetamide derivatives were subjected to cyclization to produce N-substituted 2-pyridone derivatives under conventional heating and by an ultrasonic method as well. The ultrasonic method afforded the products in less reaction time with high yields and purities compared to the conventional method, as observed from their spectral data. N-(4-Carboxy phenyl)-4,6-dimethyl-3-cyano-2-pyridone was coupled with different amino acid esters by the OxymaPure/DIC methodology under traditional and ultrasonic conditions. Again, ultrasonication assisted the coupling step and afforded the products with higher yields and purities compared to the traditional method. Fourier transform infrared spectroscopy, NMR (1H and 13C), elemental analysis, and LC-MS were used to determine the structures of all compounds. Finally, a feature of this protocol is exploring the utilization of ultrasonication as an eco-friendly alternative conventional heating method for N-cyanoacylation and synthesis of N-substituted pyridinone derivatives and as a coupling method for the formation of an amide bond, which might be of interest for many researchers.
Collapse
Affiliation(s)
- Zainab Almarhoon
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Hessa H. Al Rasheed
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
| | - Ayman El-Faham
- Department
of Chemistry, College of Science, King Saud
University, P.O. Box 2455, Riyadh 11451, Saudi Arabia
- Chemistry
Department, Faculty of Science, Alexandria
University, P.O. Box 426, Ibrahimia 12321, Alexandria, Egypt
| |
Collapse
|
27
|
structural characterization of the interaction between the C-terminal domain of the influenza polymerase PA subunit and an optimized small peptide inhibitor. Antiviral Res 2020; 185:104971. [PMID: 33166574 DOI: 10.1016/j.antiviral.2020.104971] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/26/2020] [Accepted: 11/03/2020] [Indexed: 02/07/2023]
Abstract
Influenza viruses can cause severe respiratory infections in humans, leading to nearly half a million deaths worldwide each year. Improved antiviral drugs are needed to address the threat of development of novel pandemic strains. Current therapeutic interventions target three key proteins in the viral life cycle: neuraminidase, the M2 channel and RNA-dependent-RNA polymerase. Protein-protein interactions between influenza polymerase subunits are potential new targets for drug development. Using a newly developed assay based on AlphaScreen technology, we screened a peptide panel for protein-protein interaction inhibitors to identify a minimal PB1 subunit-derived peptide that retains high inhibition potential and can be further modified. Here, we present an X-ray structure of the resulting decapeptide bound to the C-terminal domain of PA polymerase subunit from pandemic isolate A/California/07/2009 H1N1 at 1.6 Å resolution and discuss its implications for the design of specific, potent influenza polymerase inhibitors.
Collapse
|
28
|
Ginex T, Luque FJ. Searching for effective antiviral small molecules against influenza A virus: A patent review. Expert Opin Ther Pat 2020; 31:53-66. [PMID: 33012213 DOI: 10.1080/13543776.2020.1831471] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Introduction: Despite the current interest caused by SARS-Cov-2, influenza continues to be one of the most serious health concerns, with an estimated 1 billion cases across the globe, including 3-5 million severe cases and 290,000-650,000 deaths worldwide. Areas covered: This manuscript reviews the efforts made in the development of small molecules for the treatment of influenza virus, primarily focused on patent applications in the last 5 years. Attention is paid to compounds targeting key functional viral proteins, such as the M2 channel, neuraminidase, and hemagglutinin, highlighting the evolution toward new ligands and scaffolds motivated by the emergence of resistant strains. Finally, the discovery of compounds against novel viral targets, such as the RNA-dependent RNA polymerase, is discussed. Expert opinion: The therapeutic potential of antiviral agents is limited by the increasing presence of resistant strains. This should encourage research on novel strategies for therapeutic intervention. In this context, the discovery of arbidol and JNJ7918 against hemagglutinin, and current efforts on RNA-dependent RNA polymerase have disclosed novel opportunities for therapeutic treatment. Studies should attempt to expand the therapeutic arsenal of anti-flu agents, often in combined therapies, to prevent future health challenges caused by influenza virus. Abbreviations: AlphaLISA: amplified luminescent proximity homogeneous assay; HA: hemagglutinin; NA: neuraminidase; RBD: receptor binding domain; RdRp: RNA-dependent RNA polymerase; SA: sialic Acid; TBHQ: tert-butyl hydroquinone; TEVC: two-electrode voltage clamp.
