1
|
Argade MD, Achi JG, Bott R, Morsheimer KM, Owen CD, Zielinski CA, Gaisin AM, Alvarez M, Moore TW, Bu F, Li F, Cameron M, Anantpadma M, Davey RA, Peet NP, Rong L, Gaisina IN. Guardians at the Gate: Optimization of Small Molecule Entry Inhibitors of Ebola and Marburg Viruses. J Med Chem 2025; 68:135-155. [PMID: 39680623 DOI: 10.1021/acs.jmedchem.4c01646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2024]
Abstract
Ebola and Marburg (EBOV and MARV) filoviral infections lead to fatal hemorrhagic fevers and have caused over 30 outbreaks in the last 50 years. Currently, there are no FDA-approved small molecule therapeutics for effectively treating filoviral diseases. To address this unmet medical need, we have conducted a systematic structural optimization of an early lead compound, N-(4-(4-methylpiperidin-1-yl)-3-(trifluoromethyl)phenyl)-4-(morpholinomethyl)benzamide (1), borne from our previously reported hit-to-lead effort. This secondary round of structure-activity relationship (SAR) involved the design and synthesis of several deconstructed and reconstructed analogs of compound 1, which were then tested against pseudotyped EBOV and MARV. The antiviral activities of the most promising leads were further validated in infectious assays. The optimized analogs exhibited desirable antiviral activity against different ebolaviruses and reduced off-target activity. Additionally, they also possessed druglike properties, that make them ideal candidates for in vivo efficacy studies as part of our ongoing drug discovery campaign against EBOV and MARV.
Collapse
Affiliation(s)
- Malaika D Argade
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Jazmin Galván Achi
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Ryan Bott
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Kimberly M Morsheimer
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - Callum D Owen
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - Christian A Zielinski
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Arsen M Gaisin
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Mario Alvarez
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Terry W Moore
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
| | - Fan Bu
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Fang Li
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, United States
- Center for Emerging Viruses, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael Cameron
- Department of Molecular Medicine, Herbert Wertheim, UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida 33458, United States
| | - Manu Anantpadma
- The Integrated Research Facility, National Institute of Allergy and Infectious Diseases, Frederick, Maryland 20892, United States
| | - Robert A Davey
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, Massachusetts 02118, United States
| | - Norton P Peet
- Chicago BioSolutions Inc., Chicago, Illinois 60612, United States
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Chicago BioSolutions Inc., Chicago, Illinois 60612, United States
| | - Irina N Gaisina
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, Illinois 60612, United States
- Chicago BioSolutions Inc., Chicago, Illinois 60612, United States
| |
Collapse
|
2
|
Morales-Tenorio M, Lasala F, Garcia-Rubia A, Aledavood E, Heung M, Olal C, Escudero-Pérez B, Alonso C, Martínez A, Muñoz-Fontela C, Delgado R, Gil C. Discovery of Thiophene Derivatives as Potent, Orally Bioavailable, and Blood-Brain Barrier-Permeable Ebola Virus Entry Inhibitors. J Med Chem 2024; 67:16381-16402. [PMID: 39248591 PMCID: PMC11440591 DOI: 10.1021/acs.jmedchem.4c01267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/03/2024] [Revised: 08/12/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024]
Abstract
The endemic nature of the Ebola virus disease in Africa underscores the need for prophylactic and therapeutic drugs that are affordable and easy to administer. Through a phenotypic screening employing viral pseudotypes and our in-house chemical library, we identified a promising hit featuring a thiophene scaffold, exhibiting antiviral activity in the micromolar range. Following up on this thiophene hit, a new series of compounds that retain the five-membered heterocyclic scaffold while modifying several substituents was synthesized. Initial screening using a pseudotype viral system and validation assays employing authentic Ebola virus demonstrated the potential of this new chemical class as viral entry inhibitors. Subsequent investigations elucidated the mechanism of action through site-directed mutagenesis. Furthermore, we conducted studies to assess the pharmacokinetic profile of selected compounds to confirm its pharmacological and therapeutic potential.
Collapse
Affiliation(s)
| | - Fátima Lasala
- Instituto
de Investigación Hospital 12 de Octubre,, Madrid 28041, Spain
| | - Alfonso Garcia-Rubia
- Centro
de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid 28040, Spain
| | - Elnaz Aledavood
- Centro
de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid 28040, Spain
| | - Michelle Heung
- Bernhard
Nocht Institute for Tropical Medicine, Hamburg 20359, Germany
| | - Catherine Olal
- Bernhard
Nocht Institute for Tropical Medicine, Hamburg 20359, Germany
| | | | - Covadonga Alonso
- Dpt.
Biotechnology, Instituto Nacional de Investigación
y Tecnología Agraria y Alimentaria (INIA-CSIC), Madrid 28040, Spain
| | - Ana Martínez
- Centro
de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid 28040, Spain
- CIBERNED, Instituto Salud Carlos III, Madrid 28029, Spain
| | | | - Rafael Delgado
- Instituto
de Investigación Hospital 12 de Octubre,, Madrid 28041, Spain
- CIBERINFEC, Instituto Salud Carlos III, Madrid 28029, Spain
- School
of Medicine, Universidad Complutense de
Madrid, Madrid 28040, Spain
| | - Carmen Gil
- Centro
de Investigaciones Biológicas Margarita Salas (CIB-CSIC), Madrid 28040, Spain
- CIBERNED, Instituto Salud Carlos III, Madrid 28029, Spain
| |
Collapse
|
3
|
Shekunov EV, Efimova SS, Kever LV, Ishmanov TF, Ostroumova OS. Lipid Selectivity of Membrane Action of the Fragments of Fusion Peptides of Marburg and Ebola Viruses. Int J Mol Sci 2024; 25:9901. [PMID: 39337389 PMCID: PMC11432738 DOI: 10.3390/ijms25189901] [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] [Academic Contribution Register] [Received: 07/26/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The life cycle of Ebola and Marburg viruses includes a step of the virion envelope fusion with the cell membrane. Here, we analyzed whether the fusion of liposome membranes under the action of fragments of fusion peptides of Ebola and Marburg viruses depends on the composition of lipid vesicles. A fluorescence assay and electron microscopy were used to quantify the fusogenic activity of the virus fusion peptides and to identify the lipid determinants affecting membrane merging. Differential scanning calorimetry of lipid phase transitions revealed alterations in the physical properties of the lipid matrix produced by virus fusion peptides. Additionally, we found that plant polyphenols, quercetin, and myricetin inhibited vesicle fusion induced by the Marburg virus fusion peptide.
