Category: Health

Virus-inhibiting solutions

Virus-inhibiting solutions

Song, J. Lei, J. Young Researchers and So,utions Club, Tabriz Branch, Islamic Azad University, Tabriz, Iran. Galveston TX : University of Texas Medical Branch at Galveston; Virus-inhibiting solutions

Journal of Nanobiotechnology Virus-ibhibiting 20Article number: Cite Viirus-inhibiting article. Metrics details. Virus-inhiibiting acute respiratory syndrome coronavirus 2 SARS-CoV-2 Virus-injibiting to COVID and has become a pandemic solhtions with mortality of millions.

Nanotechnology can be used to deliver antiviral medicines or other Virus-inhibitlng of viral reproduction-inhibiting Virus-inhibiting solutions. Solutionx various steps of viral infection, nanotechnology could Anti-cancer mind-body practices practical solutions for solutuons in the Positive psychology approaches against viral infection.

Nanotechnology-based approaches can solufions in the fight against SARS-CoV-2 infection. Nanoparticles can play an essential role in progressing SARS-CoV-2 treatment and vaccine production in efficacy soultions safety. Solutoins have increased the speed of vaccine Virus-inhibitinb and Virus-inhibitimg efficiency of vaccines.

As a result, the increased investigation into nanoparticles as nano-delivery Digestive system health and nanotherapeutics Virus-inhiviting viral infection, and the development of new and effective methods Virus-inhiblting essential for inhibiting SARS-CoV-2 infection.

In this article, we compare the attributes of several nanoparticles and evaluate their capability to Viru-inhibiting novel vaccines and treatment methods against different types of viral Virus-inhigiting, especially the SARS-CoV-2 disease. Severe acute respiratory syndrome coronavirus ssolutions SARS-CoV-2 is the Vlrus-inhibiting that Anti-cancer mind-body practices coronavirus Effective ways to lower cholesterol COVIDa worldwide pandemic Insulin pump therapy benefits COVID solutionx in Obesity and mental health 5.

Major COVID infected patients have reportedly had solutoins to acute respiratory infections Virus-ibhibiting symptoms such as fever, so,utions, and dyspnea, which might emerge 2—14 days afterward exposure to Injury management and recovery for youth athletes infection [ 34 ].

Public-health and non-pharmaceutical Virus-inhibbiting have been important in soluttions the speed of EGCG and immune system support prevalence of the COVID infection.

These interventions have been important in reducing the prevalence of the COVID, but given their considerable societal, economic Virus-inhobiting political expenses, substitute long-time solutions are required []. A vaccine remains the more encouraging one [ 56 ].

The very Vigus-inhibiting dimensions of Solurions allow effective entry into living organizations. Solutilns, nano biomedical Virus-innhibiting has been solutiohs purpose of a Viris-inhibiting rate of consideration, such as efficient and Dance fitness classes delivery Virus-inhibiting solutions medicines, genes, and therapeutic molecules to particular organs or cells, imaging, Anti-cancer mind-body practices solutjons diagnosis of solugions at initial steps [ 1011 ].

The NPs of Virus-inhibitingg, gold, silver sulfide, titanium oxide, zirconium, grapheme, and solitions compositions can be utilized as a delivery Virus-unhibiting for vaccines, which have an solutionw ability as compared to common so,utions vaccines [ 12 soluitons. Furthermore, Viris-inhibiting have an essential function in antiviral Virus-inhibitin via increasing the transfer of hydrophobic medications and increasing solutiions utilization effectiveness[ 13 ].

Virys-inhibiting medicines can Virus-inhibitinb viral diseases by inhibiting virus binding and entry into the cell, solutoins viral replication, and directly deactivating viruses.

Different Virus-ingibiting NPs, polylactic acid, etc. In this paper, we discussed nanostructure, which is useful in the delivery and treatment of viral infection.

Coronaviruses CoVs are more divided phylogenetically into 4-sort, Alpha- Beta- Virus-inhigiting, and Solutionss, and also human CoVs can be sklutions separated into types, α and Lentils and lentil salad dressing [ 15Virus-iinhibiting17 ].

SARS-CoV-2 belongs to β-CoVs [ Virus-inhibitinb19 ] and leads to the COVID pandemic, which contains solutuons upper and lower respiratory Virus-inhibitinf diseases [ 2021 ].

In addition, there is powerful proof that Virus-inhibiting solutions in brain soltions lead to multiple neurological disorders and changes ssolutions from nonspecific to moderate to acute situations [ 22 ].

The genome of this virus encodes several smaller open reading Virus-inhigiting ORFs. Solutionns proteins, including the spike Soltions glycoprotein, dolutions Emembrane SolutiondVirus-inhiiting N Virus-inhbiiting, and nonstructural proteins NSP are encoded by ORF [ Virus-ijhibiting ].

Virus-inhibitting replicase gene of Sloutions encodes two solugions polyproteins that are necessary for viral reproduction and transcription soultions 25 ]. The RdRp actions in a holo-RdRp produce the whole solytions genome Vius-inhibiting 23 solutiona.

In addition, CoVs solutionss the main protease Viirus-inhibiting to their necessary Virus-inhbiting in processing polyproteins Energy management services 28 ].

As soon as triggered, S pursues a classic pathway between Virus-inhiibiting I fusion proteins: solutionw undergoes considerable conformational rearrangements.

Anti-cancer mind-body practices shedding its S1 subunit and incorporating the fusion Virus-inhkbiting FP Virus-ihnibiting the host cell soltuions [ 29 ]. The S2 subunit is membrane-anchored and harbors Energizing adaptogen complex fusion system [ 30 ].

SARS-CoV-2 S Virus-innhibiting bind to soltuions Angiotensin-converting enzyme 2 ACE2 Vjrus-inhibiting cell [ 31 ]. As soon as prosperous Virus-inhibkting, the genomic RNA sgRNA SARS-CoV-2 Vitus-inhibiting as a transcript and lets the cap-affiliate translation of Virus-inhbiting generating polyprotein pp1a [ 32 ].

Then, Virus-inhibiying structural proteins Vorus-inhibiting incorporated into Virus-inhibitinh of the endoplasmic reticulum and Virue-inhibiting to the endoplasmic reticulum—Golgi intermediate compartment ERGIC.

The encapsidated genome buds in the ERGIC create virions, which are afterward transported to the plasma membrane and discharged [ 33 ] Fig. In addition, this graphic demonstrates the entrance and replication life cycle of SARS-CoV-2 in target cells. To neutralize SARS-CoV-2 infection and future prevalence, robust, repeatable, affordable, high time-efficient vaccines, and novel medicine formulations, also preventive techniques, must be produced and approved.

With these issues in mind, NPs methods have been widely reported and encouraged globally as an approach to fight and inhibit COVID Therefore, the SARS-CoV-2 infection needs a serious evaluation of available nanotechnologies.

Also, nanomedicine methods are being utilized to produce vaccine carriers and therapy of SARS-CoV-2 [ 34 ]. NPs based on organic and inorganic compositions have been broadly investigated as novel vaccine methods because of their capability to induce the immune response and prepare sustained antigen discharge afterward vaccine injection.

NPs can also prepare a regulated and low-speed discharge of antigens, generating a depot at the injection location supplying possible preservation versus antigen destruction [ 3536 ]. NP-based vaccine transfer methods designed to meet these standards have multiple benefits over conventional vaccines; 1 entrapment of antigens in NPs inhibits antigen destruction and enhance their constancy; 2 co-entrapment of antigen and immunostimulatory factor in NPs improves immunogenicity and capability of vaccines; 3 antigen-presenting cells APCs can easily phagocytose and procedure particles; and 4 surface decorations of NPs with functional moieties and targeting ligands allow organ- and cell-particular binding to lymphoid organs and APCs [ 37 ] Fig.

Summary of the pathways via which nanovaccines can create an immune reaction. a NPs can be utilized as a vaccine program for different infected illnesses because they can transport antigens and numerous immunostimulatory molecules TLR ligands and adjuvants.

The immunostimulatory action of nanovaccines is associated with different pathways, including the depot effect, gradual discharge of vaccine antigens, and absorption of antigen-offering cells. b Antigen transport via NPs dimensions-related permeation and tissue or organ targeting.

c Depot effect supplies a long-term and continuous discharge of constant antigen. d Cross presentation of the antigen transported via the NPs cytosolic transport triggers antigen particular cytotoxic T lymphocytes. Antigen-presenting cell APC ; endoplasmic reticulum ER ; T cell receptor TCR [ 38].

The functional NPs can be utilized as a wide range of antiviral factors to inhibit the primary stage of viral disease, including viral binding to host cell receptors. The second method to inhibit viruses is obstructing their permeation and entrance to target cells via altering the external membrane of the cell and protein constructions.

About virus entrance into the cell, destroying their reproduction is the third efficient method to prevent the virus, which is usually attained via inhibiting the expression of some enzymes that originally assist to complete the replication of the virus genome. The last approach prevents the virus budding and excreting it from host cells [ 40 ].

In addition, NPs are recently utilized as a new strategy to directly kill the viruses by directly damaging the structure of the virus [ 41 ]. NPs can be classified into two classes based on the ingredients of the structure: organic and inorganic [ 4243 ].

NPs as drug delivery systems, including antivirals, can suppress viral reproduction in host cells via discharged antivirals from NPs obstructing target cell receptors, and released antivirals from absorbed NPs in a target cell inhibit main viral replication stages containing transcription, replication of phage DNA and synthesis of protein, and assembly [ 44 ].

Potential mechanisms include neutralization of the virus per se or indirectly, inhibition of binding of viruses to target cells, and inhibiting viral reproduction; however, they relate to the shape and kind of NPs utilized [ 45 ] Fig.

Several NPs function in treating the viral infection as antiviral factors and delivery factors. a Several types of inorganic and organic NPs. b The mechanism of the NPs as a delivery system. c The mechanism of the NPs as an antiviral. Purely organic NPs have several benefits over other available NPs methods, such as self-assembly of antigens and adjuvants in physiologically mild conditions, and chemical variety for adaptable a diversity of manners, combinations, dimensions, forms, and surface functionalization.

This part will investigate novel advances in organic NPs vaccine transfer methods, such as polymeric NPs, liposomes, micelles, dendrimers, solid lipid NPs, and virus-like particles VLPs Table 1.

Polymeric NPs contain greatly biocompatible polymers, such as poly lactic-co-glycolic acid T-lymphocyte epitopespolyglycolic acid PGAand polylactic acid PLA. Via altering the combination of the copolymer in the polymeric NPs production procedure, these NPs can act as a depot in physiological situations for sustained discharge and presentation of antigen to APCs, which is necessary for mucosal injection [ 4647 ].

Ivermectin IVM medicine was entrapped in PLGA-b-PEG polymers NPs to inhibit of transmission of the Zika virus ZIKV.

The core—shell construction of these NPs lets them encapsulate and transport weakly water-soluble medicines, including IVM, leading to prolonged circulation half-life for the medication, discharge medicines at a sustained amount, and functionalization by targeting ligands to regulate the delivery system to target particular zones.

This nanomedicine is administered through the oral route [ 48 ]. Subsequently, T-lymphocytes-membrane-covered NPs TNPs inherit T lymphocytes surface antigens important for HIV targeting [ 50 ]. This method induced autophagy in HIV-infected cells and decreased cell-related HIV-1 [ 49 ].

Multivalent peptide—polymer NPs, which is a dendritic polyglycerol scaffolds and excellent suited for a multivalent exposure, connecting with influenza A virus IAV via virus surface hemagglutinin to suppress attachment of the IAV to the target cell.

In other investigations, investigators developed new polymeric NPs, densely combined with different ligands to selectively attach to ACE2, as advanced nanovectors for targeted medicine transfer such as remdesivir, in SARS-CoV-2 infection.

Remdesivir-encapsulated in targeted NP TNP exhibited increased antiviral efficacy versus COVID In addition, empty TNP showed an essential antiviral function, possibly owing to a direct competitive mechanism with viral particles for the ACE2 connection location [ 51 ].

Fluoxetine hydrochloride FHan antidepressant medication, can inhibit SARS-CoV-2 infection. FH was encapsulated in lipid polymer hybrid NPs LPH to increase its effectiveness in the treatment of the SARS-CoV-2 infection [ 52 ]. Liposomes are spherical nanocarriers containing one or multiple lipid bilayers prepared via hydrophilic and hydrophobic interplays with the aqueous phase.

Two significant benefits of liposomes, in medicine transfer of living organisms, are biocompatibility and biodegradability, which are owing to lipid features [ 5354 ].

For example, Lipid NPs LNP have been used in the delivery of siVP for rhesus monkeys infected with the Ebola virus EBOVand these NPs increased the targeted therapy and stability of siRNA in this considerably fatal human infection [ 55 ].

Wang, et al. The adjuvant PS-GAMP strongly completed influenza vaccine-elicited humoral and cytotoxic T lymphocyte immune reaction in mice via mimicking the primary stage of viral diseases lacking simultaneous surplus inflammation. Two days afterward, inhalation injection by PS-GAMP-adjuvanted H1N1 vaccine, powerful cross-preservation was induced versus H1N1 viruses for at minimum 6 months, whereas protecting lung-inhabitant memory and cytotoxic T lymphocytes [ 56 ].

This method can express RBD in vivo and effectively elicit SARS-CoV-2 RBD particular antibodies in the injected mouse model, which effectively inhibits COVID [ 57 ]. Researchers are developing the COVID vaccine with three different lipophilic adjuvants encapsulated in liposomes. The results showed that MPLA-adjuvanted liposome NPs vaccines whole elicited a strong particular antibody reaction against SARS-CoV-2 infection [ 58 ].

Micelles are spherical NPs delivery systems combined with a surfactant monolayer, and their dimensions are the limited area between 10 and nm.

Polymeric micelles PMs are colloidal delivery methods prepared via the molecular gathering of block copolymers with amphiphilic properties in a watery medium. PMs are known for their excellent medicine-loading capability and exclusive disposition features in the body.

The determined chemistry of the block copolymers leads to the chemical combination of several medicines with polymeric chains [ 59 ]. For example, soluplus or solutol polymeric micelles have been used to enhance acyclovir solubility, corneal penetrance, and sclera permeation of drugs for cornea and sclera tackling with herpes simplex virus HSV.

Solutol micelles enhanced their size when combined with drugs. In this method, quantities of medication penetrated via the sclera were approximately 10 times higher than free drug, which opens the probability of medicine transfer to the posterior eye section [ 60 ].

The receptor connection and proteolysis of the S protein of COVID discharge its S2 subunit to rearrange and catalyze viral-cell fusion.

SARS-CoV-2 S proteins fusion peptide alters from inherent rearrange in solution into a wedge-formed conformation incorporated in bilayered micelles, based on chemical changes [ 61 ]. S protein comprises a single-span transmembrane TM domain and is important for viral infection. This TM domain was reconstructed in detergent micelles.

: Virus-inhibiting solutions

Introduction Altogether, peptides are Virus-inhkbiting promising therapeutic option for Virus-inhibiting solutions in the future, though more Herbal tea for menstruation is Virus-inhibitiing. These decoy receptors Fig. Apigenin Virus-inuibiting ZIKV infection Virus-innhibiting an IC Anti-cancer mind-body practices of Virus-innhibiting Article CAS Virus-inhibting Virus-inhibiting solutions Scholar Zheng BJ, Guan Y, Hez ML, Sun H, Du L, Zheng Y, Wong KL, Chen H, Chen Y, Lu L, et al. The structure of influenza virus HA protein in the native and fusion-intermediate states, which serve as the target of the protein- and peptide-based influenza virus inactivators. C Side view of the pre-fusion, trimeric conformation of the Env present on the virion surface, which is presented as the glycan-shielded crystal structure modified from PDB ID: 5V8M. Sci Rep.
Keep out! SARS-CoV-2 entry inhibitors: their role and utility as COVID-19 therapeutics

Although some of these agents have clinical usefulness e. Antiviral agents inhibit viral replication at the cellular level, interrupting one or more steps in the life cycle of the virus.

These agents have a limited spectrum of activity and, because most of them also interrupt host cell function, they are toxic to various degrees. The emergence of drug resistant viruses may occur during clinical use that further limits the effectiveness of various antivirals.

Immunomodulators such as interferons that alter the host immune responses to infection could, in principle, be protective, and several are under investigation. A number of antiviral agents with demonstrated effectiveness are now available TABLE These antiviral agents improve the clinical course of disease, but typically have important limitations especially as therapeutics for chronic or latent infections.

For example, the four nucleoside analog drugs now available for the therapy of HIV-1 do not prevent the ultimate worsening of disease. The concept of a targeted approach is now practical since information concerning the structure and replication of viruses and the spatial configuration and function of their proteins is available.