Collapse
Affiliation(s)
- Tiziana Ginex
- Translational Medicinal and Biological Chemistry Group, Centro de Investigaciones Biológicas Margarita Salas, Consejo Superior de Investigaciones Biológicas (CIB-CSIC) , Madrid, Spain
| | - F Javier Luque
- Department of Nutrition, Food Science and Gastronomy, Faculty of Pharmacy and Food Sciences, Institute of Biomedicine (IBUB), and Institute of Theoretical and Computational Chemistry (IQTCUB), University of Barcelona , Santa Coloma de Gramanet, Spain
| |
Collapse
|
29
|
Massari S, Bertagnin C, Pismataro MC, Donnadio A, Nannetti G, Felicetti T, Di Bona S, Nizi MG, Tensi L, Manfroni G, Loza MI, Sabatini S, Cecchetti V, Brea J, Goracci L, Loregian A, Tabarrini O. Synthesis and characterization of 1,2,4-triazolo[1,5-a]pyrimidine-2-carboxamide-based compounds targeting the PA-PB1 interface of influenza A virus polymerase. Eur J Med Chem 2020; 209:112944. [PMID: 33328103 PMCID: PMC7561591 DOI: 10.1016/j.ejmech.2020.112944] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 01/06/2023]
Abstract
Influenza viruses (Flu) are responsible for seasonal epidemics causing high rates of morbidity, which can dramatically increase during severe pandemic outbreaks. Antiviral drugs are an indispensable weapon to treat infected people and reduce the impact on human health, nevertheless anti-Flu armamentarium still remains inadequate. In search for new anti-Flu drugs, our group has focused on viral RNA-dependent RNA polymerase (RdRP) developing disruptors of PA-PB1 subunits interface with the best compounds characterized by cycloheptathiophene-3-carboxamide and 1,2,4-triazolo[1,5-a]pyrimidine-2-carboxamide scaffolds. By merging these moieties, two very interesting hybrid compounds were recently identified, starting from which, in this paper, a series of analogues were designed and synthesized. In particular, a thorough exploration of the cycloheptathiophene-3-carboxamide moiety led to acquire important SAR insight and identify new active compounds showing both the ability to inhibit PA-PB1 interaction and viral replication in the micromolar range and at non-toxic concentrations. For few compounds, the ability to efficiently inhibit PA-PB1 subunits interaction did not translate into anti-Flu activity. Chemical/physical properties were investigated for a couple of compounds suggesting that the low solubility of compound 14, due to a strong crystal lattice, may have impaired its antiviral activity. Finally, computational studies performed on compound 23, in which the phenyl ring suitably replaced the cycloheptathiophene, suggested that, in addition to hydrophobic interactions, H-bonds enhanced its binding within the PAC cavity.
Collapse
Affiliation(s)
- Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy.
| | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | | | - Anna Donnadio
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Giulio Nannetti
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Tommaso Felicetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Stefano Di Bona
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy
| | - Maria Giulia Nizi
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Leonardo Tensi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy
| | - Giuseppe Manfroni
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Maria Isabel Loza
- CIMUS Research Center, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| | - Jose Brea
- CIMUS Research Center, University of Santiago de Compostela, 15782, Santiago de Compostela, Spain
| | - Laura Goracci
- Department of Chemistry, Biology and Biotechnology, University of Perugia, 06123, Perugia, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, 35121, Padua, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, 06123, Perugia, Italy
| |
Collapse
|
30
|
Hwang J, Borgelt L, Wu P. Multicomponent Petasis Reaction for the Synthesis of Functionalized 2-Aminothiophenes and Thienodiazepines. ACS COMBINATORIAL SCIENCE 2020; 22:495-499. [PMID: 32985878 PMCID: PMC7584359 DOI: 10.1021/acscombsci.0c00173] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
![]()
Multicomponent
Petasis reaction has been widely applied for the synthesis of functionalized
amine building blocks and biologically active compounds. Employing
primary aromatic amines that are not typical reactive substrates contributes
to expand the application scope of the Petasis reaction. In this study,
we demonstrated the synthesis of functionalized 2-aminothiophenes
using Gewald-reaction-derived 2-aminothiophenes as the amine substrates,
whose low reactivity in the Petasis reaction was overcome using hexafluoro-2-propanol
as the solvent in a mild condition. The obtained Petasis products
are amenable for further transformations owing to the presence of
multiple functional handles. A following intramolecular cyclization
of selected Petasis products afforded substituted tricyclic heterocycles
that incorporate a pharmaceutically interesting thienodiazepine moiety.