Collapse
Affiliation(s)
- Egor V Shekunov
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Svetlana S Efimova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Lyudmila V Kever
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Tagir F Ishmanov
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| | - Olga S Ostroumova
- Laboratory of Membrane and Ion Channel Modeling, Institute of Cytology of Russian Academy of Sciences, Tikhoretsky 4, 194064 Saint Petersburg, Russia
| |
Collapse
|
4
|
Sokolova AS, Baev DS, Mordvinova ED, Yarovaya OI, Volkova NV, Shcherbakov DN, Okhina AA, Rogachev AD, Shnaider TA, Chvileva AS, Nikitina TV, Tolstikova TG, Salakhutdinov NF. (+)-fenchol and (-)-isopinocampheol derivatives targeting the entry process of filoviruses. Eur J Med Chem 2024; 275:116596. [PMID: 38889610 DOI: 10.1016/j.ejmech.2024.116596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/27/2024] [Revised: 06/02/2024] [Accepted: 06/11/2024] [Indexed: 06/20/2024]
Abstract
The increasing frequency of filovirus outbreaks in African countries has led to a pressing need for the development of effective antifilovirus agents. In continuation of our previous research on the antifilovirus activity of monoterpenoid derivatives, we synthesized a series of (+)-fenchol and (-)-isopinocampheol derivatives by varying the type of heterocycle and linker length. Derivatives with an N-alkylpiperazine cycle proved to be the most potent antiviral compounds, with half-maximal inhibitory concentration (IC50) 1.4-20 μМ against Lenti-EboV-GP infection and 11.3-47 μМ against Lenti-MarV-GP infection. Mechanism-of-action experiments revealed that the compounds may exert their action by binding to surface glycoproteins (GPs). It was demonstrated that the binding of the synthesized compounds to the Marburg virus GP is less efficient as compared to the Ebola virus GP. Furthermore, it was shown that the compounds possess lysosomotropic properties. Thus, the antiviral activity may be due to dual effects. This study offers new antiviral agents that are worthy of further exploration.
Collapse
Affiliation(s)
- Anastasiya S Sokolova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation.
| | - Dmitriy S Baev
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation; SRF SKIF, Koltsovo, Novosibirsk Oblast, 630559, Russian Federation
| | - Ekaterina D Mordvinova
- State Research Center of Virology and Biotechnology VECTOR (Rospotrebnadzor), Koltsovo, Novosibirsk Oblast, 630559, Russian Federation
| | - Olga I Yarovaya
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation
| | - Natalia V Volkova
- State Research Center of Virology and Biotechnology VECTOR (Rospotrebnadzor), Koltsovo, Novosibirsk Oblast, 630559, Russian Federation
| | - Dmitriy N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR (Rospotrebnadzor), Koltsovo, Novosibirsk Oblast, 630559, Russian Federation
| | - Alina A Okhina
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation
| | - Artem D Rogachev
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation
| | - Tatiana A Shnaider
- Institute of Cytology and Genetics (ICG), SB RAS, Novosibirsk, 630090, Russian Federation
| | | | - Tatiana V Nikitina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center, Russian Academy of Sciences, Tomsk, 634050, Russian Federation
| | - Tatyana G Tolstikova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation
| | - Nariman F Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences (SB RAS), Novosibirsk, 630090, Russian Federation
| |
Collapse
|
5
|
Durante D, Bott R, Cooper L, Owen C, Morsheimer KM, Patten J, Zielinski C, Peet NP, Davey RA, Gaisina IN, Rong L, Moore TW. N-Substituted Pyrrole-Based Heterocycles as Broad-Spectrum Filoviral Entry Inhibitors. J Med Chem 2024; 67:13737-13764. [PMID: 39169825 PMCID: PMC11812679 DOI: 10.1021/acs.jmedchem.4c00527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 08/23/2024]
Abstract
Since the largest and most fatal Ebola virus epidemic during 2014-2016, there have been several consecutive filoviral outbreaks in recent years, including those in 2021, 2022, and 2023. Ongoing outbreak prevalence and limited FDA-approved filoviral therapeutics emphasize the need for novel small molecule treatments. Here, we showcase the structure-activity relationship development of N-substituted pyrrole-based heterocycles and their potent, submicromolar entry inhibition against diverse filoviruses in a target-based pseudovirus assay. Inhibitor antiviral activity was validated using replication-competent Ebola, Sudan, and Marburg viruses. Mutational analysis was used to map the targeted region within the Ebola virus glycoprotein. Antiviral counter-screen and phospholipidosis assays were performed to demonstrate the reduced off-target activity of these filoviral entry inhibitors. Favorable antiviral potency, selectivity, and drug-like properties of the N-substituted pyrrole-based heterocycles support their potential as broad-spectrum antifiloviral treatments.
