1
|
Liang L, Meng Y, Chang X, Li E, Huang Y, Yan L, Lou Z, Peng Y, Zhu B, Yu W, Chang J. Discovery of a 2'-α-Fluoro-2'-β- C-(fluoromethyl) Purine Nucleotide Prodrug as a Potential Oral Anti-SARS-CoV-2 Agent. J Med Chem 2025; 68:1994-2007. [PMID: 39804580 DOI: 10.1021/acs.jmedchem.4c02769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
A novel 2'-α-fluoro-2'-β-C-(fluoromethyl) purine nucleoside phosphoramidate prodrug 15 has been designed and synthesized to treat SARS-CoV-2 infection. The SARS-CoV-2 central replication transcription complex (C-RTC, nsp12-nsp7-nsp82) catalyzed in vitro RNA synthesis was effectively inhibited by the corresponding bioactive nucleoside triphosphate (13-TP). The cryo-electron microscopy structure of the C-RTC:13-TP complex was also determined. Compound 15 exhibited potent in vitro antiviral activity against the SARS-CoV-2 20SF107 strain (EC50 = 0.56 ± 0.06 μM) and the Omicron BA.5 variant (EC50 = 0.96 ± 0.23 μM) with low cytotoxicity. Furthermore, it was well tolerated in rats at doses of up to 2000 mg/kg, and a single oral dose of this prodrug at 40 mg/kg led to high levels of 13-TP in the target organ lungs of rats with a long half-life. These findings support the further development of compound 15 as an orally available antiviral agent for the treatment of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lan Liang
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, NMPA Key Laboratory for Research and Evaluation of Innovative Drug, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Yonggang Meng
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xiaoyu Chang
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Ertong Li
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yucen Huang
- State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences and College of Pharmacy, Nankai University, Tianjin 300071, China
| | - Liming Yan
- MOE Key Laboratory of Protein Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Zhiyong Lou
- MOE Key Laboratory of Protein Science, School of Medicine, Tsinghua University, Beijing 100084, China
| | - Youmei Peng
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Bo Zhu
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, NMPA Key Laboratory for Research and Evaluation of Innovative Drug, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
| | - Wenquan Yu
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Junbiao Chang
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, NMPA Key Laboratory for Research and Evaluation of Innovative Drug, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang, Henan 453007, China
- State Key Laboratory of Antiviral Drugs, Pingyuan Laboratory, College of Chemistry, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
2
|
Kumar N, Shukla A, Kumar S, Ulasov I, Singh RK, Kumar S, Patel A, Yadav L, Tiwari R, Paswan R, Mohanta SP, Kaushalendra, Antil J, Acharya A. FNC (4'-azido-2'-deoxy-2'-fluoro(arbino)cytidine) as an Effective Therapeutic Agent for NHL: ROS Generation, Cell Cycle Arrest, and Mitochondrial-Mediated Apoptosis. Cell Biochem Biophys 2024; 82:623-639. [PMID: 38253918 DOI: 10.1007/s12013-023-01193-6] [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: 01/19/2023] [Accepted: 10/12/2023] [Indexed: 01/24/2024]
Abstract
Cytotoxic nucleoside analogs (NAs) hold great promise in cancer therapeutics by mimicking endogenous nucleosides and interfering with crucial cellular processes. Here, we investigate the potential of the novel cytidine analog, 4'-azido-2'-deoxy-2'-fluoro(arbino)cytidine (FNC), as a therapeutic agent for Non-Hodgkin lymphoma (NHL) using Dalton's lymphoma (DL) as a T-cell lymphoma model. FNC demonstrated dose- and time-dependent inhibition of DL cell growth and proliferation. IC-50 values of FNC were measured at 1 µM, 0.5 µM, and 0.1 µM after 24, 48, and 72 h, respectively. Further elucidation of FNC's mechanism of action uncovers its role in inducing apoptosis in DL cells. Notable DNA fragmentation and nuclear condensation point to activated apoptotic pathways. FNC-induced apoptosis was concomitant with changes in cellular membranes, characterized by membrane rupture and altered morphology. The robust anticancer effects of FNC are linked to its capacity to induce reactive oxygen species (ROS) production, prompting oxidative stress-mediated apoptosis. Additionally, FNC disrupted mitochondrial membrane potential (MMP), leading to mitochondrial dysfunction, further promoting apoptosis. Dysregulation of apoptotic genes, with upregulation of Bax and downregulation of Bcl-2 and Bcl-xl, implicates the mitochondrial-mediated apoptosis pathway. Furthermore, FNC-induced G2/M phase cell cycle arrest was mediated through modulation of the cell cycle inhibitor p21. Overall, this study highlights the potential of FNC as a promising therapeutic agent for NHL.
Collapse
Affiliation(s)
| | | | | | - Ilya Ulasov
- Sechenov First Moscow State Medical University, Moscow, Russia
| | | | | | | | | | | | | | | | - Kaushalendra
- Pachhunga University College Campus, Mizoram University, Aizawl, India
| | | | | |
Collapse
|
3
|
Hou J, Peng Y, Liu B, Zhang Q, Wang JH, Yu W, Chang J. 4'-Ethynyl-2'-deoxy-2'-β-fluoro-2-fluoroadenosine: A Highly Potent and Orally Available Clinical Candidate for the Treatment of HIV-1 Infection. J Med Chem 2023; 66:11282-11293. [PMID: 37535016 DOI: 10.1021/acs.jmedchem.3c00761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/04/2023]
Abstract
2'-Deoxy-2'-β-fluoroadenosines bearing 4'-azido or 4'-ethynyl groups designed for the treatment of HIV-1 infection have been synthesized. All these compounds possess nanomolar anti-HIV-1 activity, with the 4'-ethynyl-2-fluoroadenosine analog 1c (CL-197) being the most potent compound with low cytotoxicity (EC50 = 0.9 nM, CC50 > 100 μM). It also shows potent inhibitory activities on drug resistant and clinical HIV-1 strains. Oral administration of 1c to Beagle dogs resulted in high levels of its bioactive form 1c-TP in peripheral blood mononuclear cells, the HIV-1 target cells, where the resulting triphosphate exhibited a long-term intracellular retention and could prevent HIV-1 infection for an extended time. 1c displayed low in vivo toxicity and favorable pharmacokinetics profiles in Sprague-Dawley rats. The preclinical data support further development of 1c as a highly potent and orally bioavailable clinical candidate to treat HIV-1 infection. Currently, CL-197 is in clinical trials in China (registration number: CXHL2200529).