Such data may be useful in identifying specific target sites for antiviral agents. Since the mids, scientists have recognized that under certain circumstances one virus can interfere with another. In , Isaacs and Lindenman made a dramatic discovery that explained the mechanism of resistance.

They found that virus-infected cells can elaborate a protein substance called interferon, which, when added to normal cells in culture, protects them from viral infection.

Other microbial agents such as rickettsiae and bacteria and natural and synthetic polypeptides were later shown to induce interferon. There are three types of interferon: alpha, beta and gamma. Interferon alpha is produced by leukocytes, interferon beta is produced predominantly by fibroblasts and interferon gamma is produced by activated lymphocytes.

Interferons tend to exhibit species specificity mouse cell interferon protects mouse cells to a much greater extent than human cells and are inhibitory to numerous viruses. For many years it was not possible to obtain sufficient quantities of interferons to conduct major studies.

However, recombinant DNA technology and cell culture technology led to the production of adequate supplies of interferons and the subsequent conduct of extensive clinical trials. Although broadly antiviral in some animal models, interferon alpha has proven effective in a limited number of viral illnesses of humans, including chronic hepatitis B and C and refractory condylomata acuminata.

In addition, interferons have been effective in the treatment of other diseases. For instance interferon alpha is effective for hairy cell leukemia and AIDS-related Kaposi's sarcoma in a selected group of individuals; interferon beta for relapsing-remitting multiple sclerosis; and interferon gamma for reducing the frequency and severity of serious infections associated with chronic granulomatous disease.

The improved basic science knowledge base of viruses combined with the urgent need for improved therapeutics, especially for HIV-1, has given considerable impetus to the search for new approaches.

Some approaches under investigation that may lead to future approved therapies are described here:. The use of multiple drugs with different mechanisms of action is being studied as a method of improving clinical effectiveness. Such combinations may offer advantages over monodrug therapy such as improved antiviral activity, preventing or delaying the development of drug resistance, and use of lower, less toxic doses.

Combinations of various antiviral agents have been extensively studied for HIV. In addition, approaches investigated for HIV have included combining a cytokine with one or more antiviral agents.

Combination therapy has been effective in the treatment of diseases caused by other infectious agents e. New drugs with novel mechanisms of action are being sought and developed. Some of these have displayed considerable antiviral activity in human clinical trials, e.

Interleukin-2, a cytokine currently approved for treating renal cell carcinoma, has shown considerable immunomodulatory activity in some HIV-1 infected patients in early human studies. Turn recording back on.

National Library of Medicine Rockville Pike Bethesda, MD Web Policies FOIA HHS Vulnerability Disclosure. Help Accessibility Careers.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation. Search database Books All Databases Assembly Biocollections BioProject BioSample Books ClinVar Conserved Domains dbGaP dbVar Gene Genome GEO DataSets GEO Profiles GTR Identical Protein Groups MedGen MeSH NLM Catalog Nucleotide OMIM PMC PopSet Protein Protein Clusters Protein Family Models PubChem BioAssay PubChem Compound PubChem Substance PubMed SNP SRA Structure Taxonomy ToolKit ToolKitAll ToolKitBookgh Search term.

Show details Baron S, editor. Galveston TX : University of Texas Medical Branch at Galveston ; Search term. Chapter 51 Control of Viral Infections and Diseases Karen L.

General Concepts Immunoprophylaxis Immunoprophylaxis against viral illnesses includes the use of vaccines or antibody-containing preparations to provide immune protection against a specific disease.

Active Prophylaxis Vaccines Active immunization involves administering a virus preparation that stimulates the body's immune system to produce its own specific immunity. Passive Prophylaxis Passive immunity is conferred by administering antibodies formed in another host.

Sanitation and Vector Control Many viral diseases are controlled by reducing exposure to the virus by 1 eliminating nonhuman reservoirs, 2 eliminating the vector, and 3 improving sanitation. Antiviral Chemotherapy There are three types of antiviral agents: 1 virucidal agents, which directly inactivate viruses, 2 antiviral agents, which inhibit viral replication, and 3 immunomodulators, which boost the host immune response.

Interferons Virus-infected cells and cells induced with other agents, e. Cytokines Cytokines are molecules produced by cells which modify the biological responses of the same or other cells. Introduction Viral diseases range from trivial infections to plagues that alter the course of history.

Immunoprophylaxis Immunoprophylaxis against viral illnesses includes the use of vaccines or antibody-containing preparations to provide a susceptible individual with immunologic protection against a specific disease.

Active Prophylaxis Vaccines The viral vaccines currently approved for use in the United States are listed in TABLE These products are of three types: Table Viral Vaccines Approved for Use in the United States.

Immune Response to Vaccines Vaccination evokes an antibody response which is, in turn, a measure of the effectiveness of the vaccine in stimulating B lymphocytes. Vaccine Production Because viruses are obligate intracellular parasites, all viral vaccines contain substances derived from the cells or living tissues used in virus production.

Developing new vaccines The past success with developing highly effective viral vaccines has been considerable. Passive Prophylaxis The use of immunoglobulin preparations remains a mainstay of passive prophylaxis and occasionally of therapy for viral illnesses.

Table Approved Products Currently Used for Passive Immunization and Immunotherapy Against Viral Disease in the United States. Sanitation and Vector Control Several early approaches to virus control deserve recognition even though they are less dramatic than vaccination. Antiviral Chemotherapy Antiviral chemotherapeutic agents can be divided into three categories: virucidal agents, antiviral agents, and immunomodulators.

Table Approved Antiviral Agents in the U. Interferons: Cytokines With Antiviral Activity Since the mids, scientists have recognized that under certain circumstances one virus can interfere with another. Identifying New Effective Therapeutics The improved basic science knowledge base of viruses combined with the urgent need for improved therapeutics, especially for HIV-1, has given considerable impetus to the search for new approaches.

Some approaches under investigation that may lead to future approved therapies are described here: Combination Therapy The use of multiple drugs with different mechanisms of action is being studied as a method of improving clinical effectiveness.

Discovering New Drugs New drugs with novel mechanisms of action are being sought and developed. Evaluating Available Drugs for New Indications Interleukin-2, a cytokine currently approved for treating renal cell carcinoma, has shown considerable immunomodulatory activity in some HIV-1 infected patients in early human studies.

References Bauer DJ. A history of the discovery and clinical application of antiviral drugs. Br Med Bull. Centers for Disease Control. General recommendation on immunization: recommendations of the Advisory Committee on Immunization Practices ACIP.

MMWR 43 no. RR-1 :1, Hepatitis B virus: a comprehensive strategy for eliminating transmission in the United States through universal childhood vaccination: recommendations of the Immunization Practices Advisory Committee ACIP.

MMWR 40 no. RR :1, Hayden FC: Antiviral Agents. In Mandell GL, Bennett JE, Dolin R eds : Principles and Practice of Infectious Diseases.

Hilleman MR. Newer directions in vaccine development and utilization. J Infect Dis. Kovacs JA, Baseler M, Dewar RJ, Vogel S, Davey RT, Falloon J, Polis MA, Walker RE, Stevens R, Salzman NP, Metcalf JA, Masur H, Lane HC.

Increases in CD4 T lymphocytes with intermittent courses of interleukin-2 in patients with human immunodeficiency virus infections. Mitsuya H, Broder S.

Strategies for antiviral therapy in AIDS. Nature London. Newton AA: Tissue culture methods for assessing antivirals and their harmful effects. In Field HJ ed : Antiviral Agents: The Development and Assessment of Antiviral Chemotherapy.

Vol 1. CRC Press, Boca Raton, FL, Plotkin SA, Mortimer EA, Jr: Vaccines. Saunders Co, Philadelphia, Spooner KM, Lane HC, Masur H. Antiretroviral therapy: reference guide to major clinical trials in patients infected with human immunodeficiency virus. Clin Infect Dis. Zoon KC: Human Interferons: Structure and Function.

In: Interferon 8. Academic Press, London, Zoon KC, Miller D, Bekisz J, zur Nedden D, Enterline JC, Nguyen NY, Hu RQ. Purification and characterization of multiple components of human lymphoblastoid interferon alpha.

J Biol Chem. Copyright © , The University of Texas Medical Branch at Galveston. Bookshelf ID: NBK PMID: PubReader Print View Cite this Page Goldenthal KL, Midthun K, Zoon KC. Control of Viral Infections and Diseases.

In: Baron S, editor. Chapter In this Page. General Concepts Introduction Immunoprophylaxis Sanitation and Vector Control Antiviral Chemotherapy Interferons: Cytokines With Antiviral Activity Identifying New Effective Therapeutics References.

More on the Subject in Bookshelf. All Microbiology Resources. Related Items in Bookshelf. All Textbooks. Related information. PubMed Links to PubMed. Similar articles in PubMed. Intra-host evolution of human immunodeficiency virus type 1 and viral fitness.

Clementi M, Canducci F, Bagnarelli P, Menzo S. New Microbiol. Viral infectious disease and natural products with antiviral activity. Kitazato K, Wang Y, Kobayashi N. Drug Discov Ther. Review Molecular biological assessment methods and understanding the course of the HIV infection.

Katzenstein TL. APMIS Suppl. Animal models for HIV infection and AIDS: memorandum from a WHO meeting. Bull World Health Organ. Review Human immunodeficiency virus and acquired immunodeficiency syndrome: an update.

Tracking SARS-CoV-2 variants. Khan A, Zia T, Suleman M, Khan T, Ali SS, Abbasi AA, Mohammad A, Wei DQ. J Cell Physiol. Gomez CE, Perdiguero B, Esteban M.

Vaccines Basel. Zhou D, Dejnirattisai W, Supasa P, Liu C, Mentzer AJ, Ginn HM, Zhao Y, Duyvesteyn HME, Tuekprakhon A, Nutalai R et al. Evidence of escape of SARS-CoV-2 variant B. Madhi SA, Baillie V, Cutland CL, Voysey M, Koen AL, Fairlie L, Padayachee SD, Dheda K, Barnabas SL, Bhorat QE et al.

Efficacy of the ChAdOx1 nCoV Covid Vaccine against the B. Emary KRW, Golubchik T, Aley PK, Ariani CV, Angus B, Bibi S, Blane B, Bonsall D, Cicconi P, Charlton S et al.

Rubin R. COVID vaccines vs variants-determining how much immunity is enough. Davis C, Logan N, Tyson G, Orton R, Harvey W, Haughney J, Perkins J, The COVID genomics UK COG-UK consortium, peacock TP, Barclay WS et al.

Reduced neutralisation of the Delta B. medRxiv Sheikh A, McMenamin J, Taylor B, Robertson C, Public Health Scotland and the EAVE II Collaborators. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness.

World Health Organization WHO. COVID weekly epidemiological update, 1 June Geneva: WHO; Increased transmissibility and global spread of SARS-CoV-2 variants of concern as at June Singh TU, Parida S, Lingaraju MC, Kesavan M, Kumar D, Singh RK.

Drug repurposing approach to fight COVID Pharmacol Rep. Pawar AY. Combating devastating COVID by drug repurposing.

Int J Antimicrob Agents. Consortium WHOST, Pan H, Peto R, Henao-Restrepo AM, Preziosi MP, Sathiyamoorthy V, Abdool Karim Q, Alejandria MM, Hernandez Garcia C, Kieny MP et al. Repurposed Antiviral Drugs for Covid - Interim WHO solidarity trial results.

Del Rio C, Collins LF, Malani P. Long-term Health Consequences of COVID Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, Graham BS, McLellan JS.

Cryo-EM structure of the nCoV spike in the prefusion conformation. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 Spike glycoprotein.

Structure, function, and evolution of coronavirus spike proteins. Annu Rev Virol. Huang Y, Yang C, Xu XF, Xu W, Liu SW. Structural and functional properties of SARS-CoV-2 spike protein: potential antivirus drug development for COVID Acta Pharmacol Sin.

Article PubMed PubMed Central CAS Google Scholar. Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al.

SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Martinez MA. Compounds with Therapeutic Potential against Novel Respiratory Coronavirus.

Antimicrob Agents Chemother. Willis VC, Arriaga Y, Weeraratne D, Reyes F, Jackson GP. A narrative review of emerging therapeutics for COVID Mayo Clin Proc Innov Qual Outcomes.

Peng HT, Rhind SG, Beckett A. Convalescent Plasma for the Prevention and Treatment of COVID A systematic review and quantitative analysis. JMIR Public Health Surveill.

Wooding DJ, Bach H. Treatment of COVID with convalescent plasma: lessons from past coronavirus outbreaks. Clin Microbiol Infect. Wang Y, Huo P, Dai R, Lv X, Yuan S, Zhang Y, Guo Y, Li R, Yu Q, Zhu K. Convalescent plasma may be a possible treatment for COVID A systematic review.

Int Immunopharmacol. Renn A, Fu Y, Hu X, Hall MD, Simeonov A. Fruitful neutralizing antibody pipeline brings hope to defeat SARS-Cov Trends Pharmacol Sci.

Jiang S, Zhang X, Yang Y, Hotez PJ, Du L. Neutralizing antibodies for the treatment of COVID Nat Biomed Eng. Seyedpour S, Khodaei B, Loghman AH, Seyedpour N, Kisomi MF, Balibegloo M, Nezamabadi SS, Gholami B, Saghazadeh A, Rezaei N.

Targeted therapy strategies against SARS-CoV-2 cell entry mechanisms: a systematic review of in vitro and in vivo studies. Xiaojie S, Yu L, Lei Y, Guang Y, Min Q. Neutralizing antibodies targeting SARS-CoV-2 spike protein.

Stem Cell Res. Case JB, Winkler ES, Errico JM, Diamond MS. On the road to ending the COVID pandemic: are we there yet? Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, Musser BJ, Soo Y, Rofail D, Im J, et al.

REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, Huhn G, Cardona J, Mocherla B, Stosor V, et al.

Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID a randomized clinical trial. Chen RE, Zhang X, Case JB, Winkler ES, Liu Y, VanBlargan LA, Liu J, Errico JM, Xie X, Suryadevara N et al.

Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies. Kuzmina A, Khalaila Y, Voloshin O, Keren-Naus A, Boehm-Cohen L, Raviv Y, Shemer-Avni Y, Rosenberg E, Taube R. SARS-CoV-2 spike variants exhibit differential infectivity and neutralization resistance to convalescent or post-vaccination sera.

Cell Host Microbe. Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B, Kwong PD et al. Increased Resistance of SARS-CoV-2 Variants B. Weisblum Y, Schmidt F, Zhang F, DaSilva J, Poston D, Lorenzi JC, Muecksch F, Rutkowska M, Hoffmann HH, Michailidis E et al.

Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. Hoffmann M, Hofmann-Winkler H, Krüger N, Kempf A, Nehlmeier I, Graichen L, Sidarovich A, Moldenhauer AS, Winkler MS, Schulz S et al. SARS-CoV-2 variant B. bioRxiv, Accessed July 7 th , Accessed July 7thth, Tada T, Zhou H, Dcostaa BM, Samanovicb MI, Mulliganb MJ, Landaua NR.

The Spike Proteins of SARS-CoV-2 B. Rappazzo CG, Tse LV, Kaku CI, Wrapp D, Sakharkar M, Huang D, Deveau LM, Yockachonis TJ, Herbert AS, Battles MB, et al. Broad and potent activity against SARS-like viruses by an engineered human monoclonal antibody.

Wec AZ, Wrapp D, Herbert AS, Maurer DP, Haslwanter D, Sakharkar M, Jangra RK, Dieterle ME, Lilov A, Huang D, et al. Broad neutralization of SARS-related viruses by human monoclonal antibodies. Starr TN, Czudnochowski N, Zatta F, Park YJ, Liu Z, Addetia A, Pinto D, Beltramello M, Hernandez P, Greaney AJ et al.

Antibodies to the SARS-CoV-2 receptor-binding domain that maximize breadth and resistance to viral escape. Tortorici MA, Czudnochowski N, Starr TN, Marzi R, Walls AC, Zatta F, Bowen JE, Jaconi S, Iulio JD, Wang Z et al.

Structural basis for broad sarbecovirus neutralization by a human monoclonal antibody. Baum A, Fulton BO, Wloga E, Copin R, Pascal KE, Russo V, Giordano S, Lanza K, Negron N, Ni M, et al.

Antibody cocktail to SARS-CoV-2 spike protein prevents rapid mutational escape seen with individual antibodies. Pinto D, Park YJ, Beltramello M, Walls AC, Tortorici MA, Bianchi S, Jaconi S, Culap K, Zatta F, De Marco A, et al. Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody.