Collapse
Affiliation(s)
- Destiny Durante
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Ryan Bott
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Laura Cooper
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, United States
| | - Callum Owen
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - Kimberly M. Morsheimer
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - J.J. Patten
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - Christian Zielinski
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, IL, 60612, United States
| | - Norton P. Peet
- Chicago BioSolutions Inc., Chicago, IL 60612, United States
| | - Robert A. Davey
- Department of Virology, Immunology, and Microbiology, National Emerging Infectious Diseases Laboratories, Boston University Medical Campus, Boston, MA 02118, United States
| | - Irina N. Gaisina
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, United States
- Chicago BioSolutions Inc., Chicago, IL 60612, United States
- UICentre: Drug Discovery, University of Illinois Chicago, Chicago, IL, 60612, United States
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL 60612, United States
- Chicago BioSolutions Inc., Chicago, IL 60612, United States
| | - Terry W. Moore
- Department of Pharmaceutical Sciences, University of Illinois Chicago, Chicago, IL 60612, United States
- University of Illinois Cancer Center, University of Illinois Chicago, Chicago, IL 60612, United States
| |
Collapse
|
6
|
Caffrey M, Jayakumar N, Caffrey V, Anirudhan V, Rong L, Paprotny I. VLP-based model for the study of airborne viral pathogens. Microbiol Spectr 2024; 12:e0001324. [PMID: 38752752 PMCID: PMC11237701 DOI: 10.1128/spectrum.00013-24] [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] [Academic Contribution Register] [Received: 01/03/2024] [Accepted: 04/21/2024] [Indexed: 05/28/2024] Open
Abstract
The recent COVID-19 pandemic has underscored the danger of airborne viral pathogens. The lack of model systems to study airborne pathogens limits the understanding of airborne pathogen distribution as well as potential surveillance and mitigation strategies. In this work, we develop a novel model system to study airborne pathogens using virus-like particles (VLPs). Specifically, we demonstrate the ability to aerosolize VLP and detect and quantify aerosolized VLP RNA by reverse transcription-loop-mediated isothermal amplification in real-time fluorescent and colorimetric assays. Importantly, the VLP model presents many advantages for the study of airborne viral pathogens: (i) similarity in size and surface components; (ii) ease of generation and noninfectious nature enabling the study of biosafety level 3 and biosafety level 4 viruses; (iii) facile characterization of aerosolization parameters; (iv) ability to adapt the system to other viral envelope proteins, including those of newly discovered pathogens and mutant variants; and (v) the ability to introduce viral sequences to develop nucleic acid amplification assays. IMPORTANCE The study and detection of airborne pathogens are hampered by the lack of appropriate model systems. In this work, we demonstrate that noninfectious virus-like particles (VLPs) represent attractive models to study airborne viral pathogens. Specifically, VLPs are readily prepared, are similar in size and composition to infectious viruses, and are amenable to highly sensitive nucleic acid amplification techniques.
Collapse
Affiliation(s)
- Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Nitin Jayakumar
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Veronique Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Varada Anirudhan
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, Illinois, USA
| | - Igor Paprotny
- Department of Electrical and Computer Engineering, University of Illinois at Chicago, Chicago, Illinois, USA
| |
Collapse
|
7
|
Srivastava S, Kumar S, Ashique S, Sridhar SB, Shareef J, Thomas S. Novel antiviral approaches for Marburg: a promising therapeutics in the pipeline. Front Microbiol 2024; 15:1387628. [PMID: 38725678 PMCID: PMC11079314 DOI: 10.3389/fmicb.2024.1387628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/18/2024] [Accepted: 04/11/2024] [Indexed: 05/12/2024] Open
Abstract
Marburg virus disease (MVD) presents a significant global health threat, lacking effective antivirals and with current supportive care offering limited therapeutic options. This mini review explores the emerging landscape of novel antiviral strategies against MVD, focusing on promising therapeutics currently in the development pipeline. We delve into direct-acting antiviral approaches, including small molecule inhibitors targeting viral entry, replication, and assembly, alongside nucleic acid antisense and RNA interference strategies. Host-targeting antivirals are also considered, encompassing immune modulators like interferons and cytokine/chemokine modulators, broad-spectrum antivirals, and convalescent plasma and antibody-based therapies. The paper then examines preclinical and clinical development for the novel therapeutics, highlighting in vitro and in vivo models for antiviral evaluation, safety and efficacy assessments, and the critical stages of clinical trials. Recognizing the challenges of drug resistance and viral escape, the mini review underscores the potential of combination therapy strategies and emphasizes the need for rapid diagnostic tools to optimize treatment initiation. Finally, we discuss the importance of public health preparedness and equitable access to these promising therapeutics in achieving effective MVD control and global health security. This mini review presents a comprehensive overview of the burgeoning field of MVD antivirals, highlighting the potential of these novel approaches to reshape the future of MVD treatment and prevention.