Collapse
Affiliation(s)
- Jiao Hou
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Youmei Peng
- Henan Key Laboratory for Pharmacology of Liver Diseases, Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, China
| | - Bingjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Qianqian Zhang
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Jian-Hua Wang
- Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Henan Normal University, Xinxiang 453007, China
| | - Wenquan Yu
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Junbiao Chang
- Green Catalysis Center, and College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
- Pingyuan Laboratory, State Key Laboratory of Antiviral Drugs, Henan Normal University, Xinxiang 453007, China
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
4
|
Yin Q, Song X, Yang P, Yang W, Li X, Wang X, Wang S. Incorporation of glycyrrhizic acid and polyene phosphatidylcholine in lipid nanoparticles ameliorates acute liver injury via delivering p65 siRNA. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2023; 48:102649. [PMID: 36584740 DOI: 10.1016/j.nano.2022.102649] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 12/08/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022]
Abstract
Liver injury caused by hepatitis is the pathological basis of varied hepatic diseases with high morbidity and mortality. Although siRNA appears promising in therapeutics of hepatitis, efficient and safe delivery remains a challenge. In this study, we developed a new strategy of incorporating glycyrrhizic acid (GA) and polyene phosphatidylcholine (PPC) into lipid nanoparticles (GA/PPC-modified LNPs), which was capable of promoting cellular uptake, enhancing gene-silencing, reducing cytotoxicity and improving siRNA stability. GA/PPC-modified LNP and siRNA lipoplex targeting NF-κB, a key mediator of inflammation, mitigates acute liver injury, as assessed by liver histology, hematological and pro-inflammatory cytokine analysis. Furthermore, GA/PPC-modified LNPs reveal efficiently intracellular delivery of antisense oligonucleotides (ASOs) and mRNA inhibiting viral infection. In conclusion, GA/PPC-modified LNPs could be used as a promising delivery system for nucleic acid-based therapy.
Collapse
Affiliation(s)
- Qiming Yin
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, People's Republic of China
| | - Xiang Song
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China.
| | - Peng Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China
| | - Wen Yang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, People's Republic of China
| | - Xinyu Li
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China; Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, People's Republic of China
| | - Xuejun Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China.
| | - Shengqi Wang
- State Key Laboratory of Pathogen and Biosecurity, Beijing Institute of Microbiology and Epidemiology, AMMS, Beijing 100071, People's Republic of China; School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, People's Republic of China.
| |
Collapse
|
5
|
Wang Q, Mu J, Zeng J, Wan L, Zhong Y, Li Q, Li Y, Wang H, Chen F. Additive-controlled asymmetric iodocyclization enables enantioselective access to both α- and β-nucleosides. Nat Commun 2023; 14:138. [PMID: 36627283 PMCID: PMC9831021 DOI: 10.1038/s41467-022-35610-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
β-Nucleosides and their analogs are dominant clinically-used antiviral and antitumor drugs. α-Nucleosides, the anomers of β-nucleosides, exist in nature and have significant potential as drugs or drug carriers. Currently, the most widely used methods for synthesizing β- and α-nucleosides are via N-glycosylation and pentose aminooxazoline, respectively. However, the stereoselectivities of both methods highly depend on the assisting group at the C2' position. Herein, we report an additive-controlled stereodivergent iodocyclization method for the selective synthesis of α- or β-nucleosides. The stereoselectivity at the anomeric carbon is controlled by the additive (NaI for β-nucleosides; PPh3S for α-nucleosides). A series of β- and α-nucleosides are prepared in high yields (up to 95%) and stereoselectivities (β:α up to 66:1, α:β up to 70:1). Notably, the introduced iodine at the C2' position of the nucleoside is readily functionalized, leading to multiple structurally diverse nucleoside analogs, including stavudine, an FDA-approved anti-HIV agent, and molnupiravir, an FDA-approved anti-SARS-CoV-2 agent.
Collapse
Affiliation(s)
- Qi Wang
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jiayi Mu
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Jie Zeng
- Pharmaceutical Research Institute, Wuhan Institute of Technology, 430205, Wuhan, China
| | - Linxi Wan
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yangyang Zhong
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Qiuhong Li
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Yitong Li
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China
| | - Huijing Wang
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Fener Chen
- Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China. .,Engineering Center of Catalysis and synthesis for Chiral Molecules, Department of chemistry, Fudan University, Shanghai, 200433, China. .,Shanghai Engineering Center of Industrial Asymmetric Catalysis for Chiral Drugs, Shanghai, 200433, China.
| |
Collapse
|
6
|
Pal S, Chandra G, Patel S, Singh S. Fluorinated Nucleosides: Synthesis, Modulation in Conformation and Therapeutic Application. CHEM REC 2022; 22:e202100335. [PMID: 35253973 DOI: 10.1002/tcr.202100335] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/22/2022] [Indexed: 12/17/2022]
Abstract
Over the last twenty years, fluorination on nucleoside has established itself as the most promising tool to use to get biologically active compounds that could sustain the clinical trial by affecting the pharmacodynamics and pharmacokinetic properties. Due to fluorine's inherent unique properties and its judicious introduction into the molecule, makes the corresponding nucleoside metabolically very stable, lipophilic, and opens a new site of intermolecular binding. Fluorination on various nucleosides has been extensively studied as a result, a series of fluorinated nucleosides come up for different therapeutic uses which are either approved by the FDA or under the advanced stage of the clinical trial. Here in this review, we are summarizing the latest development in the chemistry of fluorination on nucleoside that led to varieties of new analogs like carbocyclic, acyclic, and conformationally biased nucleoside and their biological properties, the influence of fluorine on conformation, oligonucleotide stability, and their use in therapeutics.