Collapse
Affiliation(s)
- Jimin Hwang
- Chemical Genomics Centre and Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany
| | - Lydia Borgelt
- Chemical Genomics Centre and Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany
| | - Peng Wu
- Chemical Genomics Centre and Department of Chemical Biology, Max Planck Institute of Molecular Physiology, Dortmund 44227, Germany
| |
Collapse
|
31
|
Recent developments of gallic acid derivatives and their hybrids in medicinal chemistry: A review. Eur J Med Chem 2020; 204:112609. [DOI: 10.1016/j.ejmech.2020.112609] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023]
|
32
|
Singh A, Singh G, Bedi PMS. Thiophene derivatives: A potent multitargeted pharmacological scaffold. J Heterocycl Chem 2020. [DOI: 10.1002/jhet.3990] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Atamjit Singh
- Department of Pharmaceutical SciencesGuru Nanak Dev University Amritsar Punjab India
| | - Gurvinder Singh
- Department of Pharmaceutical ChemistryKhalsa College of Pharmacy Amritsar Punjab India
| | | |
Collapse
|
33
|
Zhang J, Hu Y, Wu N, Wang J. Discovery of Influenza Polymerase PA-PB1 Interaction Inhibitors Using an In Vitro Split-Luciferase Complementation-Based Assay. ACS Chem Biol 2020; 15:74-82. [PMID: 31714745 DOI: 10.1021/acschembio.9b00552] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The limited therapeutic options and increasing drug-resistance call for next-generation influenza antivirals. Due to the essential function in viral replication and high sequence conservation among influenza viruses, influenza polymerase PA-PB1 protein-protein interaction becomes an attractive drug target. Here, we developed an in vitro split luciferase complementation-based assay to speed up screening of PA-PB1 interaction inhibitors. By screening 10,000 compounds, we identified two PA-PB1 interaction inhibitors, R160792 and R151785, with potent and broad-spectrum antiviral activity against a panel of influenza A and B viruses, including amantadine-, oseltamivir-, or dual resistant strains. Further mechanistic study reveals that R151785 inhibits PA nuclear localization, reduces the levels of viral RNAs and proteins, and inhibits viral replication at the intermediate stage, all of which are in line with its antiviral mechanism of action. Overall, we developed a robust high throughput-screening assay for screening broad-spectrum influenza antivirals targeting PA-PB1 interaction and identified R151785 as a promising antiviral drug candidate.
Collapse
Affiliation(s)
- Jiantao Zhang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Yanmei Hu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Nan Wu
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| | - Jun Wang
- Department of Pharmacology and Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
34
|
Nguyen TTT, Le VA, Retailleau P, Nguyen TB. Access to 2‐Amino‐3‐Arylthiophenes by Base‐Catalyzed Redox Condensation Reaction Between Arylacetonitriles, Chalcones, and Elemental Sulfur. Adv Synth Catal 2019. [DOI: 10.1002/adsc.201901235] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Thi Thu Tram Nguyen
- Department of Chemistry, Faculty of ScienceCan Tho University of Medicine and Pharmacy Vietnam
| | - Van Anh Le
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-SudUniversité Paris-Saclay, 1 avenue de la Terrasse 91198 Gif-sur-Yvette France
| | - Pascal Retailleau
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-SudUniversité Paris-Saclay, 1 avenue de la Terrasse 91198 Gif-sur-Yvette France
| | - Thanh Binh Nguyen
- Institut de Chimie des Substances Naturelles, CNRS UPR 2301, Université Paris-SudUniversité Paris-Saclay, 1 avenue de la Terrasse 91198 Gif-sur-Yvette France
| |
Collapse
|
35
|
Mahdavi B, Hosseyni-Tabar SM, Rezaei-Seresht E, Rezaei-Seresht H, Falanji F. Synthesis and biological evaluation of novel hybrid compounds derived from gallic acid and the 2-aminothiophene derivatives. JOURNAL OF THE IRANIAN CHEMICAL SOCIETY 2019. [DOI: 10.1007/s13738-019-01813-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
36
|
Nannetti G, Massari S, Mercorelli B, Bertagnin C, Desantis J, Palù G, Tabarrini O, Loregian A. Potent and broad-spectrum cycloheptathiophene-3-carboxamide compounds that target the PA-PB1 interaction of influenza virus RNA polymerase and possess a high barrier to drug resistance. Antiviral Res 2019; 165:55-64. [PMID: 30885750 DOI: 10.1016/j.antiviral.2019.03.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 02/25/2019] [Accepted: 03/04/2019] [Indexed: 12/17/2022]