Collapse
Affiliation(s)
- Shriyansh Srivastava
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
- Department of Pharmacy, School of Medical and Allied Sciences, Galgotias University, Greater Noida, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi, India
| | - Sumel Ashique
- Department of Pharmaceutical Sciences, Bengal College of Pharmaceutical Sciences & Research, Durgapur, West Bengal, India
| | - Sathvik Belagodu Sridhar
- RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras al Khaimah, United Arab Emirates
| | - Javedh Shareef
- Clinical Pharmacy & Pharmacology, RAK College of Pharmacy, RAK Medical & Health Sciences University, Ras al Khaimah, United Arab Emirates
| | - Sabin Thomas
- College of Health Sciences, University of Nizwa, Nizwa, Oman
| |
Collapse
|
8
|
Zhang Y, Zhang M, Wu H, Wang X, Zheng H, Feng J, Wang J, Luo L, Xiao H, Qiao C, Li X, Zheng Y, Huang W, Wang Y, Wang Y, Shi Y, Feng J, Chen G. A novel MARV glycoprotein-specific antibody with potentials of broad-spectrum neutralization to filovirus. eLife 2024; 12:RP91181. [PMID: 38526940 PMCID: PMC10963030 DOI: 10.7554/elife.91181] [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] [Academic Contribution Register] [Indexed: 03/27/2024] Open
Abstract
Marburg virus (MARV) is one of the filovirus species that cause deadly hemorrhagic fever in humans, with mortality rates up to 90%. Neutralizing antibodies represent ideal candidates to prevent or treat virus disease. However, no antibody has been approved for MARV treatment to date. In this study, we identified a novel human antibody named AF-03 that targeted MARV glycoprotein (GP). AF-03 possessed a high binding affinity to MARV GP and showed neutralizing and protective activities against the pseudotyped MARV in vitro and in vivo. Epitope identification, including molecular docking and experiment-based analysis of mutated species, revealed that AF-03 recognized the Niemann-Pick C1 (NPC1) binding domain within GP1. Interestingly, we found the neutralizing activity of AF-03 to pseudotyped Ebola viruses (EBOV, SUDV, and BDBV) harboring cleaved GP instead of full-length GP. Furthermore, NPC2-fused AF-03 exhibited neutralizing activity to several filovirus species and EBOV mutants via binding to CI-MPR. In conclusion, this work demonstrates that AF-03 represents a promising therapeutic cargo for filovirus-caused disease.
Collapse
Affiliation(s)
- Yuting Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical UniversityHohhotChina
| | - Min Zhang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Haiyan Wu
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Xinwei Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical UniversityHohhotChina
| | - Hang Zheng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical UniversityHohhotChina
| | - Junjuan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical UniversityHohhotChina
| | - Jing Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Longlong Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - He Xiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Chunxia Qiao
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Xinying Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Yuanqiang Zheng
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical UniversityHohhotChina
| | - Weijin Huang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug ControlBeijingChina
| | - Youchun Wang
- Division of HIV/AIDS and Sex-transmitted Virus Vaccines, National Institutes for Food and Drug ControlBeijingChina
| | - Yi Wang
- Department of Hematology, Fifth Medical Center of Chinese PLA General HospitalBeijingChina
| | - Yanchun Shi
- Inner Mongolia Key Lab of Molecular Biology, School of Basic Medical Sciences, Inner Mongolia Medical UniversityHohhotChina
| | - Jiannan Feng
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| | - Guojiang Chen
- State Key Laboratory of Toxicology and Medical Countermeasures, Institute of Pharmacology and ToxicologyBeijingChina
| |
Collapse
|
9
|
Sokolova AS, Yarovaya OI, Artyushin OI, Sharova EV, Baev DS, Mordvinova ED, Shcherbakov DN, Shnaider TA, Nikitina TV, Esaulkova IL, Ilyina PA, Zarubaev VV, Brel VK, Tolstikova TG, Salakhutdinov NF. Design, synthesis and antiviral evaluation of novel conjugates of the 1,7,7-trimethylbicyclo[2.2.1]heptane scaffold and saturated N-heterocycles via 1,2,3-triazole linker. Arch Pharm (Weinheim) 2024; 357:e2300549. [PMID: 38036303 DOI: 10.1002/ardp.202300549] [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] [Academic Contribution Register] [Received: 09/29/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 12/02/2023]
Abstract
A new series of heterocyclic derivatives with a 1,7,7-trimethylbicyclo[2.2.1]heptane fragment was designed, synthesised and biologically evaluated. Synthesis of the target compounds was performed using the Cu(I) catalysed cycloaddition reaction. The key starting substances in the click reaction were an alkyne containing a 1,7,7-trimethylbicyclo[2.2.1]heptane fragment and a series of azides with saturated nitrogen-containing heterocycles. Some of the derivatives were found to exhibit strong antiviral activity against Marburg and Ebola pseudotype viruses. Lysosomal trapping assays revealed the derivatives to possess lysosomotropic properties. The molecular modelling study demonstrated the binding affinity between the compounds investigated and the possible active site to be mainly due to hydrophobic interactions. Thus, combining a natural hydrophobic structural fragment and a lysosome-targetable heterocycle may be an effective strategy for designing antiviral agents.