Collapse
Affiliation(s)
- Shantanu Pal
- School of Basic Sciences, Indian Institute of Technology, Bhubaneswar Argul, Odisha, India, 752050
| | - Girish Chandra
- Department of Chemistry, School of Physical and Chemical Sciences, Central University of South Bihar, SH-7, Gaya Panchanpur Road, Gaya, Bihar, India, 824236
| | - Samridhi Patel
- Department of Chemistry, School of Physical and Chemical Sciences, Central University of South Bihar, SH-7, Gaya Panchanpur Road, Gaya, Bihar, India, 824236
| | - Sakshi Singh
- School of Basic Sciences, Indian Institute of Technology, Bhubaneswar Argul, Odisha, India, 752050
| |
Collapse
|
7
|
Chang J. 4'-Modified Nucleosides for Antiviral Drug Discovery: Achievements and Perspectives. Acc Chem Res 2022; 55:565-578. [PMID: 35077644 DOI: 10.1021/acs.accounts.1c00697] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Modified nucleosides show therapeutic promise for antiviral therapies. However, issues including the emergence of drug resistance, toxicity, and coinfections have posed new challenges for nucleoside-based antiviral drug discovery, particularly in the era of the coronavirus disease 2019 (COVID-19) pandemic. Chemical manipulation could impact the antiviral potency, safety, and drug resistance of nucleosides. Generally, modified nucleosides are difficult to recognize by intracellular important enzymes as substrates and thus exhibit low toxicity. 4'-Modified nucleosides represent an important subclass of modified nucleosides for antiviral therapies. To prevent the occurrence of drug resistance, 4'-modified nucleosides should have 3'-OH, which should also be chemically unreactive for proviral DNA biosynthesis. The absence of 3'-OH may explain the occurrence of drug resistance for censavudine. The introduction of 4'-substituents improves enzymatic and acidic stability and makes the nucleosides more lipophilic, thus improving cell permeability and bioavailability. Steric hindrance between the 4'-substituent and 3'-OH changes the furanose conformation to the 3'-endo type, in which the oxygen lone pair on the furanose ring could not form an oxocarbonium ion for glycolysis. Currently, seven 4'-modified nucleoside drug candidates such as azvudine (also known as FNC), islatravir, censavudine, balapiravir, lumicitabine, AL-335, and 4-azidothymidine have progressed into clinical stages for treating viral infections. Of note, FNC was officially approved by NMPA in July 2021 for use in adult patients with high HIV-1 virus loads (nos. H20210035 and H20210036), providing an alternative therapeutic for patients with HIV-1. The long-term cellular retention of FNC suggests its potential as a long-lasting pre-exposure prophylaxis (PrEP) agent for preventing HIV-1 infection. Mechanistically, FNC not only inhibited HIV-1 reverse transcription and replication but also restored A3G expression in peripheral blood CD4+ T cells in HIV-1 patients receiving FNC. The 4'-azido group in azvudine stabilizes the 3'-C-endo (north) conformation by steric effects and the formation of an intramolecular hydrogen bond with the 3'-OH group, thus decreasing the nucleophilicity of 3'-OH. The north conformation may also enhance the phosphorylation efficiency of FNC by cellular kinases. Encouragingly, FNC, islatravir, and balapiravir show promise for the treatment of coronaviruses, of which FNC has advanced to phase 3 clinical trials in different countries to treat patients with COVID-19 (clinical trial numbers: NCT04668235 and NCT04425772). FNC cured the COVID-19 disease in almost all patients and showed better therapeutic efficacy than remdesivir. In this Account, we provide an overview of 4'-modified nucleoside analogs in clinical stages for antiviral therapies, highlighting the drug discovery strategies, structure-activity relationship studies, and preclinical/clinical studies and also give our perspectives on nucleoside-based antiviral drug discovery.
Collapse
Affiliation(s)
- Junbiao Chang
- Henan Key Laboratory of Organic Functional Molecules and Drug Innovation, School of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| |
Collapse
|
8
|
Fayzullina D, Kharwar RK, Acharya A, Buzdin A, Borisov N, Timashev P, Ulasov I, Kapomba B. FNC: An Advanced Anticancer Therapeutic or Just an Underdog? Front Oncol 2022; 12:820647. [PMID: 35223502 PMCID: PMC8867032 DOI: 10.3389/fonc.2022.820647] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022] Open
Abstract
Azvudine (FNC) is a novel cytidine analogue that has both antiviral and anticancer activities. This minireview focuses on its underlying molecular mechanisms of suppressing viral life cycle and cancer cell growth and discusses applications of this nucleoside drug for advanced therapy of tumors and malignant blood diseases. FNC inhibits positive-stand RNA viruses, like HCV, EV, SARS-COV-2, HBV, and retroviruses, including HIV, by suppressing their RNA-dependent polymerase enzymes. It may also inhibit such enzyme (reverse transcriptase) in the human retrotransposons, including human endogenous retroviruses (HERVs). As the activation of retrotransposons can be the major factor of ongoing cancer genome instability and consequently higher aggressiveness of tumors, FNC has a potential to increase the efficacy of multiple anticancer therapies. Furthermore, FNC also showed other aspects of anticancer activity by inhibiting adhesion, migration, invasion, and proliferation of malignant cells. It was also reported to be involved in cell cycle arrest and apoptosis, thereby inhibiting the progression of cancer through different pathways. To the date, the grounds of FNC effects on cancer cells are not fully understood and hence additional studies are needed for better understanding molecular mechanisms of its anticancer activities to support its medical use in oncology.