Abstract
Influenza viruses are major respiratory pathogens responsible for both seasonal epidemics and occasional pandemics worldwide. The current available treatment options have limited efficacy and thus the development of new antivirals is highly needed. We previously reported the identification of a series of cycloheptathiophene-3-carboxamide compounds as influenza A virus inhibitors that act by targeting the protein-protein interactions between the PA-PB1 subunits of the viral polymerase. In this study, we characterized the antiviral properties of the most promising compounds as well as investigated their propensity to induce drug resistance. Our results show that some of the selected compounds possess potent, broad-spectrum anti-influenza activity as they efficiently inhibited the replication of several strains of influenza A and B viruses, including an oseltamivir-resistant clinical isolate, with nanomolar or low-micromolar potency. The most promising compounds specifically inhibited the PA-PB1 binding in vitro and interfered with the influenza A virus polymerase activity in a cellular context, without showing cytotoxicity. The most active PA-PB1 inhibitors showed to possess a drug resistance barrier higher than that of oseltamivir. Indeed, no viral variants with reduced susceptibility to the selected compounds emerged after serial passages of influenza A virus under drug selective pressure. Overall, our studies identified potent PA-PB1 inhibitors as promising candidates for the development of new anti-influenza drugs.
Collapse
Affiliation(s)
- Giulio Nannetti
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Serena Massari
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | | | - Chiara Bertagnin
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Jenny Desantis
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Giorgio Palù
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences, University of Perugia, Perugia, Italy
| | - Arianna Loregian
- Department of Molecular Medicine, University of Padua, Padua, Italy.
| |
Collapse
|
37
|
Mohl G, Liddle N, Nygaard J, Dorius A, Lyons N, Hodek J, Weber J, Michaelis DJ, Busath DD. Novel influenza inhibitors designed to target PB1 interactions with host importin RanBP5. Antiviral Res 2019; 164:81-90. [PMID: 30742842 DOI: 10.1016/j.antiviral.2019.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Revised: 01/30/2019] [Accepted: 02/03/2019] [Indexed: 11/19/2022]
Abstract
In search of novel targets for influenza inhibitors, a site on PB1 was selected for its high conservation and probable interaction with a host protein, RanBP5, that is key to nuclear import of PB1, where it complexes with PB2, PA, and NP to transcribe viral RNA. Docking with libraries of drug-like compounds led to a selection of five candidates that bound tightly and with a pose likely to inhibit protein binding. These were purchased and tested in vitro, found to be active, and then one was synthetically expanded to explore the structure-activity relationship. The top candidates had a carboxylic acid converted to an ester and electron-withdrawing substituents added to a phenyl group in the original structure. Resistance was slow to develop, but cytotoxicity was moderately high. Nuclear localization of PB1 and in vitro polymerase activity were both strongly inhibited.
Collapse
Affiliation(s)
- Gregory Mohl
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| | - Nathan Liddle
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Joseph Nygaard
- Department of Chemistry and Biochemistry, Brigham Young University, USA
| | - Alexander Dorius
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA
| | - Nathan Lyons
- Department of Chemistry and Biochemistry, Brigham Young University, USA
| | - Jan Hodek
- Institute of Organic Chemistry and Biochemistry of the CAS, Prague, Czech Republic
| | - Jan Weber
- Institute of Organic Chemistry and Biochemistry of the CAS, Prague, Czech Republic
| | - David J Michaelis
- Department of Chemistry and Biochemistry, Brigham Young University, USA.
| | - David D Busath
- Department of Physiology and Developmental Biology, Brigham Young University, Provo, UT 84602, USA.