Collapse
Affiliation(s)
- Anastasiya S Sokolova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Olga I Yarovaya
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Oleg I Artyushin
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, Russian Federation
| | - Elena V Sharova
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, Russian Federation
| | - Dmitriy S Baev
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
- Synchrotron Radiation Facility SKIF, G.K. Boreskov Institute of Catalysis SB RAS, Koltsovo, Russian Federation
| | - Ekaterina D Mordvinova
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk Region, Russian Federation
| | - Dmitriy N Shcherbakov
- State Research Center of Virology and Biotechnology VECTOR, Rospotrebnadzor, Koltsovo, Novosibirsk Region, Russian Federation
| | - Tatiana A Shnaider
- Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Tatiana V Nikitina
- Research Institute of Medical Genetics, Tomsk National Research Medical Center of the Russian Academy of Sciences, Tomsk, Russian Federation
| | - Iana L Esaulkova
- Pasteur Institute of Epidemiology and Microbiology, St. Petersburg, Russian Federation
| | - Polina A Ilyina
- Pasteur Institute of Epidemiology and Microbiology, St. Petersburg, Russian Federation
| | - Vladimir V Zarubaev
- Pasteur Institute of Epidemiology and Microbiology, St. Petersburg, Russian Federation
| | - Valery K Brel
- A.N. Nesmeyanov Institute of Organoelement Compounds, Russian Academy of Sciences, Moscow, Russian Federation
| | - Tatyana G Tolstikova
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| | - Nariman F Salakhutdinov
- N.N. Vorozhtsov Novosibirsk Institute of Organic Chemistry, Siberian Branch of Russian Academy of Sciences, Novosibirsk, Russian Federation
| |
Collapse
|
10
|
da Rocha JM, Campos DMDO, Esmaile SC, Menezes GDL, Bezerra KS, da Silva RA, Junior EDDS, Tayyeb JZ, Akash S, Fulco UL, Alqahtani T, Oliveira JIN. Quantum biochemical analysis of the binding interactions between a potential inhibitory drug and the Ebola viral glycoprotein. J Biomol Struct Dyn 2024:1-17. [PMID: 38258414 DOI: 10.1080/07391102.2024.2305314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/19/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024]
Abstract
Ebola virus disease (EVD) causes outbreaks and epidemics in West Africa that persist until today. The envelope glycoprotein of Ebola virus (GP) consists of two subunits, GP1 and GP2, and plays a key role in anchoring or fusing the virus to the host cell in its active form on the virion surface. Toremifene (TOR) is a ligand that mainly acts as an estrogen receptor antagonist; however, a recent study showed a strong and efficient interaction with GP. In this context, we aimed to evaluate the energetic affinity features involved in the interaction between GP and toremifene by computer simulation techniques using the Molecular Fractionation Method with Conjugate Caps (MFCC) scheme and quantum-mechanical (QM) calculations, as well as missense mutations to assess protein stability. We identified ASP522, GLU100, TYR517, THR519, LEU186, LEU515 as the most attractive residues in the EBOV glycoprotein structure that form the binding pocket. We divided toremifene into three regions and evaluated that region i was more important than region iii and region ii for the formation of the TOR-GP1/GP2 complex, which might control the molecular remodeling process of TOR. The mutations that caused more destabilization were ARG134, LEU515, TYR517 and ARG559, while those that caused stabilization were GLU523 and ASP522. TYR517 is a critical residue for the binding of TOR, and is highly conserved among EBOV species. Our results may help to elucidate the mechanism of drug action on the GP protein of the Ebola virus and subsequently develop new pharmacological approaches against EVD.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jaerdyson M da Rocha
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Daniel M de O Campos
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Stephany C Esmaile
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Gabriela de L Menezes
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Katyanna S Bezerra
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Roosevelt A da Silva
- Core Collaboratives of BioSistemas, Special Unit of Exact Sciences, Federal University of Jataí, Jataí, GO, Brazil
| | - Edilson D da S Junior
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Jehad Zuhair Tayyeb
- Department of Clinical Biochemistry, College of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| | - Shopnil Akash
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Birulia, Ashulia, Dhaka, Bangladesh
| | - Umberto L Fulco
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Jonas I N Oliveira
- Department of Biophysics and Pharmacology, Bioscience Center, Federal University of Rio Grande do Norte, Natal, RN, Brazil
| |
Collapse
|
11
|
Caffrey M, Jayakumar N, Caffrey V, Anirudan V, Rong L, Paprotny I. VLP-Based Model for Study of Airborne Viral Pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.03.574055. [PMID: 38260552 PMCID: PMC10802359 DOI: 10.1101/2024.01.03.574055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 01/24/2024]
Abstract
The recent COVID-19 pandemic has underscored the danger of airborne viral pathogens. The lack of model systems to study airborne pathogens limits the understanding of airborne pathogen distribution, as well as potential surveillance and mitigation strategies. In this work, we develop a novel model system to study airborne pathogens using virus like particles (VLP). Specifically, we demonstrate the ability to aerosolize VLP and detect and quantify aerosolized VLP RNA by Reverse Transcription-Loop-Mediated Isothermal Amplification (RT-LAMP) in real-time fluorescent and colorimetric assays. Importantly, the VLP model presents many advantages for the study of airborne viral pathogens: (i) similarity in size and surface components; (ii) ease of generation and noninfectious nature enabling study of BSL3 and BSL4 viruses; (iii) facile characterization of aerosolization parameters; (iv) ability to adapt the system to other viral envelope proteins including those of newly discovered pathogens and mutant variants; (v) the ability to introduce viral sequences to develop nucleic acid amplification assays.
Collapse
Affiliation(s)
- Michael Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607
| | - Nitin Jayakumar
- Department of Mechanical and Industrial Engineering, University of Illinois at Chicago, Chicago, IL 60607
| | - Veronique Caffrey
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, IL 60607
| | - Varada Anirudan
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL 60612
| | - Igor Paprotny
- Department of Electrical and Computer Engineering, University of Illinois at Chicago, Chicago, IL 60607
| |
Collapse
|
12
|
Liang J, Wu Y, Lan K, Dong C, Wu S, Li S, Zhou HB. Antiviral PROTACs: Opportunity borne with challenge. CELL INSIGHT 2023; 2:100092. [PMID: 37398636 PMCID: PMC10308200 DOI: 10.1016/j.cellin.2023.100092] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Academic Contribution Register] [Received: 12/23/2022] [Revised: 03/15/2023] [Accepted: 03/16/2023] [Indexed: 07/04/2023]
Abstract
Proteolysis targeting chimera (PROTAC) degradation of pathogenic proteins by hijacking of the ubiquitin-proteasome-system has become a promising strategy in drug design. The overwhelming advantages of PROTAC technology have ensured a rapid and wide usage, and multiple PROTACs have entered clinical trials. Several antiviral PROTACs have been developed with promising bioactivities against various pathogenic viruses. However, the number of reported antiviral PROTACs is far less than that of other diseases, e.g., cancers, immune disorders, and neurodegenerative diseases, possibly because of the common deficiencies of PROTAC technology (e.g., limited available ligands and poor membrane permeability) plus the complex mechanism involved and the high tendency of viral mutation during transmission and replication, which may challenge the successful development of effective antiviral PROTACs. This review highlights the important advances in this rapidly growing field and critical limitations encountered in developing antiviral PROTACs by analyzing the current status and representative examples of antiviral PROTACs and other PROTAC-like antiviral agents. We also summarize and analyze the general principles and strategies for antiviral PROTAC design and optimization with the intent of indicating the potential strategic directions for future progress.