Collapse
Affiliation(s)
- Daria Fayzullina
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Rajesh Kumar Kharwar
- Endocrine Research Lab, Department of Zoology, Kutir Post Graduate College, Chakkey, Jaunpur, India
| | - Arbind Acharya
- Tumor Immunology Lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Anton Buzdin
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Nicolas Borisov
- Department of Medical and Biological Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Ilya Ulasov
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Byron Kapomba
- Department of General Surgery, Parirenyatwa Group of Hospitals, Harare, Zimbabwe,*Correspondence: Byron Kapomba,
| |
Collapse
|
9
|
Alexandrova LA, Jasko MV, Negrya SD, Solyev PN, Shevchenko OV, Solodinin AP, Kolonitskaya DP, Karpenko IL, Efremenkova OV, Glukhova AA, Boykova YV, Efimenko TA, Kost NV, Avdanina DA, Nuraeva GK, Volkov IA, Kochetkov SN, Zhgun AA. Discovery of novel N 4-alkylcytidines as promising antimicrobial agents. Eur J Med Chem 2021; 215:113212. [PMID: 33582576 DOI: 10.1016/j.ejmech.2021.113212] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/18/2020] [Accepted: 01/12/2021] [Indexed: 10/22/2022]
Abstract
The emergence of drug-resistant strains of pathogenic microorganisms necessitates the creation of new drugs. In order to find new compounds that effectively inhibit the growth of pathogenic bacteria and fungi, we synthesized a set of N4-derivatives of cytidine, 2'-deoxycytidine and 5-metyl-2'-deoxycytidine bearing extended N4-alkyl and N4-phenylalkyl groups. The derivatives demonstrate activity against a number of Gram-positive bacteria, including Mycobacterium smegmatis (MIC = 24-200 μM) and Staphylococcus aureus (MIC = 50-200 μM), comparable with the activities of some antibiotics in medical use. The most promising compound appeared to be N4-dodecyl-5-metyl-2'-deoxycytidine 4h with activities of 24 and 48 μM against M. smegmatis and S. aureus, respectively, and high inhibitory activity of 0.5 mM against filamentous fungi that can, among other things, damage works of art, such as tempera painting. Noteworthy, some of other synthesized compounds are active against fungal growth with the inhibitory concentration in the range of 0.5-3 mM.
Collapse
Affiliation(s)
| | - Maxim V Jasko
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Sergey D Negrya
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Pavel N Solyev
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia.
| | - Oleg V Shevchenko
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Andrei P Solodinin
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Daria P Kolonitskaya
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Inna L Karpenko
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Olga V Efremenkova
- Gause Institute of New Antibiotics, 11 Bol'shaya Pirogovskaya St., 119021, Moscow, Russia
| | - Alla A Glukhova
- Gause Institute of New Antibiotics, 11 Bol'shaya Pirogovskaya St., 119021, Moscow, Russia
| | - Yuliya V Boykova
- Gause Institute of New Antibiotics, 11 Bol'shaya Pirogovskaya St., 119021, Moscow, Russia
| | - Tatiana A Efimenko
- Gause Institute of New Antibiotics, 11 Bol'shaya Pirogovskaya St., 119021, Moscow, Russia
| | - Natalya V Kost
- Federal State Budgetary Scientific Institution "Mental Health Research Centre", 34 Kashirskoe Highway, 115522, Moscow, Russia
| | - Darya A Avdanina
- Research Center of Biotechnology RAS, 33 Leninsky Ave, 119071, Moscow, Russia
| | - Gulgina K Nuraeva
- Moscow Institute of Physics and Technology (National Research University), 9 Institutsky Alley, 141700, Dolgoprudny, Russia
| | - Ivan A Volkov
- Moscow Institute of Physics and Technology (National Research University), 9 Institutsky Alley, 141700, Dolgoprudny, Russia
| | - Sergey N Kochetkov
- Engelhardt Institute of Molecular Biology RAS, 32 Vavilova St., 119991, Moscow, Russia
| | - Alexander A Zhgun
- Research Center of Biotechnology RAS, 33 Leninsky Ave, 119071, Moscow, Russia
| |
Collapse
|
10
|
Sun L, Peng Y, Yu W, Zhang Y, Liang L, Song C, Hou J, Qiao Y, Wang Q, Chen J, Wu M, Zhang D, Li E, Han Z, Zhao Q, Jin X, Zhang B, Huang Z, Chai J, Wang JH, Chang J. Mechanistic Insight into Antiretroviral Potency of 2'-Deoxy-2'-β-fluoro-4'-azidocytidine (FNC) with a Long-Lasting Effect on HIV-1 Prevention. J Med Chem 2020; 63:8554-8566. [PMID: 32678592 DOI: 10.1021/acs.jmedchem.0c00940] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In preclinical and phase I and II clinical studies, 2'-deoxy-2'-β-fluoro-4'-azidocytidine (FNC) displays a potent and long-lasting inhibition of HIV-1 infection. To investigate its mechanism of action, we compared it with the well-documented lamivudine (3TC). Pharmacokinetic studies revealed that the intracellular retention of FNC triphosphate in peripheral blood mononuclear cells was markedly longer than that of the 3TC triphosphate. FNC selectively enters and is retained in HIV target cells, where it exerts long-lasting prevention of HIV-1 infection. In addition to inhibition of HIV-1 reverse transcription, FNC also restores A3G expression in CD4+ T cells in FNC-treated HIV-1 patients. FNC binds to the Vif-E3 ubiquitin ligase complex, enabling A3G to avoid Vif-induced ubiquitination and degradation. These data reveal the mechanisms underlying the superior anti-HIV potency and long-lasting action of FNC. Our results also suggest a potential clinical application of FNC as a long-lasting pre-exposure prophylactic agent capable of preventing HIV infection.