| |
Collapse
|
38
|
Synthesis and biological evaluation of a library of hybrid derivatives as inhibitors of influenza virus PA-PB1 interaction. Eur J Med Chem 2018; 157:743-758. [PMID: 30142611 DOI: 10.1016/j.ejmech.2018.08.032] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 07/03/2018] [Accepted: 08/11/2018] [Indexed: 11/21/2022]
Abstract
The limited treatment options against influenza virus along with the growing public health concerns regarding the continuous emergence of drug-resistant viruses make essential the development of new anti-flu agents with novel mechanisms of action. One of the most attractive targets is the interaction between two subunits of the RNA-dependent RNA polymerase, PA and PB1. Herein we report the rational design of hybrid compounds starting from a 3-cyano-4,6-diphenylpyridine scaffold recently identified as disruptor of PA-PB1 interactions. Guided by the previously reported SAR data, a library of amino acid derivatives was synthesized. The biological evaluation led to the identification of new PA-PB1 inhibitors, that do not show appreciable toxicity. Molecular modeling shed further lights on the inhibition mechanism of these compounds.
Collapse
|
39
|
2-Aminothiophene scaffolds: Diverse biological and pharmacological attributes in medicinal chemistry. Eur J Med Chem 2017; 140:465-493. [PMID: 28987607 DOI: 10.1016/j.ejmech.2017.09.039] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/02/2017] [Accepted: 09/19/2017] [Indexed: 12/30/2022]
Abstract
2-Aminothiophenes are important five-membered heterocyclic building blocks in organic synthesis, and the chemistry of these small molecules is still developing based on the discovery of cyclization by Gewald. Another attractive feature of 2-aminothiophene scaffolds is their ability to act as synthons for the synthesis of biological active thiophene-containing heterocycles, conjugates and hybrids. Currently, the biological actions of 2-aminothiophenes or their 2-N-substituted analogues are still being investigated because of their various mechanisms of action (e.g., pharmacophore and pharmacokinetic properties). Likewise, the 2-aminothiophene family is used as diverse promising selective inhibitors, receptors, and modulators in medicinal chemistry, and these compounds even exhibit effective pharmacological properties in the various clinical phases of appropriate diseases. In this review, major biological and pharmacological reports on 2-aminothiophenes and related compounds have been highlighted; most perspective drug-candidate hits were selected for discussion and described, along with additional synthetic pathways. In addition, we focused on the literature dedicated to 2-aminothiophenes and 2-N-substituted derivatives, which have been published from 2010 to 2017.
Collapse
|
40
|
Massari S, Desantis J, Nannetti G, Sabatini S, Tortorella S, Goracci L, Cecchetti V, Loregian A, Tabarrini O. Efficient and regioselective one-step synthesis of 7-aryl-5-methyl- and 5-aryl-7-methyl-2-amino-[1,2,4]triazolo[1,5-a]pyrimidine derivatives. Org Biomol Chem 2017; 15:7944-7955. [DOI: 10.1039/c7ob02085f] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Two facile and efficient one-step procedures for the regioselective synthesis of 7-aryl-5-methyl- and 5-aryl-7-methyl-2-amino-[1,2,4]triazolo[1,5-a]pyrimidines.
Collapse
Affiliation(s)
- Serena Massari
- Department of Pharmaceutical Sciences
- University of Perugia
- 06123 Perugia
- Italy
| | - Jenny Desantis
- Department of Pharmaceutical Sciences
- University of Perugia
- 06123 Perugia
- Italy
| | - Giulio Nannetti
- Department of Molecular Medicine
- University of Padua
- 35121 Padua
- Italy
| | - Stefano Sabatini
- Department of Pharmaceutical Sciences
- University of Perugia
- 06123 Perugia
- Italy
| | - Sara Tortorella
- Department of Chemistry
- Biology and Biotechnology
- University of Perugia
- 06123 Perugia
- Italy
| | - Laura Goracci
- Department of Chemistry
- Biology and Biotechnology
- University of Perugia
- 06123 Perugia
- Italy
| | - Violetta Cecchetti
- Department of Pharmaceutical Sciences
- University of Perugia
- 06123 Perugia
- Italy
| | - Arianna Loregian
- Department of Molecular Medicine
- University of Padua
- 35121 Padua
- Italy
| | - Oriana Tabarrini
- Department of Pharmaceutical Sciences
- University of Perugia
- 06123 Perugia
- Italy
| |
Collapse
|