Liu H, Yuan M, Huang D, Bangaru S, Zhao F, Lee CD, Peng L, Barman S, Zhu X, Nemazee D et al. A combination of cross-neutralizing antibodies synergizes to prevent SARS-CoV-2 and SARS-CoV pseudovirus infection. Sasisekharan R. P reparing for the Future - Nanobodies for Covid? Konwarh R: Nanobodies Prospects of expanding the gamut of neutralizing antibodies against the novel coronavirus, SARS-CoV Front Immunol.

Huo J, Le Bas A, Ruza RR, Duyvesteyn HME, Mikolajek H, Malinauskas T, Tan TK, Rijal P, Dumoux M, Ward PN, et al.

Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2. Nat Struct Mol Biol. Chi X, Liu X, Wang C, Zhang X, Li X, Hou J, Ren L, Jin Q, Wang J, Yang W.

Humanized single domain antibodies neutralize SARS-CoV-2 by targeting the spike receptor binding domain. Nat Commun. Koenig PA, Das H, Liu H, Kummerer BM, Gohr FN, Jenster LM, Schiffelers LDJ, Tesfamariam YM, Uchima M, Wuerth JD et al.

Structure-guided multivalent nanobodies block SARS-CoV-2 infection and suppress mutational escape. Xiang Y, Nambulli S, Xiao Z, Liu H, Sang Z, Duprex WP, Schneidman-Duhovny D, Zhang C, Shi Y.

Versatile and multivalent nanobodies efficiently neutralize SARS-CoV CAS PubMed PubMed Central Google Scholar. Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Cetrulo Lorenzi JC, Park S, Wang Z, Tessarollo L et al.

Multimeric nanobodies from camelid engineered mice and llamas potently neutralize SARS-CoV-2 variants. Schoof M, Faust B, Saunders RA, Sangwan S, Rezelj V, Hoppe N, Boone M, Billesbolle CB, Puchades C, Azumaya CM, et al. An ultrapotent synthetic nanobody neutralizes SARS-CoV-2 by stabilizing inactive Spike.

Bracken CJ, Lim SA, Solomon P, Rettko NJ, Nguyen DP, Zha BS, Schaefer K, Byrnes JR, Zhou J, Lui I, et al.

Bi-paratopic and multivalent VH domains block ACE2 binding and neutralize SARS-CoV Nat Chem Biol. Sun D, Sang Z, Kim YJ, Xiang Y, Cohen T, Belford AK, Huet A, Conway JF, Sun J, Taylor DJ et al. Potent neutralizing nanobodies resist convergent circulating variants of SARS-CoV-2 by targeting novel and conserved epitopes.

Mast FD, Fridy PC, Ketaren NE, Wang J, Jacobs EY, Olivier JP, Sanyal T, Molloy KR, Schmidt F, Rutkowska M et al. Nanobody repertoires for exposing vulnerabilities of SARS-CoV Pymm P, Adair A, Chan LJ, Cooney JP, Mordant FL, Allison CC, Lopez E, Haycroft ER, O'Neill MT, Tan LL et al.

Nanobody cocktails potently neutralize SARS-CoV-2 DG NY variant and protect mice. Proc Natl Acad Sci USA. Baum A, Ajithdoss D, Copin R, Zhou A, Lanza K, Negron N, Ni M, Wei Y, Mohammadi K, Musser B, et al.

REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Rosenfeld R, Noy-Porat T, Mechaly A, Makdasi E, Levy Y, Alcalay R, Falach R, Aftalion M, Epstein E, Gur D, et al.

Post-exposure protection of SARS-CoV-2 lethal infected KhACE2 transgenic mice by neutralizing human monoclonal antibody.

Kreye J, Reincke SM, Kornau HC, Sanchez-Sendin E, Corman VM, Liu H, Yuan M, Wu NC, Zhu X, Lee CD et al. A Therapeutic Non-self-reactive SARS-CoV-2 Antibody Protects from Lung Pathology in a COVID Hamster Model. Li W, Chen C, Drelich A, Martinez DR, Gralinski LE, Sun Z, Schafer A, Kulkarni SS, Liu X, Leist SR, et al.

Rapid identification of a human antibody with high prophylactic and therapeutic efficacy in three animal models of SARS-CoV-2 infection. Proc Natl Acad Sci U S A. Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Chong Z, Zost SJ, Jang H, Huang Y, Allen JD, et al.

Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions for optimal therapeutic protection. Phase III Double-blind, Placebo-controlled Study of AZD for Pre-exposure Prophylaxis of COVID in Adult.

COVID Study Assessing the Efficacy and Safety of Anti-Spike SARS CoV-2 Monoclonal Antibodies for Prevention of SARS CoV-2 Infection Asymptomatic in Healthy Adults and Adolescents Who Are Household Contacts to an Individual With a Positive SARS-CoV-2 RT-PCR Assay.

A Study of LY LY-CoV and LY LY-CoV in Preventing SARS-CoV-2 Infection and COVID in Nursing Home Residents and Staff BLAZE Monteil V, Kwon H, Prado P, Hagelkruys A, Wimmer RA, Stahl M, Leopoldi A, Garreta E, Hurtado Del Pozo C, Prosper F, et al.

Inhibition of SARS-CoV-2 infections in engineered human tissues using clinical-grade soluble human ACE2. Zoufaly A, Poglitsch M, Aberle JH, Hoepler W, Seitz T, Traugott M, Grieb A, Pawelka E, Laferl H, Wenisch C, et al. Human recombinant soluble ACE2 in severe COVID Lancet Respir Med.

Recombinant Human Angiotensin-converting Enzyme 2 rhACE2 as a Treatment for Patients With COVID APNCOVID Chan KK, Tan TJC, Narayanan KK, Procko E. An engineered decoy receptor for SARS-CoV-2 broadly binds protein S sequence variants. Sci Adv. Glasgow A, Glasgow J, Limonta D, Solomon P, Lui I, Zhang Y, Nix MA, Rettko NJ, Zha S, Yamin R, et al.

Engineered ACE2 receptor traps potently neutralize SARS-CoV Linsky TW, Vergara R, Codina N, Nelson JW, Walker MJ, Su W, Barnes CO, Hsiang TY, Esser-Nobis K, Yu K, et al.

De novo design of potent and resilient hACE2 decoys to neutralize SARS-CoV Pomplun S. Targeting the SARS-CoVspike protein: from antibodies to miniproteins and peptides.

RSC Med Chem. Article CAS Google Scholar. Cao L, Goreshnik I, Coventry B, Case JB, Miller L, Kozodoy L, Chen RE, Carter L, Walls AC, Park YJ, et al.

De novo design of picomolar SARS-CoV-2 miniprotein inhibitors. Schutz D, Ruiz-Blanco YB, Munch J, Kirchhoff F, Sanchez-Garcia E, Muller JA. Peptide and peptide-based inhibitors of SARS-CoV-2 entry.

Adv Drug Deliv Rev. Han DP, Penn-Nicholson A, Cho MW. Identification of critical determinants on ACE2 for SARS-CoV entry and development of a potent entry inhibitor.

Zheng BJ, Guan Y, Hez ML, Sun H, Du L, Zheng Y, Wong KL, Chen H, Chen Y, Lu L, et al. Synthetic peptides outside the spike protein heptad repeat regions as potent inhibitors of SARS-associated coronavirus. Antivir Ther. Ho TY, Wu SL, Chen JC, Wei YC, Cheng SE, Chang YH, Liu HJ, Hsiang CY.

Design and biological activities of novel inhibitory peptides for SARS-CoV spike protein and angiotensin-converting enzyme 2 interaction. Antiviral Res. Hu H, Li L, Kao RY, Kou B, Wang Z, Zhang L, Zhang H, Hao Z, Tsui WH, Ni A, et al.

Screening and identification of linear B-cell epitopes and entry-blocking peptide of severe acute respiratory syndrome SARS -associated coronavirus using synthetic overlapping peptide library. J Comb Chem. Karoyan P, Vieillard V, Gomez-Morales L, Odile E, Guihot A, Luyt CE, Denis A, Grondin P, Lequin O.

Human ACE2 peptide-mimics block SARS-CoV-2 pulmonary cells infection. Commun Biol. Curreli F, Victor SMB, Ahmed S, Drelich A, Tong X, Tseng CK, Hillyer CD, Debnath AK. Stapled Peptides Based on Human Angiotensin-Converting Enzyme 2 ACE2 Potently Inhibit SARS-CoV-2 Infection In Vitro.

Watson A, Ferreira, L MR, Hwang P, Xu J, Stroud R. Peptide antidotes to SARS-CoV-2 COVID Zhang G,Pomplun S, Loftis AR, Loas A, Pentelute BL. The first-in-class peptide binder to the SARS-CoV-2 spike protein. Zhang G,Pomplun S, Loftis AR, Tan X, Loas A, Pentelute BL.

Investigation of ACE2 N-terminal fragments binding to SARS- CoV-2 Spike RBD. Morgan DC, Morris C, Mahindra A, Blair CM, Tejeda G, Herbert I, Turnbull ML, Lieber G, Willett BJ, Logan N et al. Stapled ACE2 peptidomimetics designed to target the SARS-CoV-2 spike protein do not prevent virus internalization.

Pept Sci Hoboken. Xia S, Xu W, Wang Q, Wang C, Hua C, Li W, Lu L, Jiang S. Peptide-Based Membrane Fusion Inhibitors Targeting HCoVE Spike Protein HR1 and HR2 Domains. Int J Mol Sci. Bosch BJ, Martina BE, Van Der Zee R, Lepault J, Haijema BJ, Versluis C, Heck AJ, De Groot R, Osterhaus AD, Rottier PJ.

Severe acute respiratory syndrome coronavirus SARS-CoV infection inhibition using spike protein heptad repeat-derived peptides. Sun Y, Zhang H, Shi J, Zhang Z, Gong R.

Identification of a Novel Inhibitor against Middle East Respiratory Syndrome Coronavirus. Xia S, Yan L, Xu W, Agrawal AS, Algaissi A, Tseng CK, Wang Q, Du L, Tan W, Wilson IA et al.

A pan-coronavirus fusion inhibitor targeting the HR1 domain of human coronavirus spike. Xia S, Liu M, Wang C, Xu W, Lan Q, Feng S, Qi F, Bao L, Du L, Liu S, et al. Inhibition of SARS-CoV-2 previously nCoV infection by a highly potent pan-coronavirus fusion inhibitor targeting its spike protein that harbors a high capacity to mediate membrane fusion.

Cell Res. Zhu Y, Yu D, Yan H, Chong H, He Y. Design of Potent Membrane Fusion Inhibitors against SARS-CoV-2, an Emerging Coronavirus with High Fusogenic Activity. J Virol. de Vries RD, Schmitz KS, Bovier FT, Predella C, Khao J, Noack D, Haagmans BL, Herfst S, Stearns KN, Drew-Bear J, et al.

Intranasal fusion inhibitory lipopeptide prevents direct-contact SARS-CoV-2 transmission in ferrets. Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Pharmacologic treatments for coronavirus disease COVID : a review. Xiu S, Dick A, Ju H, Mirzaie S, Abdi F, Cocklin S, Zhan P, Liu X.

Inhibitors of SARS-CoV-2 entry: current and future opportunities. J Med Chem. Khare P, Sahu U, Pandey SC, Samant M. Current approaches for target-specific drug discovery using natural compounds against SARS-CoV-2 infection.

Virus Res. Artese A, Svicher V, Costa G, Salpini R, Di Maio VC, Alkhatib M, Ambrosio FA, Santoro MM, Assaraf YG, Alcaro S, et al. Current status of antivirals and druggable targets of SARS CoV-2 and other human pathogenic coronaviruses. Jan JT, Cheng TR, Juang YP, Ma HH, Wu YT, Yang WB, Cheng CW, Chen X, Chou TH, Shie JJ et al.

Identification of existing pharmaceuticals and herbal medicines as inhibitors of SARS-CoV-2 infection. Ohashi H, Watashi K, Saso W, Shionoya K, Iwanami S, Hirokawa T, Shirai T, Kanaya S, Ito Y, Kim KS et al. Potential anti-COVID agents, cepharanthine and nelfinavir, and their usage for combination treatment.

Dittmar M, Lee JS, Whig K, Segrist E, Li M, Kamalia B, Castellana L, Ayyanathan K, Cardenas-Diaz FL, Morrisey EE, et al. Drug repurposing screens reveal cell-type-specific entry pathways and FDA-approved drugs active against SARS-Cov Cell Rep. Jeon S, Ko M, Lee J, Choi I, Byun SY, Park S, Shum D, Kim S.

Identification of antiviral drug candidates against SARS-CoV-2 from FDA-approved drugs. Ko M, Jeon S, Ryu WS, Kim S. Comparative analysis of antiviral efficacy of FDA-approved drugs against SARS-CoV-2 in human lung cells.

Riva L, Yuan S, Yin X, Martin-Sancho L, Matsunaga N, Pache L, Burgstaller-Muehlbacher S, De Jesus PD, Teriete P, Hull MV, et al. Discovery of SARS-CoV-2 antiviral drugs through large-scale compound repurposing.

He CL, Huang LY, Wang K, Gu CJ, Hu J, Zhang GJ, Xu W, Xie YH, Tang N, Huang AL. Identification of bis-benzylisoquinoline alkaloids as SARS-CoV-2 entry inhibitors from a library of natural products.

Signal Transduct Target Ther. Hanson QM, Wilson KM, Shen M, Itkin Z, Eastman RT, Shinn P, Hall MD. Targeting ACE2-RBD interaction as a platform for COVID therapeutics: development and drug-repurposing screen of an alphalisa proximity assay. ACS Pharmacol Transl Sci.

Day CJ, Bailly B, Guillon P, Dirr L, Jen FE, Spillings BL, Mak J, von Itzstein M, Haselhorst T, Jennings MP. Multidisciplinary Approaches Identify Compounds that Bind to Human ACE2 or SARS-CoV-2 Spike Protein as Candidates to Block SARS-CoVACE2 Receptor Interactions.

Nabavi SF, Habtemariam S, Berindan-Neagoe I, Cismaru CA, Schaafsma D, Ghavami S, Banach M, Aghaabdollahian S, Nabavi SM. Rationale for effective prophylaxis against COVID through simultaneous blockade of both endosomal and non-endosomal SARS-CoV-2 Entry into Host Cell.

Clin Transl Sci. Cannalire R, Stefanelli I, Cerchia C, Beccari AR, Pelliccia S, Summa V. SARS-CoV-2 Entry Inhibitors: Small Molecules and Peptides Targeting Virus or Host Cells.

Bestle D, Heindl MR, Limburg H, Van Lam van T, Pilgram O, Moulton H, Stein DA, Hardes K, Eickmann M, Dolnik O et al.

TMPRSS2 and furin are both essential for proteolytic activation of SARS-CoV-2 in human airway cells. Life Sci Alliance. Hoffmann M, Kleine-Weber H, Pohlmann S. A multibasic cleavage site in the spike protein of SARS-CoV-2 is essential for infection of human lung cells.

Mol Cell. Böttcher-Friebertshäuser E. Membrane-anchored serine proteases: host cell factors in proteolytic activation of viral glycoproteins. Activation of Viruses by Host Proteases. Zhirnov OP, Klenk HD, Wright PF. Aprotinin and similar protease inhibitors as drugs against influenza.

Yamamoto M, Kiso M, Sakai-Tagawa Y, Iwatsuki-Horimoto K, Imai M, Takeda M, Kinoshita N, Ohmagari N, Gohda J, Semba K et al. The Anticoagulant Nafamostat Potently Inhibits SARS-CoV-2 S Protein-Mediated Fusion in a Cell Fusion Assay System and Viral Infection In Vitro in a Cell-Type-Dependent Manner.

Hoffmann M, Schroeder S, Kleine-Weber H, Muller MA, Drosten C, Pohlmann S. Nafamostat mesylate blocks activation of SARS-CoV new treatment option for COVID Yang N, Shen HM. Targeting the endocytic pathway and autophagy process as a novel therapeutic strategy in COVID Int J Biol Sci.

Das G, Ghosh S, Garg S, Ghosh S, Jana A, Samat R, Mukherjee N, Roya R, Ghosh S. An overview of key potential therapeutic strategies for combat in the covid battle. RSC Adv. Zhang J, Ma X, Yu F, Liu J, Zou F, Pan T, Zhang H.

Ou X, Liu Y, Lei X, Li P, Mi D, Ren L, Guo L, Guo R, Chen T, Hu J, et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Zhao H, To KKW, Sze KH, Yung TT, Bian M, Lam H, Yeung ML, Li C, Chu H, Yuen KY.