Collapse
Affiliation(s)
- Li Sun
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Youmei Peng
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wenquan Yu
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Zhang
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China
| | - Lan Liang
- College of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| | - Chuanjun Song
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Jiao Hou
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Yan Qiao
- College of Chemistry, Zhengzhou University, Zhengzhou 450001, China
| | - Qingduan Wang
- Henan Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Jingyu Chen
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Mengli Wu
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Dongwei Zhang
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China.,Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Ertong Li
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Zhifu Han
- Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Qingxia Zhao
- Department of Infection, Zhengzhou Sixth People's Hospital, Zhengzhou 450000, China
| | - Xia Jin
- Shanghai Public Health Clinical Center Affiliated to Fudan University, Shanghai 201508, China
| | - Bailing Zhang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Zhiwei Huang
- HIT Center for Life Sciences, School of Life Science and Technology, Harbin Institute of Technology, Harbin 150080, China
| | - Jijie Chai
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China.,Tsinghua-Peking Center for Life Sciences, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Jian-Hua Wang
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou 510530, China
| | - Junbiao Chang
- Henan Key Laboratory of Organic Functional Molecule and Drug Innovation, College of Life Science, Henan Normal University, Xinxiang 453007, China.,College of Chemistry, Zhengzhou University, Zhengzhou 450001, China.,College of Chemistry and Chemical Engineering, Henan Normal University, Xinxiang 453007, China
| |
Collapse
|
11
|
Qiu J, Zhou Q, Zhang Y, Guan M, Li X, Zou Y, Huang X, Zhao Y, Chen W, Gu X. Discovery of novel quinazolinone derivatives as potential anti-HBV and anti-HCC agents. Eur J Med Chem 2020; 205:112581. [PMID: 32791397 DOI: 10.1016/j.ejmech.2020.112581] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 11/29/2022]
Abstract
As a continuation of earlier works, a series of novel quinazolinone derivatives (5a-s) were synthesized and evaluated for their in vitro anti-HBV and anti-hepatocellular carcinoma cell (HCC) activities. Among them, compounds 5j and 5k exhibited most potent inhibitory effect on HBV DNA replication in both drug sensitive and resistant (lamivudine and entecavir) HBV strains. Interestingly, besides the anti-HBV effect, compound 5k could significantly inhibit the proliferation of HepG2, HUH7 and SK- cells, with IC50 values of 5.44, 6.42 and 6.75 μM, respectively, indicating its potential anti-HCC activity. Notably, the in vitro anti-HCC activity of 5k were more potent than that of positive control 5-fluorouracil and sorafenib. Further studies revealed that compound 5k could induce HepG2 cells apoptosis by dose-dependently upregulating Bad and Bax expression and decreasing Bcl-2 and Bcl-xl protein level. Considering the potent anti-HBV and anti-HCC effect, compound 5k might be a promising lead to develop novel therapeutic agents towards HBV infection and HBV-induced HCC.
Collapse
Affiliation(s)
- Jingying Qiu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China; Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Qingqing Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yinpeng Zhang
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Mingyu Guan
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xin Li
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yueting Zou
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xuan Huang
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Yali Zhao
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Wang Chen
- Department of Pharmaceutical Analysis, School of Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China
| | - Xiaoke Gu
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Xuzhou Medical University, Xuzhou, 221004, People's Republic of China.
| |
Collapse
|
12
|
Onitsuka K, Tokuda R, Kuwata-Higashi N, Kumamoto H, Aoki M, Amano M, Kohgo S, Das D, Haraguchi K, Mitsuya H, Imoto S. Synthesis and evaluation of the anti-hepatitis B virus activity of 4'-Azido-thymidine analogs and 4'-Azido-2'-deoxy-5-methylcytidine analogs: structural insights for the development of a novel anti-HBV agent. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2019; 39:518-529. [PMID: 31514570 DOI: 10.1080/15257770.2019.1664749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Hepatitis B virus (HBV) infection is a major worldwide health problem that requires the development of improved antiviral therapies. Here, a series of 4'-Azido-thymidine/4'-Azido-2'-deoxy-5-methylcytidine derivatives (6, 10-15) were synthesized, and their anti-HBV activities evaluated. Compounds 10-15 were synthesized via an SNAr reaction of 18, in which the 4-position of the thymine moiety was activated as the 2,4,6-triisopropylbenzenesulfonate. Compounds 11-15 showed no antiviral activity. However, 4'-Azido thymidine (6) and 4'-Azido-2'-deoxy-5-methylcytidine (10) displayed significant anti-HBV activity (EC50 = 0.63 and 5.99 µM, respectively) with no detectable cytotoxicity against MT-2 cells up to 100 µM.
Collapse
Affiliation(s)
- Kengo Onitsuka
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Ryoh Tokuda
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| | - Nobuyo Kuwata-Higashi
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Hiroki Kumamoto
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama, Japan
| | - Manabu Aoki
- Experimental Retrovirology Section, HIV & AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Masayuki Amano
- Departments of Infectious Diseases and Hematology, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Satoru Kohgo
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan
| | - Debananda Das
- Experimental Retrovirology Section, HIV & AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kazuhiro Haraguchi
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama, Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Infections, National Center for Global Health and Medicine Research Institute, Tokyo, Japan.,Experimental Retrovirology Section, HIV & AIDS Malignancy Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.,Departments of Infectious Diseases and Hematology, Kumamoto University School of Medicine, Kumamoto, Japan
| | - Shuhei Imoto
- Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Sojo University, Kumamoto, Japan
| |
Collapse
|
13
|
Yang W, Peng Y, Wang J, Song C, Yu W, Zhou Y, Jiang J, Wang Q, Wu J, Chang J. Design, synthesis, and biological evaluation of novel 2'-deoxy-2'-fluoro-2'-C-methyl 8-azanebularine derivatives as potent anti-HBV agents. Bioorg Med Chem Lett 2019; 29:1291-1297. [PMID: 30962085 DOI: 10.1016/j.bmcl.2019.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 03/29/2019] [Accepted: 04/03/2019] [Indexed: 01/01/2023]
Abstract
Hepatitis B virus (HBV) is a global health problem requiring more efficient and better tolerated anti-HBV agent. In this paper, a series of novel 2'-deoxy-2'-fluoro-2'-C-methyl-β-d-arabinofuranosyl 8-azanebularine analogues (1 and 2a) and N4-substituted 8-azaadenosine derivatives (2b-g) were designed, synthesized and screened for in vitro anti-HBV activity. Two concise and practical synthetic routes were developed toward the structural motif construction of 2'-deoxy-2'-fluoro-2'-C-methyl-β-d-arabinofuranosyl 8-azainosine from the ribonolactone 3 under mild conditions. The in vitro anti-HBV screening results showed that these 8-azanebularine analogues had a significant inhibitory effect on the expression of HBV antigens and HBV DNA at a concentration of 20 μM. Among them, halogen-substituted 8-azaadenosine derivative 2g displayed activities comparable to that of 3TC. In particular, 2g retained excellent activity against lamivudine-resistant HBV mutants.