Collapse
Affiliation(s)
- Jinsen Liang
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Yihe Wu
- 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, 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
| | - Chune Dong
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Shuwen Wu
- State Key Laboratory of Virology, College of Life Sciences, Wuhan University, Wuhan, 430072, China
| | - Shu Li
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
| | - Hai-Bing Zhou
- Medical Research Institute, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, 430071, China
- 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, School of Pharmaceutical Sciences, Wuhan University, Wuhan, 430071, China
| |
Collapse
|
13
|
Yousef M, Le TS, Zuo J, Park C, Chacra NB, Davies NM, Löbenberg R. Sub-cellular sequestration of alkaline drugs in lysosomes: new insights for pharmaceutical development of lysosomal fluid. Res Pharm Sci 2022; 18:1-15. [PMID: 36846734 PMCID: PMC9951787 DOI: 10.4103/1735-5362.363591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/22/2022] [Revised: 10/23/2022] [Accepted: 11/22/2022] [Indexed: 12/25/2022] Open
Abstract
Background and purpose Lysosomal-targeted drug delivery can open a new strategy for drug therapy. However, there is currently no universally accepted simulated or artificial lysosomal fluid utilized in the pharmaceutical industry or recognized by the United States Pharmacopeia (USP). Experimental procedure We prepared a simulated lysosomal fluid (SLYF) and compared its composition to a commercial artificial counterpart. The developed fluid was used to test the dissolution of a commercial product (Robitussin®) of a lysosomotropic drug (dextromethorphan) and to investigate in-vitro lysosomal trapping of two model drugs (dextromethorphan and (+/-) chloroquine). Findings/Results The laboratory-prepared fluid or SLYF contained the essential components for the lysosomal function in concentrations reflective of the physiological values, unlike the commercial product. Robitussin® passed the acceptance criteria for the dissolution of dextromethorphan in 0.1 N HCl medium (97.7% in less than 45 min) but not in the SLYF or the phosphate buffer media (72.6% and 32.2% within 45 min, respectively). Racemic chloroquine showed higher lysosomal trapping (51.9%) in the in-vitro model than dextromethorphan (28.3%) in a behavior supporting in-vivo findings and based on the molecular descriptors and the lysosomal sequestration potential of both. Conclusion and implication A standardized lysosomal fluid was reported and developed for in-vitro investigations of lysosomotropic drugs and formulations.
Collapse
Affiliation(s)
- Malaz Yousef
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada,Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Tyson S. Le
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Jieyu Zuo
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - Chulhun Park
- College of Pharmacy, Jeju National University, Jeju 63243, South Korea
| | - Nadia Bou Chacra
- Faculty of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Neal M. Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada,Corresponding authors: N.M. Davies, Tel: +1-7802210828, Fax: +1-7804921217
R. Löbenberg, Tel: +1-7804921255, Fax: +1-7804921217
| | - Raimar Löbenberg
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada,Corresponding authors: N.M. Davies, Tel: +1-7802210828, Fax: +1-7804921217
R. Löbenberg, Tel: +1-7804921255, Fax: +1-7804921217
| |
Collapse
|
14
|
Plescia CB, Lindstrom AR, Quintero MV, Keiser P, Anantpadma M, Davey R, Stahelin RV, Davisson VJ. Evaluation of Phenol-Substituted Diphyllin Derivatives as Selective Antagonists for Ebola Virus Entry. ACS Infect Dis 2022; 8:942-957. [PMID: 35357134 PMCID: PMC9112336 DOI: 10.1021/acsinfecdis.1c00474] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/28/2022]
Abstract
![]()
Ebola
virus (EBOV) is an aggressive filoviral pathogen that can
induce severe hemorrhagic fever in humans with up to 90% fatality
rate. To date, there are no clinically effective small-molecule drugs
for postexposure therapies to treat filoviral infections. EBOV cellular
entry and infection involve uptake via macropinocytosis, navigation
through the endocytic pathway, and pH-dependent escape into the cytoplasm.
We report the inhibition of EBOV cell entry via selective inhibition
of vacuolar (V)-ATPase by a new series of phenol-substituted derivatives
of the natural product scaffold diphyllin. In cells challenged with
Ebola virus, the diphyllin derivatives inhibit viral entry dependent
upon structural variations to low nanomolar potencies. Mechanistically,
the diphyllin derivatives had no effect on uptake and colocalization
of viral particles with endocytic marker LAMP1 but directly modulated
endosomal pH. The most potent effects were reversible exhibiting higher
selectivity than bafilomycin or the parent diphyllin. Unlike general
lysosomotrophic agents, the diphyllin derivatives showed no major
disruptions of endocytic populations or morphology when examined with
Rab5 and LAMP1 markers. The dilated vacuole phenotype induced by apilimod
treatment or in constitutively active Rab5 mutant Q79L-expressing
cells was both blocked and reversed by the diphyllin derivatives.
The results are consistent with the action of the diphyllin scaffold
as a selective pH-dependent viral entry block in late endosomes. Overall,
the compounds show improved selectivity and minimal cytotoxicity relative
to classical endosomal acidification blocking agents.