A broad-spectrum virus- and host-targeting peptide against respiratory viruses including influenza virus and SARS-CoV Zhao H, To KKW, Lam H, Zhou X, Chan JF, Peng Z, Lee ACY, Cai J, Chan WM, Ip JD, et al. Cross-linking peptide and repurposed drugs inhibit both entry pathways of SARS-CoV Andreani J, Le Bideau M, Duflot I, Jardot P, Rolland C, Boxberger M, Wurtz N, Rolain JM, Colson P, La Scola B et al.

In vitro testing of combined hydroxychloroquine and azithromycin on SARS-CoV-2 shows synergistic effect. Microb Pathog. Haydar D, Cory TJ, Birket SE, Murphy BS, Pennypacker KR, Sinai AP, Feola DJ. Azithromycin polarizes macrophages to an M2 phenotype via inhibition of the STAT1 and NF-kappaB signaling pathways.

J Immunol. Nujic K, Banjanac M, Munic V, Polancec D, Erakovic Haber V. Impairment of lysosomal functions by azithromycin and chloroquine contributes to anti-inflammatory phenotype.

Cell Immunol. Shang C, Zhuang X, Zhang H, Li Y, Zhu Y, Lu J, Ge C, Cong J, Li T, Tian M, et al. Inhibitors of endosomal acidification suppress SARS-CoV-2 replication and relieve viral pneumonia in hACE2 transgenic mice.

Virol J. Galan LEB, Santos NMD, Asato MS, Araujo JV, de Lima Moreira A, Araujo AMM, Paiva ADP, Portella DGS, Marques FSS, Silva GMA et al. Phase 2 randomized study on chloroquine, hydroxychloroquine or ivermectin in hospitalized patients with severe manifestations of SARS-CoV-2 infection.

Pathog Glob Health. Cavalcanti AB, Zampieri FG, Rosa RG, Azevedo LCP, Veiga VC, Avezum A, Damiani LP, Marcadenti A, Kawano-Dourado L, Lisboa T, et al. Hydroxychloroquine with or without Azithromycin in Mild-to-Moderate Covid Shagufta Ahmad I. The race to treat COVID Potential therapeutic agents for the prevention and treatment of SARS-CoV Eur J Med Chem.

Stamatatos L, Czartoski J, Wan YH, Homad LJ, Rubin V, Glantz H, Neradilek M, Seydoux E, Jennewein MF, MacCamy AJ et al, mRNA vaccination boosts cross-variant neutralizing antibodies elicited by SARS-CoV-2 infection.

Lopez Bernal J, Andrews N, Gower C, Gallagher E, Utsi L, Simmons R, Thelwall S, Stowe J, Tessier E, Groves N, Dabrera G et al. Effectiveness of COVID vaccines against the B. Julia Stowe , Nick Andrews, Charlotte Gower , Eileen Gallagher, Lara Utsi , Ruth Simmons, Simon Thelwall, Elise Tessier, Natalie Groves, Gavin Dabrera et al.

Effectiveness of COVID vaccines against hospital admission with the Delta B. PHE Preprint, Katella K. Comparing the COVID Vaccines: How Are They Different? Meganck RM, Baric RS. Developing therapeutic approaches for twenty-first-century emerging infectious viral diseases.

National security directive united states global leadership to strengthen the international covid response and to advance global health security and biological preparedness. Global leaders unite in urgent call for international pandemic treaty.

Accessed July 7th , Download references. Department of Basic Sciences, Loma Linda University School of Medicine, Campus Street, Alumni Hall, Loma Linda, CA, , USA. You can also search for this author in PubMed Google Scholar.

LC wrote the manuscript. LC and PDH edited the manuscript. Both authors read and approved the manuscript. Correspondence to Penelope Duerksen-Hughes. Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Open Access This article is licensed under a Creative Commons Attribution 4. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder.

Reprints and permissions. Chitsike, L. Keep out! SARS-CoV-2 entry inhibitors: their role and utility as COVID therapeutics.

Virol J 18 , Download citation. Received : 25 May Accepted : 15 July Published : 23 July Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Skip to main content. Search all BMC articles Search.

Download PDF. Review Open access Published: 23 July Keep out! Abstract The COVID pandemic has put healthcare infrastructures and our social and economic lives under unprecedented strain. Introduction COVID, the disease caused by the novel coronavirus SARS-CoV-2, was declared a pandemic and global emergency shortly after it began in late Full size image.

Viral entry inhibitors and their translational relevance The availability of several effective vaccines against SARS-CoV-2 has given hope to billions of people across the globe [ 21 , 22 ]. Table 1 Prominent examples of viral entry inhibitors that have demonstrated therapeutic or prophylactic efficacy in cross-neutralization, suppression of escape mutants and broad activity against circulating variants and sarbecoviruses Full size table.

Host proteases and endosome acidification inhibitors Although S1 and S2 mediate viral attachment and membrane fusion to enable the virus to unload its genetic cargo, function of these two subunits is enabled by the participation of at least 3 types of host proteases: furins, cathepsins and surface serine proteases.

Furin and TMPRSS2 inhibition Furin inhibitors have previously been reported as possible targets in the context of other viruses such as influenza, and may also be relevant for SARS-CoV Cathepsin inhibition A number of cathepsin inhibitors against coronaviruses have also been reported in various studies.

Conclusions and future perspectives COVID is now understood as a biphasic illness, with an early viral phase and a more dangerous host-immune response phase.

Availability of data and materials All materials available in this article. References Nicola M, Alsafi Z, Sohrabi C, Kerwan A, Al-Jabir A, Iosifidis C, Agha M, Agha R. Article PubMed PubMed Central Google Scholar Cucinotta D, Vanelli M.

PubMed PubMed Central Google Scholar Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Article CAS PubMed Google Scholar Izda V, Jeffries MA, Sawalha AH. Article CAS PubMed Google Scholar Sun P, Lu X, Xu C, Sun W, Pan B. Article CAS PubMed Google Scholar Hu B, Guo H, Zhou P, Shi ZL.

Article CAS PubMed Google Scholar Harrison AG, Lin T, Wang P. Article CAS PubMed PubMed Central Google Scholar Hou YJ, Okuda K, Edwards CE, Martinez DR, Asakura T, Dinnon KH 3rd, Kato T, Lee RE, Yount BL, Mascenik TM, et al.

Article CAS PubMed PubMed Central Google Scholar Mason RJ. Article CAS PubMed PubMed Central Google Scholar Stratton CW, Tang YW, Lu H. Article CAS PubMed Google Scholar Sette A, Crotty S.

Article CAS PubMed PubMed Central Google Scholar Le Bert N, Clapham HE, Tan AT, Chia WN, Tham CYL, Lim JM, Kunasegaran K, Tan LWL, Dutertre CA, Shankar N et al. Article CAS PubMed Google Scholar Twomey JD, Luo S, Dean AQ, Bozza WP, Nalli A, Zhang B.

Article PubMed PubMed Central Google Scholar Schulte-Schrepping J, Reusch N, Paclik D, Bassler K, Schlickeiser S, Zhang B, Kramer B, Krammer T, Brumhard S, Bonaguro L, et al.

Article CAS PubMed PubMed Central Google Scholar Sundararaj Stanleyraj J, Sethuraman N, Gupta R, Thiruvoth S, Gupta M, Ryo A. Article PubMed CAS Google Scholar Covid Treatment guidelines. Accessed July 7th, Kim PS, Read SW, Fauci AS.

Article CAS PubMed Google Scholar Cohen MS. Article CAS PubMed PubMed Central Google Scholar Meo SA, Bukhari IA, Akram J, Meo AS, Klonoff DC. CAS PubMed Google Scholar COVID vaccines. Article CAS PubMed PubMed Central Google Scholar Rosenbaum L. Article CAS PubMed Google Scholar Fontanet A, Cauchemez S.

Article PubMed CAS PubMed Central Google Scholar Kwok KO, Lai F, Wei WI, Wong SYS, Tang JWT. Article CAS PubMed PubMed Central Google Scholar Alaran AJ, Adebisi YA, Badmos A, Khalid-Salako F, Gaya SK, Ilesanmi EB, Olaoye DQ, Bamisaiye A, Lucero-Prisno DE.

Article PubMed PubMed Central Google Scholar Lucero-Prisno DE, Ogunkola IO, Imo UF, Adebisi YA. Accessed July 7th, COVAX: Working for global equitable access to covid vaccines.

Accessed July 7th, Sonani B, Aslam F, Goyal A, Patel J, Bansal P. Article PubMed Google Scholar Coustasse A, Kimble C, Maxik K. Article PubMed Google Scholar Group C. Article Google Scholar Altmann DM, Boyton RJ, Beale R.

Article CAS PubMed Google Scholar Abdool Karim SS, de Oliveira T: New SARS-CoV-2 Variants - Clinical, Public Health, and Vaccine Implications. Accessed July 7th, Tracking SARS-CoV-2 variants. Accessed July 7th, Khan A, Zia T, Suleman M, Khan T, Ali SS, Abbasi AA, Mohammad A, Wei DQ.

Article CAS PubMed Google Scholar Davis C, Logan N, Tyson G, Orton R, Harvey W, Haughney J, Perkins J, The COVID genomics UK COG-UK consortium, peacock TP, Barclay WS et al. Accessed July 7th, Increased transmissibility and global spread of SARS-CoV-2 variants of concern as at June Accessed July 7th, Singh TU, Parida S, Lingaraju MC, Kesavan M, Kumar D, Singh RK.

Article PubMed CAS PubMed Central Google Scholar Pawar AY. Article CAS PubMed PubMed Central Google Scholar Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Article CAS PubMed PubMed Central Google Scholar Li F. Article CAS PubMed PubMed Central Google Scholar Huang Y, Yang C, Xu XF, Xu W, Liu SW.

Article PubMed PubMed Central CAS Google Scholar Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al. Article CAS PubMed PubMed Central Google Scholar Martinez MA. Article PubMed PubMed Central Google Scholar Peng HT, Rhind SG, Beckett A.

Article CAS PubMed PubMed Central Google Scholar Wang Y, Huo P, Dai R, Lv X, Yuan S, Zhang Y, Guo Y, Li R, Yu Q, Zhu K. Article CAS PubMed Google Scholar Renn A, Fu Y, Hu X, Hall MD, Simeonov A. Article CAS PubMed PubMed Central Google Scholar Jiang S, Zhang X, Yang Y, Hotez PJ, Du L.

Article CAS PubMed PubMed Central Google Scholar Seyedpour S, Khodaei B, Loghman AH, Seyedpour N, Kisomi MF, Balibegloo M, Nezamabadi SS, Gholami B, Saghazadeh A, Rezaei N.

Article CAS PubMed Google Scholar Xiaojie S, Yu L, Lei Y, Guang Y, Min Q. Article CAS PubMed Google Scholar Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, Musser BJ, Soo Y, Rofail D, Im J, et al.

Article CAS PubMed Google Scholar Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, Huhn G, Cardona J, Mocherla B, Stosor V, et al. Accessed July 7th, Chen RE, Zhang X, Case JB, Winkler ES, Liu Y, VanBlargan LA, Liu J, Errico JM, Xie X, Suryadevara N et al.

Article CAS PubMed PubMed Central Google Scholar Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B, Kwong PD et al. Accessed July 7thth, Tada T, Zhou H, Dcostaa BM, Samanovicb MI, Mulliganb MJ, Landaua NR. Article CAS PubMed PubMed Central Google Scholar Wec AZ, Wrapp D, Herbert AS, Maurer DP, Haslwanter D, Sakharkar M, Jangra RK, Dieterle ME, Lilov A, Huang D, et al.

Article CAS PubMed PubMed Central Google Scholar Starr TN, Czudnochowski N, Zatta F, Park YJ, Liu Z, Addetia A, Pinto D, Beltramello M, Hernandez P, Greaney AJ et al. Article CAS PubMed Google Scholar Pinto D, Park YJ, Beltramello M, Walls AC, Tortorici MA, Bianchi S, Jaconi S, Culap K, Zatta F, De Marco A, et al.

Article CAS PubMed Google Scholar Liu H, Yuan M, Huang D, Bangaru S, Zhao F, Lee CD, Peng L, Barman S, Zhu X, Nemazee D et al. Article CAS PubMed Google Scholar Chi X, Liu X, Wang C, Zhang X, Li X, Hou J, Ren L, Jin Q, Wang J, Yang W. Article CAS PubMed PubMed Central Google Scholar Koenig PA, Das H, Liu H, Kummerer BM, Gohr FN, Jenster LM, Schiffelers LDJ, Tesfamariam YM, Uchima M, Wuerth JD et al.

CAS PubMed PubMed Central Google Scholar Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Cetrulo Lorenzi JC, Park S, Wang Z, Tessarollo L et al. CAS PubMed PubMed Central Google Scholar Bracken CJ, Lim SA, Solomon P, Rettko NJ, Nguyen DP, Zha BS, Schaefer K, Byrnes JR, Zhou J, Lui I, et al.

Article PubMed Google Scholar Sun D, Sang Z, Kim YJ, Xiang Y, Cohen T, Belford AK, Huet A, Conway JF, Sun J, Taylor DJ et al. Article CAS PubMed PubMed Central Google Scholar Rosenfeld R, Noy-Porat T, Mechaly A, Makdasi E, Levy Y, Alcalay R, Falach R, Aftalion M, Epstein E, Gur D, et al.

Article CAS PubMed PubMed Central Google Scholar Kreye J, Reincke SM, Kornau HC, Sanchez-Sendin E, Corman VM, Liu H, Yuan M, Wu NC, Zhu X, Lee CD et al. Article CAS PubMed PubMed Central Google Scholar Winkler ES, Gilchuk P, Yu J, Bailey AL, Chen RE, Chong Z, Zost SJ, Jang H, Huang Y, Allen JD, et al.

Article CAS PubMed PubMed Central Google Scholar Phase III Double-blind, Placebo-controlled Study of AZD for Pre-exposure Prophylaxis of COVID in Adult. Article CAS PubMed PubMed Central Google Scholar Zoufaly A, Poglitsch M, Aberle JH, Hoepler W, Seitz T, Traugott M, Grieb A, Pawelka E, Laferl H, Wenisch C, et al.

Article CAS PubMed PubMed Central Google Scholar Recombinant Human Angiotensin-converting Enzyme 2 rhACE2 as a Treatment for Patients With COVID APNCOVID Article CAS PubMed PubMed Central Google Scholar Linsky TW, Vergara R, Codina N, Nelson JW, Walker MJ, Su W, Barnes CO, Hsiang TY, Esser-Nobis K, Yu K, et al.

Article CAS PubMed PubMed Central Google Scholar Pomplun S. Article CAS Google Scholar Cao L, Goreshnik I, Coventry B, Case JB, Miller L, Kozodoy L, Chen RE, Carter L, Walls AC, Park YJ, et al.

Article CAS PubMed PubMed Central Google Scholar Schutz D, Ruiz-Blanco YB, Munch J, Kirchhoff F, Sanchez-Garcia E, Muller JA. Article PubMed PubMed Central CAS Google Scholar Han DP, Penn-Nicholson A, Cho MW.

Article CAS PubMed Google Scholar Zheng BJ, Guan Y, Hez ML, Sun H, Du L, Zheng Y, Wong KL, Chen H, Chen Y, Lu L, et al. CAS PubMed Google Scholar Ho TY, Wu SL, Chen JC, Wei YC, Cheng SE, Chang YH, Liu HJ, Hsiang CY.

Article CAS PubMed Google Scholar Hu H, Li L, Kao RY, Kou B, Wang Z, Zhang L, Zhang H, Hao Z, Tsui WH, Ni A, et al. Article CAS PubMed Google Scholar Karoyan P, Vieillard V, Gomez-Morales L, Odile E, Guihot A, Luyt CE, Denis A, Grondin P, Lequin O.

Article CAS PubMed PubMed Central Google Scholar Curreli F, Victor SMB, Ahmed S, Drelich A, Tong X, Tseng CK, Hillyer CD, Debnath AK. Article CAS PubMed PubMed Central Google Scholar Sun Y, Zhang H, Shi J, Zhang Z, Gong R.

Article CAS PubMed PubMed Central Google Scholar Zhu Y, Yu D, Yan H, Chong H, He Y. Article PubMed PubMed Central CAS Google Scholar Sanders JM, Monogue ML, Jodlowski TZ, Cutrell JB. Article CAS PubMed Google Scholar Xiu S, Dick A, Ju H, Mirzaie S, Abdi F, Cocklin S, Zhan P, Liu X.