Collapse
Affiliation(s)
- Wu Yang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Youmei Peng
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, PR China
| | - Jingwen Wang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Chuanjun Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Wenquan Yu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Yubing Zhou
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Jinhua Jiang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, PR China
| | - Qingduan Wang
- Institute of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, PR China
| | - Jie Wu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China.
| | - Junbiao Chang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, PR China.
| |
Collapse
|
14
|
Guo S, Xu M, Guo Q, Zhu F, Jiang X, Xie Y, Shen J. Discovery of pyrimidine nucleoside dual prodrugs and pyrazine nucleosides as novel anti-HCV agents. Bioorg Med Chem 2019; 27:748-759. [PMID: 30683552 DOI: 10.1016/j.bmc.2019.01.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2018] [Revised: 12/27/2018] [Accepted: 01/14/2019] [Indexed: 10/27/2022]
Abstract
To explore the application potential of dual prodrug strategies in the development of anti-HCV agents, a variety of sofosbuvir derivatives with modifications at the C4 or N3 position of the uracil moiety were designed and synthesized. Some compounds exhibited potent anti-HCV activities, such as 4e and 8a-8c with similar EC50 values (0.20-0.22 μM) comparative to that of sofosbuvir (EC50 = 0.18 μM) in a genotype 1b based replicon Huh-7 cell line. Moreover, 8b displayed a good human plasma stability profile, and was easily metabolized in human liver microsomes expectantly. On the other hand, aiming to discover novel anti-HCV nucleosides, pyrazin-2(1H)-one nucleosides and their phosphoramidate prodrugs were investigated. Several active compounds were discovered, such as 25e (EC50 = 7.3 μM) and S-29b (EC50 = 19.5 μM). This kind of nucleosides were interesting and would open a new avenue for the development of antiviral agents.
Collapse
Affiliation(s)
- Shuang Guo
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Mingshuo Xu
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Qi Guo
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| | - Fuqiang Zhu
- Topharman Shanghai Co., Ltd, No. 388 Jialilue Road, Zhangjiang Hitech Park, Shanghai 201203, China
| | - Xiangrui Jiang
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China
| | - Yuanchao Xie
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China.
| | - Jingshan Shen
- CAS Key Laboratory for Receptor Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, 555 Zuchongzhi Road, Shanghai 201203, China; University of Chinese Academy of Sciences, No. 19A Yuquan Road, Beijing 100049, China
| |
Collapse
|
15
|
Zhang J, Wang Y, Peng Y, Qin C, Liu Y, Li J, Jiang J, Zhou Y, Chang J, Wang Q. Novel fluoronucleoside analog NCC inhibits lamivudine-resistant hepatitis B virus in a hepatocyte model. Braz J Infect Dis 2018; 22:477-486. [PMID: 30586543 PMCID: PMC9425639 DOI: 10.1016/j.bjid.2018.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 11/26/2018] [Accepted: 11/30/2018] [Indexed: 11/26/2022] Open
Abstract
Antiviral drug resistance is the most important factor contributing to treatment failure using nucleos(t)ide analogs such as lamivudine for chronic infection with hepatitis B virus (HBV). Development of a system supporting efficient replication of clinically resistant HBV strains is imperative, and new antiviral drugs are needed urgently to prevent selection of drug-resistant HBV mutants. A novel fluorinated cytidine analog, NCC (N-cyclopropyl-4′-azido-2′-deoxy-2′-fluoro-β-d-cytidine), was recently shown to strongly inhibit human HBV in vitro and in vivo. This study was designed to evaluate the antiviral activity of NCC against lamivudine-resistant HBV. We generated a stable cell line encoding the major pattern of lamivudine-resistant mutations rtL180M/M204V and designated it “HepG2.RL1”. Immuno-transmission electron microscopic examination and enzyme-linked immunosorbent assay were used to detect secretion of HBV-specific particles and antigens. Quantification of extracellular DNA and intracellular DNA of HepG2.RL1 cells by quantitative real-time polymerase chain reaction revealed >625-fold and >5556-fold increases in the 50% inhibitory concentration of lamivudine, respectively, compared with that for the wild-type virus. The results showed that NCC inhibited DNA replication and HBeAg production in wild-type or lamivudine-resistant HBV in a dose-dependent manner. In conclusion, screening for antiviral compounds active against lamivudine-resistant HBV can be carried out with relative ease using hepG2.RL1 cells. NCC is a potential antiviral agent against wild-type HBV and clinical lamivudine-resistant HBV and deserves evaluation for the treatment of HBV infection.