Collapse
Affiliation(s)
| | | | - Maritza V. Quintero
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio 78229-3900, United States
| | - Patrick Keiser
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts 02118, United States
| | - Manu Anantpadma
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts 02118, United States
| | - Robert Davey
- Department of Microbiology, National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts 02118, United States
| | | | | |
Collapse
|
15
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been prevalent in the humans since 2019 and has given rise to a pandemic situation. With the discovery and ongoing use of drugs and vaccines against SARS-CoV-2, there is still no surety of its complete suppression of this disease or if there is a need for additional booster doses. There is an urgent need for alternative treatment strategies against COVID-19. Peptides and peptidomimetics have several advantages as therapeutic agents because of their target selectivity, better interactions, and lower toxicity. Minor structural alterations to peptides can help prevent their fast metabolism and provide long-action. This comprehensive review provides an overview of different peptide-based vaccines and therapeutics against SARS-CoV-2. It discusses the design and mechanism of action of the peptide-based vaccines, peptide immunomodulators, anti-inflammatory agents, and peptides as entry inhibitors of SARS-CoV-2. Moreover, the mechanism of action, sequences and current clinical trial studies are also summarized. The review also discusses the future aspects of peptide-based vaccines and therapeutics for COVID-19. Graphical Abstract
Collapse
Affiliation(s)
- Pritam V. Bagwe
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019 India
| | - Priyal V. Bagwe
- Vaccine Nanotechnology Laboratory, Center for Drug Delivery and Research, College of Pharmacy, Mercer University, Atlanta, GA 30341 USA
| | - Sai Srinivas Ponugoti
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019 India
| | - Shreerang V. Joshi
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, Maharashtra 400019 India
| |
Collapse
|
16
|
Cooper L, Achi JG, Rong L. Comparative analyses of small molecule and antibody inhibition on glycoprotein-mediated entry of Měngla virus with other filoviruses. J Med Virol 2022; 94:3263-3269. [PMID: 35332563 PMCID: PMC9161972 DOI: 10.1002/jmv.27739] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/09/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 11/08/2022]
Abstract
The ability of viruses in the filoviridae family (Ebola virus (EBOV) and Marburg Virus (MARV)) to cause severe human disease and their pandemic potential makes all emerging filoviral pathogens a concern to humanity. Měnglà Virus (MLAV) belonging to the new genus Dianlovirus was recently discovered in the liver of bats from Měnglà County, Yunnan Province, China. The capacity of MLAV to utilize NPC1 as an endosomal receptor, to transduce mammalian cells, and suppress IFN response suggests that this potential pathogen could cause human illness. Despite great effort by researchers, only the viral genome has been recovered and isolation of live MLAV had been unsuccessful. Here using a pseudovirus model baring the MLAV glycoprotein (GP), we studied the protease dependence of the MLAV-GP, and the ability of small molecules and antibodies to inhibit MLAV viral entry. Like EBOV and MARV, the MLAV-GP requires proteolytic processing but like MARV it does not depend on cathepsin B activity for viral entry. Furthermore, previously discovered small molecule inhibitors and antibodies are MLAV inhibitors and show the possibility of developing these inhibitors as broad-spectrum filovirus antivirals. Overall, the findings in the study confirmed that MLAV viral entry is biologically distinct but has similarities to MARV. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Laura Cooper
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Jazmin Galvan Achi
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, University of Illinois at Chicago, Chicago, IL, 60612, USA
| |
Collapse
|
17
|
Tsang NY, Li WF, Varhegyi E, Rong L, Zhang HJ. Ebola Entry Inhibitors Discovered from Maesa perlarius. Int J Mol Sci 2022; 23:ijms23052620. [PMID: 35269770 PMCID: PMC8910447 DOI: 10.3390/ijms23052620] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/31/2021] [Revised: 02/08/2022] [Accepted: 02/23/2022] [Indexed: 11/21/2022] Open
Abstract
Ebola virus disease (EVD), a disease caused by infection with Ebola virus (EBOV), is characterized by hemorrhagic fever and a high case fatality rate. With limited options for the treatment of EVD, anti-Ebola viral therapeutics need to be urgently developed. In this study, over 500 extracts of medicinal plants collected in the Lingnan region were tested against infection with Ebola-virus-pseudotyped particles (EBOVpp), leading to the discovery of Maesa perlarius as an anti-EBOV plant lead. The methanol extract (MPBE) of the stems of this plant showed an inhibitory effect against EBOVpp, with an IC50 value of 0.52 µg/mL, which was confirmed by testing the extract against infectious EBOV in a biosafety level 4 laboratory. The bioassay-guided fractionation of MPBE resulted in three proanthocyanidins (procyanidin B2 (1), procyanidin C1 (2), and epicatechin-(4β→8)-epicatechin-(4β→8)-epicatechin-(4β→8)-epicatechin (3)), along with two flavan-3-ols ((+)-catechin (4) and (−)-epicatechin (5)). The IC50 values of the compounds against pseudovirion-bearing EBOV-GP ranged from 0.83 to 36.0 µM, with 1 as the most potent inhibitor. The anti-EBOV activities of five synthetic derivatives together with six commercially available analogues, including EGCG ((−)-epigallocatechin-3-O-gallate (8)), were further investigated. Molecular docking analysis and binding affinity measurement suggested the EBOV glycoprotein could be a potential molecular target for 1 and its related compounds.