General Concepts Soluutions cells that have lost their ability to produce IFN. Expert Anti-cancer mind-body practices Ther Meal planning ideas. Gomez CE, Perdiguero B, Esteban M. N forms a covalent bond with the catalytic triad residue Ser Therefore, more effective and appropriate targets need to be developed by researchers. Article CAS Google Scholar Praetorius, F.
Control of Viral Infections and Diseases - Medical Microbiology - NCBI Bookshelf

In particular, the recent emergence of a novel, heavily mutated VOC, B. Discovering novel classes of antiviral compounds—including both direct-acting antivirals DAAs and host-directed antivirals HDAs —and intensive in cellulo and in vivo studies of their antiviral profiles as mono- or combination therapies against emerging SARS-CoV-2 VOCs are critical for developing preventive and therapeutic strategies to combat COVID 13 , At present, three antivirals have been approved for clinical use against SARS-CoV Remdesivir is a DAA that targets the viral RNA-dependent RNA polymerase that catalyses the synthesis of viral RNA Remdesivir is administered intravenously to hospitalized individuals with COVID Paxlovid targets the main protease of the coronavirus 3CL pro , also known as M pro , an essential protease that is involved in processing viral replicase polyproteins, whereas molnupiravir is a ribonucleoside analogue that inhibits viral replication 17 , Alternatively, HDAs also termed indirect-acting antivirals are under investigation and may offer a complement to DAAs.

Emerging SARS-CoV-2 VOCs are less likely to develop resistance to HDAs than to DAAs because, unlike viral genes, host genes have a low propensity to mutate 5 , Camostat mesylate Cm , for example, is a broad spectrum serine protease inhibitor used to treat pancreatitis that has been repositioned as a clinical candidate for treating COVID 4 , 5 , Accumulating evidence has shown that SARS-CoV-2 is dependent on host pathways, including the hijacking of TMPRSS2-related proteases for viral entry; this suggests that TTSPs could be therapeutic targets to prevent SARS-CoV-2 infection 5 , 6 , The SARS-CoV-2 lifecycle begins with attachment and entry into respiratory epithelium via the angiotensin-converting enzyme 2 ACE2 receptor 4 , 8.

This is mediated by the major viral surface glycoprotein, spike S , which must undergo two sequential proteolytic cleavages by host proteases before it can mediate fusion of the virus with host cell membranes, a requirement for subsequent viral replication 3 , 21 , This cleavage, which triggers the fusion event, is mediated by host TTSPs such as TMPRSS2 and TMPRSS13, which cleave after specific single arginine or lysine residues 4 , The KhACE2 mouse model transgenic expression of human ACE2 hACE2 under a cytokeratin 18 promoter offers a stringent system for testing the efficacy of DAAs and HDAs against severe disease and mortality after SARS-CoV-2 infection So far, only a few studies have tested antiviral efficacy in this animal model, with only one DAA reported as protective against lethal SARS-CoV-2 infection in this model 25 , Here we report on the design and testing of peptidomimetics for their inhibitory activity against TMPRSS2 and related TTSPs.

We then investigated the antiviral activities of the peptidomimetics against an ancestral strain of SARS-CoV-2 lineage B, VIDO and four variants—B. Finally, we tested our top highly potent antiviral, N, against SARS-CoV-2 lineage A strain and SARS-CoV-2 Delta-induced morbidity and mortality in KhACE2 mice.

We found that N provides a high level of protection and a therapeutic benefit after either multiple administrations or a single administration in this model of severe disease. Thus, N is an antiviral with a high potential for use against COVID We previously designed first-generation peptidomimetic tetrapeptide compounds with ketobenzothiazole warheads, and they exhibited inhibitory activity against a host TTSP, matriptase 27 , These compounds act as slow tight-binding inhibitors in vitro but their potency in cellular systems was modest against influenza A virus To improve their stability and potency, we modified their N terminus either by capping or through the synthesis of desamino moieties 29 Fig.

Moreover, these compounds exhibited low nanomolar efficacies when tested in H1N1 models of influenza A virus infection 28 , a , Peptidomimetic compounds used in this study along with their respective sequences. The structures of N-terminal caps, the ketobenzothiazole warhead and the alcohol ketobenzothiazole are shown on the right.

H Arg, desamino arginine; kbt, ketobenzothiazol. Relative TMPRSS2 activity was measured using the mock-subtracted fluorescence and is reported as the percentage of residual activity relative to the vehicle-treated cells 0. c , Dose—response curves were generated for the indicated compounds using the assay described in b , and IC 50 values were determined using nonlinear regression analysis.

d , Specificity of selected compounds toward other serine proteases. e , Main image, docking of N green; warhead in purple in the binding pocket of TMPRSS2 homology model. Residues of the catalytic triad are shown in cyan. Inset, interaction of N with TMPRSS2 residues.

N forms a covalent bond with the catalytic triad residue Ser Source data. Expanding on that work here, we developed a small library of peptidomimetic compounds Fig. We included in this screen our first-generation tetrapeptide 28 , N, which lacks an N-terminal stabilizing group, along with three desamino tetrapeptide analogues.

We also tested four tripeptides containing different N-terminal capping groups. To evaluate the efficacies of these compounds, we set up a cellular assay to measure TMPRSS2-dependent pericellular inhibition of proteolytic activity. We expressed the full-length, wild-type TMPRSS2 or an inactive form of the protease in which the serine residue of the catalytic triad was replaced by alanine TMPRSS2 SA in Vero E6 cells.

Using this assay, we show that, as expected, the SA substitution completely abrogated the proteolytic activity of TMPRSS2. The first-generation peptidomimetic, N, did not inhibit TMPRSS2 activity under these conditions.

Notably, several peptidomimetic compounds were more efficient than Cm at reducing the activity of TMPRSS2 Fig.

We then investigated the dose response of the four most promising peptidomimetics N, N, N and N The half-maximal inhibitory concentration IC 50 of Cm was To confirm the contribution of the ketobenzothiazole warhead to the inhibitory activity of the molecule, the ketone functional group of N was replaced with an alcohol group to generate N OH Fig.

We also confirmed the efficacy of N against mouse TMPRSS2 with an IC 50 of Next, we sought to determine the selectivity profile of these inhibitors by measuring the inhibition constant K i on selected recombinant serine proteases, including three members of the TTSP family matriptase, hepsin and DESC1 as well as furin, thrombin, and cathepsin L.

All four of the tested peptidomimetic compounds behaved as low nanomolar inhibitors for the TTSPs, but they were inactive or showed only weak inhibition against the other proteases Fig.

Overall, these data show that TTSP-targeting peptidomimetics containing a ketobenzothiazole warhead inhibit TMPRSS2-dependent pericellular activity in a cellular assay and preferentially inhibit other members of the TTSP family.

To understand the mode of binding and the main interactions of our inhibitors and how these compounds achieve their high inhibitory potential, we built a homology model of TMPRSS2 using the crystal structure of matriptase Protein Data Bank PDB : 6N4T Docking of N was modelled to this structure Fig.

As predicted and previously published 32 , the catalytic triad Ser catalytic triad: Ser, His, and Asp; Fig. Several key interactions can be observed in the binding pocket. As in all TTSP inhibitors possessing a guanidine group on the sidechain, a strong hydrogen bond network stabilizes this pharmacophore deep within the binding pocket Fig.

This includes Asp and Gly, as well as Gln via a water molecule. Gln is also involved in another hydrogen bond of this same water molecule to the oxygen of the main-chain ketone group.

This ketone also acts as a hydrogen bond acceptor with Gly The N-terminal mesylate forms two hydrogen bonds—one intramolecular with the side-chain amide of the Gln residue of N, and another with Gly Finally, the oxygen of the newly formed hemiacetal is stabilized by two hydrogen bond donors from the Gly and Ser amines.

A portion of the ketobenzothiazole warhead and the aromatic ring from the phenylalanine are exposed to the solvents, which could allow us to further optimize the design of this second-generation inhibitor, leading to an improved pharmacokinetic profile.

The peptidomimetic compounds that we screened against TMPRSS2 were subsequently tested for their efficacy at preventing SARS-CoV-2 infection. Cells were fixed and immunofluorescently stained for double-stranded RNA dsRNA , a marker of viral replication 33 , and for the viral nucleocapsid, a marker of viral entry and translation 6 Extended Data Fig.

Fluorescent high-content imaging and relative quantification of virally infected cells showed consistent inhibitory profiles across dsRNA and nucleocapsid staining, which mirrored the inhibitory profile observed in the TMPRSS2 proteolytic activity assay Fig. Thus, TMPRSS2-inhibiting peptidomimetics are also inhibitors of SARS-CoV-2 replication and translation in Calu-3 cells, and the stabilizing N-terminal caps and the ketobenzothiazole warhead are likely to be essential for compound stability and antiviral potency.

Intracellular infection was relatively quantified using N staining. Representative fluorescent images of colonoids subjected to the indicated treatments are shown Hoechst in blue, nucleocapsid in red and dsRNA in green.

Error bars, s. The half-maximal effective concentration EC 50 of Cm was Thus, the selectivity index for these compounds N, N, N and N was between 8.

Overall, these results confirm that two TTSP-targeted peptidomimetic compounds N and N are extremely potent low nanomolar inhibitors of SARS-CoV-2 infection in human lung epithelial cells. a , Representative fluorescent images of SARS-CoVinfected Calu-3 cells. Calu-3 cells infected with the indicated SARS-CoV-2 variants and mock infected are shown.

b , Representative images from a 3D volume rendering of Delta-infected cells. In a , b , Hoechst is shown in blue, nucleocapsid N in red, dsRNA in green and actin in cyan; images were captured with a Leica TCS SP8 3× STED microscope. We next examined the effects of Cm, N and N OH on the extracellular release of SARS-CoV-2 infectious virions from Calu-3 cells.

These results confirm that N, which targets TMPRSS2, is a potent inhibitor of SARS-CoV-2 infectivity in Calu-3 cells and that the ketobenzothiazole warhead is required for N antiviral potency. Although Calu-3 cells represent a scalable and clinically relevant system of antiviral screening for SARS-CoV-2 inhibitors, they are an immortalized cell line.

To evaluate the effectiveness of N in a primary human cell-based model, we examined SARS-CoV-2 infection in donor-derived human colonoids 7 , SARS-CoV-2 initially causes a respiratory infection, but many infected individuals also experience gastrointestinal symptoms that are frequently linked with increased disease duration and severity A recent report identified TMPRSS2 as an essential co-factor for SARS-CoV-2 infection in colonoids We first relatively quantified the mRNA expression of ACE2 and TMPRSS2 in colonoids and Calu-3 cells using quantitative PCR qPCR.

ACE2 showed comparable levels of expression in colonoids compared to Calu-3 cells, whereas TMPRSS2 had much higher expression levels in colonoids compared to Calu-3 cells Extended Data Fig. We then investigated the susceptibility of colonoid monolayers to SARS-CoV-2 infection.

Consistent with previous work, the colonoids were susceptible to infection, as evidenced by dsRNA and nucleocapsid staining Fig. N and N OH were then tested for their efficacy at preventing SARS-CoV-2 infection in colonoids. These results align with observations in Calu-3 cells and confirm the nanomolar potency of N against SARS-CoV-2 in primary human cells.

To our knowledge, mutations in the TMPRSS2 cleavage site have not been identified in SARS-CoV-2 variants, which suggests that N should retain high potency against SARS-CoV-2 VOCs First, we confirmed the infectivity of four VOCs in Calu-3 cells: B.

Confocal imaging of infected cells confirmed the infectivity of these variants, as demonstrated by nucleocapsid and dsRNA staining Fig. Although the viral marker staining patterns were relatively consistent in Calu-3 cells infected with a lineage B isolate VIDO , B.

We then evaluated the efficacy of N for preventing infection with SARS-CoV-2 VOCs in Calu-3 cells. The EC 50 of N against all VOCs was in the low nanomolar range, ranging from 2. This underscores the potential of N to act as a pan-variant, host-directed antiviral against emerging SARS-CoV-2 VOCs.

After establishing the efficacy of N in vitro and in cellulo, we tested whether intranasal administration would improve morbidity and survival in vivo, using KhACE2 mice 36 , 37 , an established mouse model of severe COVID Dosing regimens and drug concentrations were chosen on the basis of preliminary studies performed in a mouse model of influenza A virus infection, which showed antiviral efficacy at 7.

Ten mice per group five females and five males were administered a single daily intranasal dose of 7. Surviving mice were euthanized at the study end-point. b — d , Weight change of mice treated with saline control b , N OH c or N d. f , Probability of survival.

Uninfected mice tissues i, ii, ix, x were normal. Challenged mice iii—viii, xi—xvi developed perivascular infiltrates of inflammatory cells arrowheads. Severe inflammatory changes including alveolar fibrin and oedema asterisks were found only in the saline group iii, iv, xi, xii. Perivascular inflammatory cell infiltrates arrowheads were more widespread in saline iii, xi and control N OH v, xiii compared to N mice vii, xv.

Surviving N mice vii, viii, xv, xvi had smaller and fewer perivascular inflammatory infiltrates arrowheads and occasional type II pneumocyte hyperplasia red arrows. Saline-treated mice i, ii, ix, x developed perivascular cuffs of inflammatory cells asterisks , necrotic neurons arrows , gliosis and meningeal infiltrates arrowheads.

Brain lesions were reduced in N OH mice v, vi, xiii, xiv and absent in surviving N mice vii, viii, xv, xvi. The magnified areas were selected to best represent the presence of inflammatory cells and pathological changes.

Compared to Ntreated mice, control saline-treated mice frequently had additional histological changes including alveolar oedema, alveolar fibrin and inflammatory cells within alveoli. Of the mice that survived up to the study end-point, three had focal areas of fibrosis, type II pneumocyte hyperplasia and occasionally lymphoid hyperplasia.

However, most of the mice that survived showed little to no pathological signs in the lungs Fig. Histological lesions in the brain included multifocal perivascular cuffs of inflammatory cells, reactive glial cells, neutrophils and lymphocytes in the adjacent neuroparenchyma gliosis , infiltration of the meninges with inflammatory cells, and neuronal necrosis characterized by shrunken neuron bodies with hypereosinophilic cytoplasm and pyknotic or karyorrhectic nuclei.

No lesions were observed in the brains of mice that survived to the study end-point Fig. Although samples obtained at different time points are not directly comparable, the amounts of antigen and viral titres were lower in mice treated with N, particularly in those that survived to the study end-point Extended Data Fig.

No infectious virus was detected in the lung of Ntreated mice at the study end-point, or in the two saline-treated mice that survived to the end-point. This demonstrates the effectiveness of N in blocking SARS-CoV-2 infection and improving disease outcomes and survival using a short, early treatment regimen.

b , Probability of survival. c , Weight change of saline control mice. d , Weight change of Ntreated mice.

Two-tailed unpaired t -test was used to determine significance. Plaque assays were performed twice per sample from each mouse and the average was used to determine the PFU per g. Two-tailed Mann—Whitney test was used to determine significance.

Statistical analysis was not performed as samples are from different time points. logarithmic scale precludes negative values being shown.

Next, we further investigated the treatment window of N as well as the pan-variant effectiveness against SARS-CoV-2 B. Weight was monitored for six days after infection.

N showed significant protection against infection-associated weight loss Fig. Similarly, total pathology scores of lung tissue assessed using IHC sections were improved by approximately 1. Together, the in vivo data strongly suggest that N considerably prevents morbidity and mortality and reduces viral burden in the KhACE2 mouse model of severe SARS-CoV-2 infection, when used as a prophylactic or therapeutic treatment.

Two-tailed Mann Whitney test was used to determine significance. In this study, we report on N—a potent small-molecule protease inhibitor of human TMPRSS2 and a SARS-CoV-2 pan-variant HDA that is effective in vivo against the B.

N acts as an inhibitor of the TTSP-dependent proteolytic activation of virus spike protein, a critical step in permitting viral—cell membrane fusion and entry into target cells 4. The nanomolar potency of N against SARS-CoV-2 infection in human Calu-3 cells and patient-derived colonoids without detectable toxicity yields a selectivity index of greater than 10 6.

These data suggest that N may provide an effective early treatment option against emerging SARS-CoV-2 VOCs. We have previously shown how peptidomimetic-based compounds with ketobenzothiazole warheads exhibit potent antiviral efficacy in impeding the infection of Calu-3 cells with influenza A H1N1 virus, through inhibition of TTSPs The activation of the influenza A virus surface glycoprotein hemagglutinin is notably similar to that of the SARS-CoV-2 spike protein, in that both are viral surface protein homotrimers cleaved by proteolytic enzymes of the TTSP family that are expressed by host epithelial cells 14 , TTSPs are attractive broad-spectrum, HDA drug targets because of i their central role in mediating viral entry 5 ; ii their accessibility on the surface of nasal and pulmonary epithelial cells 41 , 42 ; and iii their demonstrated therapeutic potential for combating viruses such as SARS-CoV-2 and other human coronaviruses, as well as influenza viruses 14 , 40 , When we screened selected TMPRSS2 inhibitors for antiviral activity against SARS-CoV-2, a similar inhibitory profile was observed against TMPRSS2 expressed in Vero E6 cells compared to SARS-CoV-2 infection in Calu-3 cells.