Collapse
Affiliation(s)
- Jingmin Zhang
- The First Affiliated Hospital of Zhengzhou University, Department of Pharmacy, Zhengzhou, China; Zhengzhou University, Academy of Medical and Pharmaceutical Sciences, Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, China
| | - Yafeng Wang
- Zhengzhou University, School of Pharmaceutical Sciences, Zhengzhou, China
| | - Youmei Peng
- Zhengzhou University, Academy of Medical and Pharmaceutical Sciences, Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, China
| | - Chongzhen Qin
- The First Affiliated Hospital of Zhengzhou University, Department of Pharmacy, Zhengzhou, China
| | - Yixian Liu
- Zhengzhou University, Academy of Medical and Pharmaceutical Sciences, Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, China
| | - Jingjing Li
- The First Affiliated Hospital of Zhengzhou University, Department of Pharmacy, Zhengzhou, China
| | - Jinhua Jiang
- Zhengzhou University, Academy of Medical and Pharmaceutical Sciences, Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, China
| | - Yubing Zhou
- The First Affiliated Hospital of Zhengzhou University, Department of Pharmacy, Zhengzhou, China.
| | - Junbiao Chang
- Zhengzhou University, College of Chemistry and Molecular Engineering, Zhengzhou, China.
| | - Qingduan Wang
- Zhengzhou University, Academy of Medical and Pharmaceutical Sciences, Henan Key Laboratory for Pharmacology of Liver Diseases, Zhengzhou, China.
| |
Collapse
|
16
|
Lv B, Guo S, Zhu F, Hu T, Guo Q, Lin H, Xie Y, Shen J. Facile synthesis of 2-C-methyl-d-arabino-γ-1,4-lactones and mechanism study. Tetrahedron Lett 2018. [DOI: 10.1016/j.tetlet.2018.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
|
17
|
Kohgo S, Imoto S, Tokuda R, Takamatsu Y, Higashi-Kuwata N, Aoki M, Amano M, Kansui H, Onitsuka K, Maeda K, Mitsuya H. Synthesis of 4′-Substituted Purine 2′-Deoxynucleosides and Their Activity against Human Immunodeficiency Virus Type 1 and Hepatitis B Virus. ChemistrySelect 2018. [DOI: 10.1002/slct.201800527] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Satoru Kohgo
- Department of Refractory Viral Infection; National Center for Global Health and Medicine Research Institute; 1-21-1 Toyama, Shinjuku Tokyo 162-8655 Japan
- Faculty of Pharmaceutical Sciences; Sojo University; 4-22-1 Ikeda, Nishi-ku Kumamoto 860-0082 Japan
| | - Shuhei Imoto
- Faculty of Pharmaceutical Sciences; Sojo University; 4-22-1 Ikeda, Nishi-ku Kumamoto 860-0082 Japan
| | - Ryoh Tokuda
- Faculty of Pharmaceutical Sciences; Sojo University; 4-22-1 Ikeda, Nishi-ku Kumamoto 860-0082 Japan
| | - Yuki Takamatsu
- Experimental Retrovirology Section; HIV & AIDS Malignancy Branch; National Cancer Institute; National Institutes of Health; 10 Center Drive Room 5 A11 - MSC 1868 Bethesda MD 20892-1868 USA
| | - Nobuyo Higashi-Kuwata
- Department of Refractory Viral Infection; National Center for Global Health and Medicine Research Institute; 1-21-1 Toyama, Shinjuku Tokyo 162-8655 Japan
- Departments of Hematology; Rheumatology & Clinical Immunology and Infectious Diseases; Kumamoto University Graduate School of Medical Sciences, Honjo, Chuou-ku; Kumamoto 860-8556 Japan
| | - Manabu Aoki
- Experimental Retrovirology Section; HIV & AIDS Malignancy Branch; National Cancer Institute; National Institutes of Health; 10 Center Drive Room 5 A11 - MSC 1868 Bethesda MD 20892-1868 USA
- Departments of Hematology; Rheumatology & Clinical Immunology and Infectious Diseases; Kumamoto University Graduate School of Medical Sciences, Honjo, Chuou-ku; Kumamoto 860-8556 Japan
- Department of Medical Technology; Kumamoto Health Science University; 325 Izumi-cho, Kita-ku Kumamoto 861-5598 Japan
| | - Masayuki Amano
- Departments of Hematology; Rheumatology & Clinical Immunology and Infectious Diseases; Kumamoto University Graduate School of Medical Sciences, Honjo, Chuou-ku; Kumamoto 860-8556 Japan
| | - Hisao Kansui
- Faculty of Pharmaceutical Sciences; Sojo University; 4-22-1 Ikeda, Nishi-ku Kumamoto 860-0082 Japan
| | - Kengo Onitsuka
- Faculty of Pharmaceutical Sciences; Sojo University; 4-22-1 Ikeda, Nishi-ku Kumamoto 860-0082 Japan
| | - Kenji Maeda
- Department of Refractory Viral Infection; National Center for Global Health and Medicine Research Institute; 1-21-1 Toyama, Shinjuku Tokyo 162-8655 Japan
| | - Hiroaki Mitsuya
- Department of Refractory Viral Infection; National Center for Global Health and Medicine Research Institute; 1-21-1 Toyama, Shinjuku Tokyo 162-8655 Japan
- Experimental Retrovirology Section; HIV & AIDS Malignancy Branch; National Cancer Institute; National Institutes of Health; 10 Center Drive Room 5 A11 - MSC 1868 Bethesda MD 20892-1868 USA
- Departments of Hematology; Rheumatology & Clinical Immunology and Infectious Diseases; Kumamoto University Graduate School of Medical Sciences, Honjo, Chuou-ku; Kumamoto 860-8556 Japan
| |
Collapse
|
18
|
Li Z, Wang Z, Wang N, Han X, Yu W, Wang R, Chang J. Identification of the binding between three fluoronucleoside analogues and fat mass and obesity-associated protein by isothermal titration calorimetry and spectroscopic techniques. J Pharm Biomed Anal 2018; 149:290-295. [DOI: 10.1016/j.jpba.2017.11.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2017] [Revised: 10/27/2017] [Accepted: 11/01/2017] [Indexed: 12/19/2022]
|
19
|
Liu Y, Peng Y, Lu J, Wang J, Ma H, Song C, Liu B, Qiao Y, Yu W, Wu J, Chang J. Design, synthesis, and biological evaluation of new 1,2,3-triazolo-2'-deoxy-2'-fluoro- 4'-azido nucleoside derivatives as potent anti-HBV agents. Eur J Med Chem 2017; 143:137-149. [PMID: 29174810 DOI: 10.1016/j.ejmech.2017.11.028] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2017] [Revised: 10/23/2017] [Accepted: 11/08/2017] [Indexed: 01/15/2023]
Abstract
Novel drugs are urgently needed to combat hepatitis B virus (HBV) infection due to drug-resistant virus. In this paper, a series of novel 4-monosubstituted 2'-deoxy-2'-β-fluoro-4'-azido-β-d-arabinofuranosyl 1,2,3-triazole nucleoside analogues (1a-g) were designed, synthesized and screened for in vitro anti-HBV activity. At 5.0 μM in the cellular model, all the synthetic compounds display activities comparable to that of the positive control, lamivudine at 20 μM. Of the compounds tested, the amide-substituted analogue (1a) shows the most promising anti-HBV activity and low cytotoxicity in the cell model. In particular, it retains excellent activity against lamivudine-resistant HBV mutants. In duck HBV (DHBV)-infected duck models, both the serum and liver DHBV DNA levels (67.4% and 53.3%, respectively) were reduced markedly by the treatment with 1a. Analysis of the structure of HBV polymer/1a-triphosphate (1a-TP) complex shows that 1a-TP is stabilized by specific van der Waals interactions with the enzyme residues arising from 4-amino-1,2,3-triazole and the 4'-azido group.