Collapse
Affiliation(s)
- Nga Yi Tsang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China; (N.Y.T.); (W.-F.L.)
| | - Wan-Fei Li
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China; (N.Y.T.); (W.-F.L.)
| | - Elizabeth Varhegyi
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, 909 South Wolcott Ave, Chicago, IL 60612, USA;
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois Chicago, 909 South Wolcott Ave, Chicago, IL 60612, USA;
- Correspondence: (L.R.); (H.-J.Z.); Tel.: +1-312-3550203 (L.R.); +852-34112956 (H.-J.Z.)
| | - Hong-Jie Zhang
- Teaching and Research Division, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong, China; (N.Y.T.); (W.-F.L.)
- Correspondence: (L.R.); (H.-J.Z.); Tel.: +1-312-3550203 (L.R.); +852-34112956 (H.-J.Z.)
| |
Collapse
|
18
|
Ebola Virus Entry Inhibitors. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1366:155-170. [DOI: 10.1007/978-981-16-8702-0_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Academic Contribution Register] [Indexed: 11/25/2022]
|
19
|
White JM, Schiffer JT, Bender Ignacio RA, Xu S, Kainov D, Ianevski A, Aittokallio T, Frieman M, Olinger GG, Polyak SJ. Drug Combinations as a First Line of Defense against Coronaviruses and Other Emerging Viruses. mBio 2021; 12:e0334721. [PMID: 34933447 PMCID: PMC8689562 DOI: 10.1128/mbio.03347-21] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022] Open
Abstract
The world was unprepared for coronavirus disease 2019 (COVID-19) and remains ill-equipped for future pandemics. While unprecedented strides have been made developing vaccines and treatments for COVID-19, there remains a need for highly effective and widely available regimens for ambulatory use for novel coronaviruses and other viral pathogens. We posit that a priority is to develop pan-family drug cocktails to enhance potency, limit toxicity, and avoid drug resistance. We urge cocktail development for all viruses with pandemic potential both in the short term (<1 to 2 years) and longer term with pairs of drugs in advanced clinical testing or repurposed agents approved for other indications. While significant efforts were launched against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), in vitro and in the clinic, many studies employed solo drugs and had disappointing results. Here, we review drug combination studies against SARS-CoV-2 and other viruses and introduce a model-driven approach to assess drug pairs with the highest likelihood of clinical efficacy. Where component agents lack sufficient potency, we advocate for synergistic combinations to achieve therapeutic levels. We also discuss issues that stymied therapeutic progress against COVID-19, including testing of agents with low likelihood of efficacy late in clinical disease and lack of focus on developing virologic surrogate endpoints. There is a need to expedite efficient clinical trials testing drug combinations that could be taken at home by recently infected individuals and exposed contacts as early as possible during the next pandemic, whether caused by a coronavirus or another viral pathogen. The approach herein represents a proactive plan for global viral pandemic preparedness.
Collapse
Affiliation(s)
- Judith M. White
- University of Virginia, Department of Cell Biology, Charlottesville, Virginia, USA
- University of Virginia, Department of Microbiology, Charlottesville, Virginia, USA
| | - Joshua T. Schiffer
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Rachel A. Bender Ignacio
- University of Washington, Division of Allergy and Infectious Diseases, Seattle, Washington, USA
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Shuang Xu
- Fred Hutchinson Cancer Research Center, Vaccine and Infectious Diseases Division, Seattle, Washington, USA
| | - Denis Kainov
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Institute of Technology, University of Tartu, Tartu, Estonia
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Aleksandr Ianevski
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
| | - Tero Aittokallio
- Institute for Molecular Medicine Finland, FIMM, University of Helsinki, Helsinki, Finland
- Oslo Centre for Biostatistics and Epidemiology (OCBE), University of Oslo, Oslo, Norway
- Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Matthew Frieman
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | | | - Stephen J. Polyak
- Virology Division, Department of Laboratory Medicine and Pathology, University of Washington, Seattle, Washington, USA
- Department of Global Health, University of Washington, Seattle, Washington, USA
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
20
|
Chen Z, Du R, Galvan Achi JM, Rong L, Cui Q. SARS-CoV-2 cell entry and targeted antiviral development. Acta Pharm Sin B 2021; 11:3879-3888. [PMID: 34002130 PMCID: PMC8117542 DOI: 10.1016/j.apsb.2021.05.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/17/2021] [Revised: 04/21/2021] [Accepted: 04/29/2021] [Indexed: 12/25/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the causative agent of the pandemic coronavirus disease 2019 (COVID-19), which threatens human health and public safety. In the urgent campaign to develop anti-SARS-CoV-2 therapies, the initial entry step is one of the most appealing targets. In this review, we summarize the current understanding of SARS-CoV-2 cell entry, and the development of targeted antiviral strategies. Moreover, we speculate upon future directions toward next-generation of SARS-CoV-2 entry inhibitors during the upcoming post-pandemic era.
Collapse
Affiliation(s)
- Zinuo Chen
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruikun Du
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266122, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Jazmin M. Galvan Achi
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Lijun Rong
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Qinghua Cui
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
- Qingdao Academy of Chinese Medicinal Sciences, Shandong University of Traditional Chinese Medicine, Qingdao 266122, China
- Experimental Center, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| |
Collapse
|
21
|
Abstract
In this issue of Cell, Ganaie et al. reports the identification of LRP1 as a receptor of the highly pathogenic Rift Valley fever virus. By using genome-wide CRISPR-Cas9 screening and functional studies, Ganaie et al. identified LRP1 and several co-factors as essential elements for virus infection.
Collapse
Affiliation(s)
- Nathen E Bopp
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Institute for Human Infection and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Diana Fernández
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Institute for Human Infection and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Center for Tropical Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA
| | - Patricia V Aguilar
- Department of Pathology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Institute for Human Infection and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA; Center for Tropical Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX 77550, USA.
| |
Collapse
|