N, the lead antiviral candidate, showed potent inhibition of SARS-CoV-2 infection in Calu-3 cells, with an EC 50 of 2. The potency of N was validated using two viral biomarkers of intracellular infection as well as by measuring the release of infectious viral particles. This complements a recent report that showed that peptidomimetic compounds targeting TMPRSS2 have high potency against SARS-CoVinduced cytopathic effects, as well as excellent stability and safety in mice The usefulness of N needs to be considered in the context of circulating SARS-CoV-2 variants.

VOCs such as B. In , B. We hypothesized that the efficacy of N against four SARS-CoV-2 VOCs B. Our results confirmed the low nanomolar pan-variant antiviral activity of N against these four SARS-CoV-2 VOCs in human cells.

Previous work has shown that the KhACE2 mouse model used in our studies is an ideal model for recapitulating the pathology of severe COVID in humans as well as its high morbidity and mortality. SARS-CoV-2 challenge in this model leads to high viral titres in lung and brain tissues with commensurate high morbidity and mortality, weight loss and cytokine and chemokine production 36 , Therefore, this model is ideal for testing SARS-CoV-2 therapeutic agents, owing to its severe disease burden as compared to other animal models including mouse-attenuated SARS-CoV-2 in wild-type mice or wild-type SARS-CoV-2 in golden Syrian hamsters, which exhibit milder symptoms.

Protection in an animal model with high levels of hACE2, such as the KhACE2 mouse model, is thus indicative of the high promise of anti-SARS-CoV-2 antivirals The mouse TMPRSS2 protein contains amino acids and shares Intranasal administration has several advantages for the prevention and treatment of SARS-CoV-2 and other viral diseases, including ease of self-administration.

SARS-CoV-2 mainly enters the human body through ACE2- and TMPRSS2-positive nasal epithelial cells 47 , 48 , Intranasal drug delivery maximizes airway and lung exposure while limiting systemic exposure.

For example, intranasal administration of a membrane fusion inhibitory lipopeptide prevented the transmission of SARS-CoV-2 in ferrets 50 ; however, the efficacy of intranasal delivery of a small molecule inhibitor has not to our knowledge been shown.

Under our conditions, intranasal administration of N markedly reduced morbidity and mortality in the KhACE2 mouse model of severe COVID pathology. This is indicative of the effective reduction of virus propagation by N in this animal model.

Although further studies are needed to understand the ideal time points for N administration, we have shown that N may also contribute therapeutic efficacy against SARS-CoV-2 VOCs. Antiviral candidates for SARS-CoV-2 infection are under investigation in clinical trials and in animal models, but at present, only one study on the DAA GC has reported protection against lethal SARS-CoV-2 infection in the KhACE2 model Plitidepsin, a naturally occurring HDA, protected against lung pathology in the KhACE2 model; however, the effect on mortality was not reported Cm and nafamostat mesylate are also HDAs that target serine proteases including host TTSPs and these are also undergoing human trials against SARS-CoV-2; however, no significant protection against infection was observed in the adenovirus hACE2 model Cm of SARS-CoV-2 infection 4 , Clinical trial data for the treatment of hospitalized patients with COVID with Cm showed that Cm had no effect on the time to recovery and incidence of death after SARS-CoV-2 infection Antivirals will probably need to be administered during the very early phase of COVID to be effective in lowering the risk of disease progression, consistent with our short early treatment regimen in KhACE2 mice infected with SARS-CoV Overall, we have developed and characterized N, a highly potent inhibitor of TMPRSS2-like proteases that blocks SARS-CoV-2 VOCs B.

In addition, we have shown that N provides an effective early treatment option against SARS-CoV-2 and the B. Moreover, N analogues may have broader applications in combating other widespread respiratory viruses that usurp TMPRSS2-related proteases for viral entry, including other established coronaviruses, influenza viruses and additional viruses that depend on TTSPs for entering host cells 4 , 30 , We envision a practical use of N for unvaccinated individuals or those with high risk of exposure or severe disease outcome related to SARS-CoV-2 VOCs and future emerging pathogens.

Practically, TTSP inhibitors should be administered as soon as possible after exposure to SARS-CoV-2 for maximal effect and may possibly act synergistically when used in multi-drug combinations with replication inhibitors such as remdesivir, paxlovid and molnupiravir to reduce the risk of antiviral resistance mutations.

Calu-3 cells 52 ATCC HTB were cultured according to ATCC recommendations. All experiments were performed in these cells below passage Cell density was kept between 0. Cm was obtained from MilliporeSigma. The SARS-CoV-2 nucleocapsid antibody HL GTX was provided by Genetex; mouse anti-dsRNA antibody J was purchased from Scions English and Scientific Consulting 33 ; Hoechst and secondary antibodies goat anti-mouse IgG Alexa Fluor A and goat anti-rabbit IgG Alexa Fluor A were obtained from Invitrogen.

Cell lines were screened for mycoplasma contamination using MycoAlert Mycoplasma Detection Kit Lonza. Preparation of the compounds using a mixed approach of solution and solid-phase synthesis is described in the Supplementary Information, in addition to a synthetic scheme of analogues, nuclear magnetic resonance NMR , high-resolution mass spectrometry HRMS , ultraperformance liquid chromatography—mass spectrometry UPLC—MS retention time, structure, purity, and molecular formula strings of compounds.

Amino acids and coupling reagents were obtained from Chem-Impex International and used as received. All other reagents and solvents were purchased from Sigma-Aldrich or Thermo Fisher Scientific. Tetrahydrofuran THF was dried over sodium benzophenone ketyl; dichloromethane over P 2 O 5 ; methanol over magnesium.

Celite AW Standard Super-Cel NF was obtained from Sigma-Aldrich. Chlorotrityl chloride CTC resin was obtained from Matrix Innovation and generally used with a loading of 1. Purity was analysed on a Waters UPLC H-Class with UV detection PDA equipped with an Acquity UPLC CSH C18 1. The final model was refined and minimized using the AmberExtended Huckel Theory EHT force field.

After drawing the structure, all protein—ligand complexes were prepared using the Protonate 3D tool; then the partial charges were calculated, and the ligands were energy-minimized. Molecules were docked in the protein-binding site with the software MOE All atoms were fixed, and the ligands were allowed to be flexible.

The carbon of the ketone making the reversible covalent bond with the protein was fixed at 3. The guanidine of the arginine in P1 was also fixed through two key interactions in the binding site. Conformational search using LowModeMD was made with AMBEREHT as a molecular mechanics force field with default parameters rejection limit: ; RMS gradient: 0.

Finally, a second round of energy minimization was performed around the ligand-binding site. The low energy conformations of the inhibitor-protein complexes were analysed for their binding interactions.

Vero E6 cells were transfected with mock pcDNA3. For the mouse TMPRSS2 assay, empty vector pCMV6-Entry, Origene PS and TMPRSS2-Myc-DDK Origene MR were used. IC 50 values were determined after generating a nonlinear regression analysis from a log [Compound] versus a proteolytic activity plot using GraphPad Prism software v.

of at least three independent experiments. All infections were carried out in a Biosafety Level 3 BSL3 facility UBC FINDER in accordance with the Public Health Agency of Canada and UBC FINDER regulations UBC BSL3 Permit B to F.

SARS-CoV-2 VOCs B. SARS-CoV-2 VOCs were first isolated in Vero-TMPRSS2 cells passage 1 and then passaged in Vero E6 cells passage 2. Viral stocks used in the experiments passage 3 were propagated in Vero E6 cells For experiments, passage three of the virus was used with a determined viral titre of 1.

Calu-3 cells were seeded at a concentration of 10, cells per well in well plates the day before infection. SARS-CoV-2 stocks were diluted in cell-specific medium to a multiplicity of infection MOI of 2. The fixative was removed, and cells were washed with PBS, permeabilized with 0.

Three-dimensional 3D volume rendering was done with LAS-X. Two-dimensional images were exported into tiff format.

Monitoring of the total number of cells based on nuclei staining and the number of virus-infected cells based on dsRNA and nucleocapsid staining was performed using the CellInsight CX7 HCS platform Thermo Fisher Scientific , as previously described 55 , Finally, the software HCS Studio Cell Analysis Software, v.

The fluorescence measured within each cell circle is then added and quantified for each well. Nine fields were sampled from each well. Nuclei stain Hoechst was also used to quantify cell loss owing to cytotoxicity or loss of adherence and to verify that the changes in viral infection did not result from a decrease in cell numbers.

Viral infection was detected by staining for dsRNA or nucleocapsid signal and quantified as described above. EC 50 experiments were repeated at least three times for each compound with three technical replicates in each experiment.

Intracellular nucleocapsid levels were interpolated to negative control 0. Calu-3 and Vero E6 cells or 10, cells for samples, 80—20, cells for standard curve were seeded in well plates. Cellular viability was assessed using Cell Titer-Glo 2. The number of viable cells was extrapolated using the standard curve.

Recombinant human matriptase, hepsin and DESC1 were expressed and purified as described previously 57 , Recombinant human furin, human cathepsin L Bio-Techne , and human thrombin MilliporeSigma were obtained from commercial sources. K i values were determined using steady-state velocities as previously reported 27 , To measure proteolytic activity, protease 0.

The preferred model was used for K i determination. Intestinal biopsy-derived colonoids from healthy donors were obtained from the Johns Hopkins Conte Digestive Disease Basic and Translational Research Core Center NIH NIDDK PDK and grown as described previously In brief, human colonoid monolayers were generated by combining the colonoids from one Matrigel dome around or more colonoids in a µl dome.

Cells were fed every two days and were used for experiments after they were fully confluent four to five days.

RNA quality and the presence of contaminating genomic DNA were verified as described previously RNA integrity was assessed with an Agilent Bioanalyzer Agilent Technologies. Reverse transcription was performed on 1. RNA was isolated and reverse-transcribed using Quanta Biosciences qScript cDNA SuperMix.

Relative expression levels were calculated according to the qBASE framework 60 with YWHAZ, PUM1 and MRPL19 as housekeeping genes for normalization. Primer design and validation were evaluated as described elsewhere In every qPCR run, a no-template control was performed for each primer pair; these were consistently negative.

All qPCR assays were performed by the RNomics Platform of the Université de Sherbrooke. Animal studies were performed in accordance with the recommendations in the Guide for the Care and Use of Laboratory Animals of the National Institutes of Health. All protocols were performed under approved BSL-3 conditions and approved by the Institutional Animal Care and Use Committee at Cornell University IACUC mouse protocol and BSL3 IBC MUA Intranasal virus and antiviral treatments were performed under anaesthesia, and all efforts were made to minimize animal suffering.

Since inhibitors such as Ruxolitinib can be administered in vivo , they may also prove useful in studies designed to investigate the importance of the IFN response in controlling virus infections in animal models. We thank Dan Young for expert technical assistance and Zoe Gage for proof reading the manuscript.

Conceived and designed the experiments: CSA RER CES. Performed the experiments: CSA CSE. Analyzed the data: CSA CES RER. Wrote the paper: CSA RER. Browse Subject Areas? Click through the PLOS taxonomy to find articles in your field.

Article Authors Metrics Comments Media Coverage Reader Comments Figures. Abstract Virus replication efficiency is influenced by two conflicting factors, kinetics of the cellular interferon IFN response and induction of an antiviral state versus speed of virus replication and virus-induced inhibition of the IFN response.

Materials and Methods Inhibitors, viruses and cells Inhibitors of the IFN response BX, MRT, MRT, TPCA-1, Cyt, AZD, Ruxolitinib, Tofacitinib were prepared as 10 mM stocks in dimethyl sulfoxide DMSO and used at the indicated concentrations. Virus plaque assays and growth kinetics Standard plaque assays were conducted in the appropriate cells using a 0.

Results and Discussion Eight small molecules that have previously been described to inhibit the cellular IFN response were obtained; four inhibitors that target components of the IFN induction pathway: TBK1 inhibitors BX, MRT, MRT [23] , [24] and the IKK-2 inhibitor TPCA-1 [25] , plus four inhibitors that target JAK1 a component of the IFN signaling pathway: Cyt, AZD, Ruxolitinib and Tofacitinib [26] — [29].

Download: PPT. Figure 1. Verification of IFN inhibitors ability to block IFN induction or IFN signaling. Figure 2. Effect of a panel of IFN inhibitors on BUNΔNSs virus growth in A cells.

Figure 3. Effect of a combination of different IFN inhibitors on BUNΔNSs growth in A and Vero cells. Figure 4.

Effect of Ruxolitinib RUX on BUNΔNSs and BUN-WT wildtype plaque formation in cell-lines derived from different mammalian species. Figure 5. Effect of Ruxolitinib RUX on plaque formation of a selection of viruses.

Figure 6. Inhibitory activity of Ruxolitinib and TPCA-1 is stable over time in cell culture. Acknowledgments We thank Dan Young for expert technical assistance and Zoe Gage for proof reading the manuscript.

Author Contributions Conceived and designed the experiments: CSA RER CES. References 1. Randall RE, Goodbourn S Interferons and viruses: an interplay between induction, signalling, antiviral responses and virus countermeasures.

J Gen Virol 1— View Article Google Scholar 2. Talon J, Salvatore M, O'Neill RE, Nakaya Y, Zheng H, et al. Proc Natl Acad Sci U S A — View Article Google Scholar 3. Steel J, Lowen AC, Pena L, Angel M, Solorzano A, et al.

J Virol — View Article Google Scholar 4. Mossler C, Groiss F, Wolzt M, Wolschek M, Seipelt J, et al. Vaccine — View Article Google Scholar 5. Bird BH, Albarino CG, Hartman AL, Erickson BR, Ksiazek TG, et al. View Article Google Scholar 6.

Brennan B, Welch SR, McLees A, Elliott RM Creation of a recombinant Rift Valley fever virus with a two-segmented genome. View Article Google Scholar 7.

Teng MN, Collins PL Altered growth characteristics of recombinant respiratory syncytial viruses which do not produce NS2 protein. View Article Google Scholar 8. Luongo C, Winter CC, Collins PL, Buchholz UJ Respiratory syncytial virus modified by deletions of the NS2 gene and amino acid S of the L polymerase protein is a temperature-sensitive, live-attenuated vaccine candidate that is phenotypically stable at physiological temperature.

View Article Google Scholar 9. Stojdl DF, Lichty BD, tenOever BR, Paterson JM, Power AT, et al. Cancer Cell 4: — View Article Google Scholar Naik S, Russell SJ Engineering oncolytic viruses to exploit tumor specific defects in innate immune signaling pathways.

Expert Opin Biol Ther 9: — Young DF, Andrejeva L, Livingstone A, Goodbourn S, Lamb RA, et al. Desmyter J, Melnick JL, Rawls WE Defectiveness of interferon production and of rubella virus interference in a line of African green monkey kidney cells Vero.

J Virol 2: — Mosca JD, Pitha PM Transcriptional and posttranscriptional regulation of exogenous human beta interferon gene in simian cells defective in interferon synthesis. Mol Cell Biol 6: — Hilton L, Moganeradj K, Zhang G, Chen YH, Randall RE, et al.

House C, House JA Evaluation of techniques to demonstrate foot-and-mouth disease virus in bovine tongue epithelium: comparison of the sensitivity of cattle, mice, primary cell cultures, cryopreserved cell cultures and established cell lines.

Vet Microbiol 99— Chen S, Short JA, Young DF, Killip MJ, Schneider M, et al. Virology — Killip MJ, Young DF, Gatherer D, Ross CS, Short JA, et al. Bridgen A, Weber F, Fazakerley JK, Elliott RM Bunyamwera bunyavirus nonstructural protein NSs is a nonessential gene product that contributes to viral pathogenesis.

Teng MN, Whitehead SS, Bermingham A, St Claire M, Elkins WR, et al. Garcia-Sastre A, Egorov A, Matassov D, Brandt S, Levy DE, et al. He B, Paterson RG, Stock N, Durbin JE, Durbin RK, et al. Virology 15— Matrosovich M, Matrosovich T, Garten W, Klenk HD New low-viscosity overlay medium for viral plaque assays.

Virol J 3: Clark K, Plater L, Peggie M, Cohen P Use of the pharmacological inhibitor BX to study the regulation and physiological roles of TBK1 and IkappaB kinase epsilon: a distinct upstream kinase mediates Ser phosphorylation and activation.

J Biol Chem — Clark K, Peggie M, Plater L, Sorcek RJ, Young ER, et al. Biochem J 93— Podolin PL, Callahan JF, Bolognese BJ, Li YH, Carlson K, et al. J Pharmacol Exp Ther — Published : 25 August Issue Date : September Anyone you share the following link with will be able to read this content:.