Collapse
Affiliation(s)
- Yuan Liu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Youmei Peng
- Henan Academy of Medical and Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450052, PR China
| | - Jingjing Lu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Jingwen Wang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Haoran Ma
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Chuanjun Song
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Bingjie Liu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Yan Qiao
- School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, PR China
| | - Wenquan Yu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China
| | - Jie Wu
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China.
| | - Junbiao Chang
- College of Chemistry and Molecular Engineering, Zhengzhou University, Henan 450001, PR China; Collaborative Innovation Center of New Drug Research and Safety Evaluation, Henan Province, Zhengzhou 450001, PR China.
| |
Collapse
|
20
|
Saturated Heterocycles with Applications in Medicinal Chemistry. ADVANCES IN HETEROCYCLIC CHEMISTRY 2017. [DOI: 10.1016/bs.aihch.2016.03.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
21
|
|
22
|
Chen L, Zhang YH, Zou Q, Chu C, Ji Z. Analysis of the chemical toxicity effects using the enrichment of Gene Ontology terms and KEGG pathways. Biochim Biophys Acta Gen Subj 2016; 1860:2619-26. [PMID: 27208425 DOI: 10.1016/j.bbagen.2016.05.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 04/25/2016] [Accepted: 05/13/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Chemical toxicity is one of the major barriers for designing and detecting new chemical entities during drug discovery. Unexpected toxicity of an approved drug may lead to withdrawal from the market and significant loss of the associated costs. Better understanding of the mechanisms underlying various toxicity effects can help eliminate unqualified candidate drugs in early stages, allowing researchers to focus their attention on other more viable candidates. METHODS In this study, we aimed to understand the mechanisms underlying several toxicity effects using Gene Ontology (GO) terms and KEGG pathways. GO term and KEGG pathway enrichment theories were adopted to encode each chemical, and the minimum redundancy maximum relevance (mRMR) was used to analyze the GO terms and the KEGG pathways. Based on the feature list obtained by the mRMR method, the most related GO terms and KEGG pathways were extracted. RESULTS Some important GO terms and KEGG pathways were uncovered, which were concluded to be significant for determining chemical toxicity effects. CONCLUSIONS Several GO terms and KEGG pathways are highly related to all investigated toxicity effects, while some are specific to a certain toxicity effect. GENERAL SIGNIFICANCE The findings in this study have the potential to further our understanding of different chemical toxicity mechanisms and to assist scientists in developing new chemical toxicity prediction algorithms. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.
Collapse
Affiliation(s)
- Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China.
| | - Yu-Hang Zhang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.
| | - Quan Zou
- School of Computer Science and Technology, Tianjin University, Tianjin 300072, People's Republic of China.
| | - Chen Chu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China.
| | - Zhiliang Ji
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen, Fujian 361102, People's Republic of China.
| |
Collapse
|
23
|
Study on 2-arylthio-5-iodo pyrimidine derivatives as novel nonnucleoside inhibitors against hepatitis B virus DNA replication. Future Med Chem 2016; 8:751-63. [PMID: 27172826 DOI: 10.4155/fmc.16.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Novel nonnucleoside hepatitis B virus inhibitors have been recently developed for the reason of drug-resistant mutations and adverse effects of nucleoside analogs. In this study, two series of 2-arylthio-5-iodo pyrimidine analogs were firstly reported as potential anti-HBV agents. METHODOLOGY Target compounds were prepared according to two high-yielded synthetic routes, and their anti-HBV activities were evaluated on Hep2.2.15 and HepAD38 cell lines, respectively. To probe the mechanism of active agents, a cell-based (Huh-7) study of biochemical markers (e.g., HBeAg, HBsAg, intracellular HBV DNA and pgRNA) was performed. Furthermore, the pharmacophore models were constructed for future optimization of lead compounds. CONCLUSION 2-Arylthio-5-iodo pyrimidine derivatives firstly proved to be effective against HBV, which paves the way for future development of nonnucleoside anti-HBV agents.
Collapse
|
24
|
Lv Z, He W, Tian X, Kang J, Liu Y, Peng Y, Zheng L, Wang Q, Yu W, Chang J. Design, synthesis, and biological evaluation of new N 4 -Substituted 2′-deoxy-2′-fluoro-4′-azido cytidine derivatives as potent anti-HBV agents. Eur J Med Chem 2015; 101:103-10. [DOI: 10.1016/j.ejmech.2015.06.030] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2015] [Revised: 06/08/2015] [Accepted: 06/12/2015] [Indexed: 02/08/2023]
|