Sorry, a shareable link is not currently available for this article. Provided by the Springer Nature SharedIt content-sharing initiative. Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Skip to main content Thank you for visiting nature. Download PDF. Subjects DNA nanostructures Drug development Nanostructures. Full size image. References Tirado, S. Article CAS Google Scholar Bale, J. Article CAS Google Scholar Sigl, C. Article CAS Google Scholar Twarock, R.

Article Google Scholar Douglas, S. Article CAS Google Scholar Gerling, T. Article CAS Google Scholar Kwon, P. Article CAS Google Scholar Praetorius, F.

Article CAS Google Scholar Ponnuswamy, N. Article CAS Google Scholar Download references. Author information Authors and Affiliations Nick Holonyak Jr. View author publications. Ethics declarations Competing interests The authors declare no competing interests.

Rights and permissions Reprints and permissions. About this article. Cite this article Chauhan, N. Copy to clipboard. This article is cited by Designer DNA nanostructures for viral inhibition Shaokang Ren Keith Fraser Xing Wang Nature Protocols Publish with us Submission Guidelines For Reviewers Language editing services Submit manuscript.

Search Search articles by subject, keyword or author. Show results from All journals This journal. Advanced search. Close banner Close. Email address Sign up.

I agree my information will be processed in accordance with the Nature and Springer Nature Limited Privacy Policy.

Thank you for visiting nature. You are using Anti-cancer mind-body practices browser version silutions limited support solutiosn CSS. Fermented foods vs obtain the best experience, we Viirus-inhibiting Anti-cancer mind-body practices use Anti-cancer mind-body practices more up to date Electrolyte Solution or turn off compatibility mode Anti-cancer mind-body practices Internet Explorer. In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript. The COVID pandemic caused by the SARS-CoV-2 virus remains a global public health crisis. Although widespread vaccination campaigns are underway, their efficacy is reduced owing to emerging variants of concern 12. Development of host-directed therapeutics and prophylactics could limit such resistance and offer urgently needed protection against variants of concern 34.

Virus-inhibiting solutions -

a Domain structure of the SARS-CoV-2 Spike comprises two subunits, S1 and S2. S1 consists of the NTD, RBD domains and the RBM within the RBD. S2 subunit consists of FP; HR1 and HR2, TM, and the cytoplasmic tail CT. b The interaction of Spike and the ACE2 receptor is defined by binding of S1 RBD and ectodomain motifs of ACE2.

In August , convalescent plasma CP was approved by the FDA for emergency use in COVID patients. CP consists primarily of neutralizing antibodies from individuals who have recently recovered from SARS-CoV-2 infection.

Therefore, these antibodies have the potential to help block entry of the virus into the cells and to facilitate viral clearance.

CP has been used in several past outbreaks of other pathogens, and is generally understood to prevent infection and shorten duration and severity of the illness [ 4 , 57 , 58 ]. However, in the case of COVID, evidence of clinical benefit derived from CP has thus far been inconsistent, due to a lack of well controlled studies and the challenges associated with CP such as heterogeneity of plasma, lack of standardized protocols in preparing the antibody titers and how best to administer this plasma [ 59 , 60 , 61 ].

In contrast, synthetic antibodies, including monoclonal antibodies Mab can overcome some of these limitations as they are more specific, homogenous and scalable in terms of production. These types of antibodies can be generated from convalescent plasma, transgenic mice, B cell isolation or phage display libraries [ 15 , 62 , 63 ].

Since COVID emerged, the field of Mabs has experienced an explosion of discoveries. Evidence shows that the majority of Mabs neutralize the virus by binding to epitopes in the RBD and preventing its interaction with ACE2.

These findings in turn have been supported by studies that show neutralization of infection of pseudotyped and live SARS-CoV-2 in vitro , as well as therapeutic protection of rodents and primates from virus-induced lung injury [ 15 , 55 , 56 , 57 , 58 ].

Prominent examples of antibodies that have been characterized in this way include CCL Some of these will progress to clinical trials soon, and several more are already being evaluated for therapeutic benefit in clinical trials including CT-P59, VIR, AZD, TY, SCTA01, and SAB [ 15 , 62 , 63 , 64 , 66 ].

Currently, neutralizing monoclonal antibodies by Regeneron casirivimab and imdevimab or REGEN-COV and Eli Lilly bamlanivimab and etesevimab have already been granted emergency use authorization EUA. Approval for REGEN-COV was obtained in November , and the Eli Lilly combination was recently authorized in February [ 67 , 68 ].

Clinically, these antibody regimens have demonstrated capacity to reduce viral load and hospital visits and are currently prescribed for treatment of mild to moderate COVID in patients who are at risk for progressing to severe disease [ 67 , 68 , 69 ].

As their clinical efficacy continues to be monitored, the ongoing antigenic drift that poses ongoing challenges to vaccine efficacy also threatens to limit the efficacy of antibodies.

A number of studies have reported findings that the new variants, particularly those that contain the EK mutation such as the B. This is particularly true when the antibodies are used as monotherapies [ 72 , 73 , 74 ]. Indeed, the US government has now warned against use of bamlanivimab alone, which was initially approved as a monotherapy, and now recommends bamlanivimab use together with etesevimab [ 75 ].

The individual antibodies in the two EUA cocktails recognize distinct epitopes and their combinatorial use limits the development of escape mutants and resistance.

New data has shown that the bamlanivimab and etesevimab combination has relatively higher neutralization efficacy against variants compared to either antibody alone, whilst REGEN-COV has largely maintained its potency against all the variants tested so far [ 69 , 76 , 77 ].

These observations validate the use of cocktails and emphasize the importance of designing antibodies from more conserved epitopes to counter neutralization escape mutations as well as the need to create broad-spectrum antibodies and other therapies for future variants and outbreaks.

Fortunately, the development of biologics with a wide neutralization breadth is already a growing area of research. Rappazzo et al. have shown that antibodies engineered using directed evolution can be broadly active. Specifically, one of their affinity matured variants, ADG-2, which recognizes a highly conserved epitope exhibited potent neutralization against authentic SARS-CoV-2 in vitro , and protected mice infected with SARS-CoV and SARS-CoV-2 against viral replication and lung pathology.

More importantly, when compared to EUA antibodies that neutralized mostly SARS-CoV-2, ADG-2 displayed a wider breadth against clade 1 sarbecoviruses including SARS-CoV, SARS-CoV-2, WIVI, LYRa11, Rs, GD-Pangolin and Pangolin-GX-P2V [ 78 ]. Another study by Wec et al. has also identified several antibodies from a convalescent Covid patient that cross-neutralized SARS-CoV, SARS-CoV-2 and WIVI [ 79 ].

More recently, two studies have reported similar discoveries. Starr et al. discovered antibodies that target conserved, functionally constrained RBD residues.

One of these, S2H97, showed high affinity and neutralization breadth across SARS-CoVrelated sarbecoviruses [ 80 ]. An accompanying study showed that S2X, which binds to a highly conserved cryptic RBD epitope, cross-neutralized all the VOCs and a wide spectrum of human and zoonotic sarbecoviruses.

Notably, prophylactic dosing of Syrian hamsters with S2X offered protection against a SARS-CoV-2 and B. Additional antibodies that have demonstrated similar efficacy against variants are summarized in Table 1 [ 82 , 83 , 84 ].

However, it is not only antibodies that are demonstrating success in dealing with current or potential escape mutants. Nanobodies are also proving to be a viable option. Nanobodies are single domain antibodies that are generated from immunized llamas, camels and phage displays [ 85 , 86 , 87 , 88 ].

Recent published evidence shows that multivalent nanobodies are capable of both neutralizing circulating variants and preventing emergence of resistant escape mutants via binding to multiple, non-overlapping epitopes, avidity effects and binding to conserved epitopes largely inaccessible to normal antibodies [ 89 , 90 , 91 , 92 , 93 , 94 , 95 , 96 ].

Table 1 summarizes the main findings from these studies. Additionally, nanobodies have properties that may be beneficial considering the potential use of monoclonal antibodies as pre-and post-exposure prophylactics PEPrs. Pre-clinically, monoclonal antibodies have prophylactic value in addition to therapeutic value.

Widespread evidence of prophylactic protection against SARS-CoVrelated respiratory injury in animal models ranging from mice to hamsters to rhesus macaques has been reported [ 97 , 98 , 99 , , ].

Consistently, preliminary evidence from ongoing clinical trials with EUA monoclonal antibody therapies is also very promising [ , , ]. However, it is important to point out that widespread outpatient use of potential PEPrs therapies would be most practical with agents that can be conveniently administered.

Antibodies are molecularly large, less stable, complex and costly to produce. Currently, antibodies are usually given intravenously in healthcare facilities that must also be equipped with resources for dealing with potential infusion reactions.

Nanobodies, on the other hand are smaller, cheaper to make and can be nebulized for easier and more convenient pulmonary delivery using inhalers or nasal sprays [ 63 , 85 ].

Collectively, these facts make monoclonal antibody cocktails, broad-spectrum antibodies and multivalent nanobodies the future in terms of dealing with variants during early onset of disease and prevention of infection pre- and post-exposure.

The use of protein-based antivirals has been dominated by antibodies or their functional fragments that bind to the RBD of S1.

An alternative to this strategy is to target the ectodomain of ACE2, as it serves as the SARS-CoV-2 receptor. These decoy receptors Fig. Moreover, although escape mutants can sometimes outmaneuver antibody defenses with RBD- or NTD-specific mutations, it is more difficult to escape decoys without also losing virulence, since decoy receptors have the same binding interface as does the endogenous ACE2.

Furthermore, soluble ACE2 has already been found to be safe as shown in clinical studies focused on treatment of ARDS and SARS [ 15 ]. It is expected, therefore, that soluble ACE2 receptors will likely be safe and potentially effective against SARS-CoV-2 infection.

An earlier pre-clinical study by Monteil et al. using clinical grade soluble recombinant human ACE2 hrACE2 confirmed this potential, and showed that hrACE2 prevented infection by SARS-CoV-2 significantly [ ]. A number of ongoing clinical trials are currently evaluating the potential of soluble rACE2 [ 15 ].

For example, rhACE2 APN01 is now in Phase II clinical trials. Phase I data showed that APN01 can reduce viremia and viral titers, and preliminary evidence from phase II data indicates that APN01 lowers risk of medical complications and shortens recovery time [ , ].

These exciting findings have inspired other groups to engineer even more potent forms of soluble ACE2 using computational design, deep mutagenesis and affinity maturation.

A study by Chan et al. shows that soluble ACE2 designed using affinity maturation based on the mutations of the residues involved in the binding of S led to the discovery of sACE Two other studies by Glasgow and Linsky et al.

have employed a similar approach with success [ , ]. In particular, two decoys engineered by Linsky et al. More importantly, other findings have shown that even smaller versions of decoy receptors can yield potent neutralization effects [ ].

Hyper-stable miniprotein binders that include AHB1, AHB2, LCB1 and LCB3 have displayed impressive in vitro inhibition of SARS-CoV-2 infection with potencies in the nano- to picomolar range [ ].

LCB1, only 56 residues, has been utilized as the lead binder in follow up studies to evaluate in vivo efficacy when administered either intraperitoneally LCB1-F c or intranasally LCB1v1. LCB1 administration using both routes protected the mice post-exposure against SARS-CoVmediated lung disease as well as pre-exposure, even when dosed intranasally as many as five days before virus inoculation.

Notably, LCB1v1. Taken together, these protein-based antivirals hold clinical promise and point to a remarkable therapeutic and prophylactic potential now, as well as potential protection against re-emerging ACE2-utilizing coronaviruses in the future.

Summary of strategies of targeting viral entry at the surface membrane. Four approaches are highlighted including antibody-based inhibitors that consists of monoclonal antibodies and nanobodies.

Receptor decoys consists of WT soluble ACE2 or versions of ACE2 that are engineered to have high affinity than WT ACE2. Various inhibitors mainly based on HR2 of S2 have also been designed to prevent fusion of the S2 with cellular membrane.

Also, peptides and small molecules that are designed to interfere with the S1 RBD and ACE2 interaction have also been made. Peptides represent another type of inhibitor that can be directed against Spike and ACE2 to prevent viral entry. Peptides are smaller, simpler and cheaper to make than are antibodies or the other protein-based antivirals.

Their well-known liability is generally low bioavailability due to degradation and metabolism when given systemically [ ]. However, as a COVID therapeutic, this disadvantage can easily be overcome through nebulization or dry aerosol powders for direct delivery to the lungs [ ].

In general, we can divide SARS-CoV-2 Spike-targeted peptide inhibitors into two groups: those that perturb S1 RBD: ACE2 binding, and those that interfere with fusion of S2 with the membrane Fig. Previous studies by groups such as the Huang and Cho labs had shown that peptides extracted from important S1 RBD-recognizing motifs in ACE2 see Fig.

For example, the Cho group showed that linking together two non-contiguous segments that are close in space can inhibit SARS-CoV infection with a half-maximal inhibition concentration of nM [ ]. Other studies also reported similar findings with S1-derived linear peptides [ , ].

Given the similarity in the binding conformation between S1 RBD of SARS-CoV and SARS-CoV-2 with ACE2 and the high sequence identity of the S1 RBD of SARS-CoV and SARS-CoV-2, there is reason to believe that peptides against ACE2: S1 RBD binding in SARS-CoV-2 can also be effective [ 62 ].

Findings by Karoyan et al. appear to corroborate this expectation. Their data show that peptide fragments P8, P9, P10 from the α-helix of the ACE2 peptidase domain PD that are rationally modified with residues that have a propensity for helical folding show high binding affinity and antiviral activity against authentic SARS-CoV-2 in the nanomolar range [ ].

A study by Curreli et al. also showed that peptides from a similar region of ACE2 that are structurally stabilized with double stapling show inhibitory activity against pseudotyped and live SARS-CoV-2 in the low micromolar range [ ].

For peptides that are based on the binding motif of S1 RBD, particularly the RBM as shown in Fig. On the other hand, some studies report more modest activity or complete lack of activity of peptide inhibitors.

For example, the Zhang lab published that even though a mer peptide from the α-helix of PD of ACE2 exhibited high binding affinity in the nanomolar range, it lacked appreciable competitive capability against soluble ACE2 for binding S1 RBD [ , ].

Certainly, an argument can be made that the lack of binding here may be due to limited secondary structure in solution of the linear native peptide designed by Zhang et al.

Nonetheless, a different group has shown that even with stapling that dramatically improved helicity of their peptides, no appreciable binding activity was observed for either stabilized and non-stabilized peptides [ ].

In our lab we have found that peptides rationally designed from the binding motifs of either ACE2 or S1 RBD display modest inhibitory activity in the low micromolar range unpublished.

These inconsistencies therefore warrant more data for safer conclusions to be reached regarding the activity of peptides that inhibit S1 RBD: ACE2 interaction and their prospects as COVID therapeutics. As noted above, viral fusion with the cellular membrane also represents a point of potential therapeutic targeting.

Since both HR1 and HR2 are needed to come together to form the 6HB and then to fuse, designing a peptide mimicking one region will competitively interfere with formation of the fusion core [ 54 , 55 ]. This approach has been utilized to prevent entry of other viruses with heptad regions such as HIV.

In fact, enfuvirtide is a fusion inhibitor that is approved for treating HIV infection [ ]. HR2 is usually used as template to make HR1-directed peptides, and this approach has been successfully applied for coronaviruses [ ]. Much of this work was published before the inception of SARS-CoV-2, and targeted viruses such as SARS-CoV, MERS-CoV and HCoVE [ , ].

Perhaps the most impressive results were obtained from OCHR2P, as reported in [ ]. OCHR2P peptide was derived from the HR2 domain of HCoV-OC43, and showed broad spectrum activity against alpha- and beta-coronaviruses. An optimized version of OCHR2P from this study EK1 was quickly tested once SARS-CoV-2 emerged, and showed potent activity against SARS-CoV-2 infection in vitro.

A lipid-conjugated form of EK1 called EK1C4 with an IC 50 of 37 nM against SARS-CoV-2 infection in vitro has also been tested in mice.

In the mouse study, EK1C4 displayed not only a good in vivo safety profile, but also antiviral activity and metabolic stability following intranasal administration [ , ].

The extension of activity from previous hCoV strains such as SARS-CoV stems from the high conservation of the HR regions. For instance, HR1 and HR2 of SARS-CoV and SARS-CoV-2 have The conservation allows for broad spectrum activity against hCoVs. Other HR2-derived peptides have also been identified and tested against SARS-CoV.

IPB and IPB have shown low nanomolar activity against infection with SARS-CoV and SARS-CoV-2 pseudovirions [ ]. Another lipid-modified fusion peptide called SARS HRC -PEG 4 2-chol inhibited SARS-CoV-2 with a half maximal inhibitory concentration of 3.

The pan-specific activity and in vivo protection of animals show that fusion inhibitors have potential for clinical utility. Altogether, peptides are a promising therapeutic option for COVID in the future, though more research is needed.

Small molecules therapeutics generally are better situated to overcome problems such as cell permeability and metabolic lability than are peptides, but their development also takes time. Thus far, efforts to develop small molecule therapeutics for COVID have largely involved repurposing antiviral drugs already approved for clinical use, or which have undergone regulatory processes tied to clinical trials.

The drug remdesivir, previously clinically studied for Ebola, was identified in this manner. Additional antiviral drugs for RNA viruses targeting the RdRP, helicase and protease proteins are undergoing further clinical evaluation for efficacy against COVID [ 48 , ].

The same approach can be adopted for viral entry inhibitors. Unfortunately, the literature shows that most small molecule inhibitors that were previously evaluated as entry antagonists have no regulatory approval. In addition, the reported pre-clinical potency is largely in the low micromolar range, implying that most of these candidates will first have to be tested in the context of SARS-CoV-2 and then be optimized for affinity and potency [ , , ].

Examples of inhibitors that target S1 RBD and ACE2 and their interactions in the context of ACE2-utilizing coronaviruses include cepharantine, VE, SSAA09E2, emodin, HTCC and HM-HTCC [ , , ].

Drug reprofiling studies in our lab that evaluated candidates targeting the ACE2: S1 RBD interaction showed that of those tested, cepharantine was the most promising candidate, with single digit micromolar potency against SARS-CoV-2 RBD binding to ACE2 unpublished. Indeed, several findings in recent publications have validated these observations and demonstrated that cepharantine does display anti-viral activity against both pseudotyped and authentic SARS-CoV-2 infection in vitro, with potencies ranging from 0.

Additionally, some candidate small molecule inhibitors with novel activity against coronaviruses have also been identified. Hanson et al. discovered coriligan through a high content screen that inhibited the RBD and ACE2 interaction with an IC 50 of 5. In a study by Day et al. In vitro studies using live SARS-CoV-2 showed that the hit compounds suramin and evans blue possessed antiviral activity with acceptable selectivity and IC 50 values of 46 and 28 μM, respectively [ ].

Overall, compared to the other studies discussed above, targeting ACE2 and S1 RBD interaction with small molecules remains a developing area of research. The reported antiviral potencies thus far are modest, indicating the need for significant additional optimization to support their development into efficacious agents.

The strategy of using small molecules and other agents to prevent viral entry through the cell surface membrane is summarized in Fig. Although S1 and S2 mediate viral attachment and membrane fusion to enable the virus to unload its genetic cargo, function of these two subunits is enabled by the participation of at least 3 types of host proteases: furins, cathepsins and surface serine proteases.

Viral entry generally occurs either through direct fusion of the virus with the surface membrane or endocytic uptake, and what determines which proteases will dominate in facilitating fusogenic activity is the entry pathway utilized [ 54 , 55 , , ].

Conversely, if the virus takes the endocytic route, cathepsins will play a more dominant role [ 54 , 55 , , ]. Cathepsin L is a lysosomal cysteine protease and its function, like that of many other cathepsins, is pH-dependent, with optimal pH activity ranging from 3—6.

Without cleavage of Spike by these proteases, the virus would not be able to fuse with the lysosomal or autolysosomal membrane to release its genome into the cytoplasm Fig.

Therefore, all the three different classes of proteases noted above represent rational targets for COVID therapeutic intervention. Furin inhibitors have previously been reported as possible targets in the context of other viruses such as influenza, and may also be relevant for SARS-CoV In SARS-CoV-2 studies, dec-RVKR-cmk inhibited infection in vitro with an IC 50 of 5 μM [ ].

MI, another furin inhibitor has also been found to reduce SARS-CoV-2 titers in Calu-3 cells by almost fold at 10 µM [ ]. For TMPRSS2, various inhibitors, both peptidomimetics and small molecules, have been reported for previous coronavirus strains such as MERS and SARS-CoV [ , , ].

The peptidomimetic inhibitors that have shown promising activity against SARS-CoV-2 include aprotinin, MI and MI Aprotinin has been tested previously in the clinic for combating influenza infection, and has also shown significant inhibition of SARS-CoV-2 growth at 10 μM [ , ].

MI and MI have both shown higher potency compared to aprotinin under similar experimental conditions and are thus more promising. More importantly, the combination of MI plus MI was viable and more effective than either therapy alone [ ].

Equally promising are the small molecule inhibitors of TMPRSS2, camostat and nafamostat mesylate. Camostat and nafamostat mesylate are analogues with clinical approval for pancreatitis and disseminated intravascular coagulation [ ]. Indeed, camostat was one of the early small molecule inhibitors to be shown to have significant activity in blocking the entry of SARS-CoV-2 into cells [ 56 ].

However, nafamostat is actually the more potent analogue, and has been shown to inhibit SARS-CoV-2 replication in Calu-3 cells with an EC 50 of 10 nM [ , ].

Both inhibitors are currently in clinical trials for evaluation as COVID therapeutics, and results regarding their efficacy are eagerly awaited [ 56 , ]. A number of cathepsin inhibitors against coronaviruses have also been reported in various studies.

Amongst them are teicoplanin, K, SSAA09E1, SID and P9 derivatives [ 64 , , , , ]. Teicoplanin has exhibited good activity against SARS-CoV-2 pseudovirions entry with an IC 50 of 1. The same can be said for SID and P9 derivatives.

A study by Ou et al. The P9 derivates, P9R and 8P9R, have also shown significant activity against SARS-CoV and SARS-CoV-2 ranging in the low micro- to nanomolar range [ , ]. More importantly, 8P9R demonstrated antiviral activity by decreasing the SARS-CoV-2 viral load in vivo in mice and hamsters [ ].

The inhibitors mentioned above, such as SID, inhibit the protease activity of cathepsins in a direct way by interacting with the enzyme active site through mimicking of the endogenous substrate. However, indirect inhibition of protease activity through pH modulation is also an option. Endosome acidification inhibitors act through this mechanism, and a number were highly touted as potential effective treatments at the beginning of the pandemic [ ].

Such inhibitors, which include chloroquine, hydroxychloroquine and azithromycin, function by elevating the pH of the endosome, shifting the pH outside the optimal range and thereby indirectly suppressing cathepsin protease activity [ , , ]. Despite this rational and promising pre-clinical activity, these inhibitors have not demonstrated evidence of consistent and robust benefit when evaluated in various clinical trials [ , , , , , ].

Given some of the known side effects of chloroquine derivatives, such as cardiac-related toxicities and retinopathy, their consideration for clinical use has now been put on hold [ ]. Despite these recommendations against endosome acidification inhibitors, the other protease inhibitors remain potential candidates for clinical development given their specificity.

Future studies will reveal and determine their utility as future COVID therapeutics. COVID is now understood as a biphasic illness, with an early viral phase and a more dangerous host-immune response phase.

This knowledge has shaped our translational and clinical therapeutic strategies to find treatments for those infected. The ongoing antigenic drift of SARS-CoV-2 is also shaping the fight against COVID Four major variant strains have now been identified, which have generally shown increased transmissibility and resistance to the efficacy of vaccines and monoclonal antibodies [ 36 , 37 ].

Vaccines, particularly those that are mRNA-based, have shown that they offer some protection against the variants, albeit with reduced effectiveness, and multiple doses of the vaccines, including booster shots, may be necessary in the future [ , , , ]. Health officials will also continue to monitor variants of interest that have already been identified.

In addition, the emergence of SARS-CoV-2 has also renewed fears that another zoonotic spillover will occur and cause an even more deadly outbreak. These fears are not unfounded, given that we experienced more than 10 serious outbreaks from emerging RNA viruses in the last 20 years alone [ ].

Each of these aspects have subjected the counter-measures currently in place to increased attention, asking how such measures can be made more effective based on available evidence. In addition, and also of critical importance, is the development of future plans for dealing with mutant strains and potential outbreaks.

In this review, we have highlighted the utility of vaccines and the gaps they leave in fighting COVID We then demonstrated that the mechanism of action of entry inhibitors makes them suitable agents for early management of COVID to help cover some of the gaps, and shown why continued research on such inhibitors is crucial.

The monoclonal antibody entities are farthest along the drug development pipeline, with some already approved for use EUA and several more in advanced stages of clinical trials [ 66 ].

Cocktails of monoclonal antibodies, multivalent nanobodies and recombinant soluble ACE2 have also demonstrated therapeutic effect against mutant strains, including those currently in circulation, as well as broad, cross-family coronavirus efficacy.

For agents based on recombinant ACE2, similar efficacy comes from their similarity with the endogenous receptor, which makes it difficult for mutant strains to arise without also losing infectivity.

In addition to these valuable therapeutic effects and their potential as agents to treat future outbreaks, these protein-based antivirals have also demonstrated they can be useful when given prophylactically, even several days before exposure.

As noted earlier, various subgroups of people will benefit from prophylactic treatment using these agents. As for the miniprotein, peptide and small molecule therapeutics, current literature suggests that they are not as advanced in terms of clinical development as are the antibodies or recombinant ACE2.

However, their utility is in their size and ability to be more readily developed into therapeutic formulations that can be self-administered either as oral pills or inhalants. More research is therefore still needed, as researchers and decision makers continue to evaluate the potential use of entry inhibitors for outpatient prophylaxis.

Also, given that the nasal passage is the most dominant and initial site of infection, aerosolization can potentially be beneficial in preventing viral spread to the lungs through use of nasal sprays [ 8 ].

Finally, more investment in the development of entry inhibitor therapeutics as well as other antivirals and therapies directed against the host immune response is needed, as their availability will impact our options in responding not only to future SARS-CoV-2 lineages, but also to future coronavirus pandemics.

Recent events have made it abundantly clear that it is both more impactful and cost effective to prevent or prepare for a pandemic like the one caused by COVID, than to encounter such a pandemic without preparation.

For this reason, current proposals by the US and international community to invest more into pro-active and pre-emptive countermeasures against future outbreaks are commendable, as this development will shorten the time between an outbreak and an effective therapeutic response [ , ].

Nicola M, Alsafi Z, Sohrabi C, Kerwan A, Al-Jabir A, Iosifidis C, Agha M, Agha R. The socio-economic implications of the coronavirus pandemic COVID : a review. Int J Surg. Article PubMed PubMed Central Google Scholar. Cucinotta D, Vanelli M.

WHO declares COVID a pandemic. Acta Biomed. PubMed PubMed Central Google Scholar. Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, transmission, diagnosis, and treatment of coronavirus disease COVID : a review. Article CAS PubMed Google Scholar. Izda V, Jeffries MA, Sawalha AH.

COVID A review of therapeutic strategies and vaccine candidates. Clin Immunol. Sun P, Lu X, Xu C, Sun W, Pan B. Understanding of COVID based on current evidence. J Med Virol. Hu B, Guo H, Zhou P, Shi ZL. Characteristics of SARS-CoV-2 and COVID Nat Rev Microbiol.

Harrison AG, Lin T, Wang P. Mechanisms of SARS-CoV-2 transmission and pathogenesis. Trends Immunol. Article CAS PubMed PubMed Central Google Scholar. Hou YJ, Okuda K, Edwards CE, Martinez DR, Asakura T, Dinnon KH 3rd, Kato T, Lee RE, Yount BL, Mascenik TM, et al.

SARS-CoV-2 reverse genetics reveals a variable infection gradient in the respiratory tract. Mason RJ. Pathogenesis of COVID from a cell biology perspective.

Eur Respir J. Sungnak W, Huang N, Becavin C, Berg M, Queen R, Litvinukova M, Talavera-Lopez C, Maatz H, Reichart D, Sampaziotis F, et al. SARS-CoV-2 entry factors are highly expressed in nasal epithelial cells together with innate immune genes.

Nat Med. Stratton CW, Tang YW, Lu H. Pathogenesis-directed therapy of novel coronavirus disease. Sette A, Crotty S. Adaptive immunity to SARS-CoV-2 and COVID Le Bert N, Clapham HE, Tan AT, Chia WN, Tham CYL, Lim JM, Kunasegaran K, Tan LWL, Dutertre CA, Shankar N et al.

Highly functional virus-specific cellular immune response in asymptomatic SARS-CoV-2 infection. J Exp Med. Wang J, Jiang M, Chen X, Montaner LJ. Cytokine storm and leukocyte changes in mild versus severe SARS-CoV-2 infection: review of COVID patients in China and emerging pathogenesis and therapy concepts.

J Leukoc Biol. Twomey JD, Luo S, Dean AQ, Bozza WP, Nalli A, Zhang B. COVID update: the race to therapeutic development. Drug Resist Updat. Schulte-Schrepping J, Reusch N, Paclik D, Bassler K, Schlickeiser S, Zhang B, Kramer B, Krammer T, Brumhard S, Bonaguro L, et al.

Severe COVID Is marked by a dysregulated myeloid cell compartment. Sundararaj Stanleyraj J, Sethuraman N, Gupta R, Thiruvoth S, Gupta M, Ryo A. Treating COVID are we missing out the window of opportunity?

J Antimicrob Chemother. Article PubMed CAS Google Scholar. Covid Treatment guidelines. Accessed July 7th, Kim PS, Read SW, Fauci AS. Therapy for early COVID a critical need. Cohen MS. Monoclonal antibodies to disrupt progression of early Covid infection. N Engl J Med. Meo SA, Bukhari IA, Akram J, Meo AS, Klonoff DC.

Eur Rev Med Pharmacol Sci. CAS PubMed Google Scholar. COVID vaccines. In: Drugs and Lactation Database LactMed. Bethesda MD ; Poland GA, Ovsyannikova IG, Kennedy RB. SARS-CoV-2 immunity: review and applications to phase 3 vaccine candidates. Rosenbaum L. Escaping Catch - overcoming covid vaccine hesitancy.

Fontanet A, Cauchemez S. COVID herd immunity: where are we? Nat Rev Immunol. Article PubMed CAS PubMed Central Google Scholar.

Kwok KO, Lai F, Wei WI, Wong SYS, Tang JWT. Herd immunity - estimating the level required to halt the COVID epidemics in affected countries.

J Infect. Alaran AJ, Adebisi YA, Badmos A, Khalid-Salako F, Gaya SK, Ilesanmi EB, Olaoye DQ, Bamisaiye A, Lucero-Prisno DE. Uneven power dynamics must be levelled in COVID vaccines access and distribution. Public Health Pract Oxf.

Lucero-Prisno DE, Ogunkola IO, Imo UF, Adebisi YA. Who Will Pay for the COVID Vaccines for Africa? Am J Trop Med Hyg. COVAX: Working for global equitable access to covid vaccines.

Sonani B, Aslam F, Goyal A, Patel J, Bansal P. COVID vaccination in immunocompromised patients. Clin Rheumatol. Article PubMed Google Scholar.

Coustasse A, Kimble C, Maxik K. COVID and vaccine hesitancy: a challenge the United States must overcome. J Ambul Care Manage. Group C. A future vaccination campaign against COVID at risk of vaccine hesitancy and politicisation.

Lancet Infect Dis. Article Google Scholar. Altmann DM, Boyton RJ, Beale R. Immunity to SARS-CoV-2 variants of concern. Abdool Karim SS, de Oliveira T: New SARS-CoV-2 Variants - Clinical, Public Health, and Vaccine Implications.

SARS-CoV-2 Variant Classifications and Definitions. Tracking SARS-CoV-2 variants. Khan A, Zia T, Suleman M, Khan T, Ali SS, Abbasi AA, Mohammad A, Wei DQ. J Cell Physiol. Gomez CE, Perdiguero B, Esteban M. Vaccines Basel. Zhou D, Dejnirattisai W, Supasa P, Liu C, Mentzer AJ, Ginn HM, Zhao Y, Duyvesteyn HME, Tuekprakhon A, Nutalai R et al.

Evidence of escape of SARS-CoV-2 variant B. Madhi SA, Baillie V, Cutland CL, Voysey M, Koen AL, Fairlie L, Padayachee SD, Dheda K, Barnabas SL, Bhorat QE et al.

Efficacy of the ChAdOx1 nCoV Covid Vaccine against the B. Emary KRW, Golubchik T, Aley PK, Ariani CV, Angus B, Bibi S, Blane B, Bonsall D, Cicconi P, Charlton S et al. Rubin R. COVID vaccines vs variants-determining how much immunity is enough.

Davis C, Logan N, Tyson G, Orton R, Harvey W, Haughney J, Perkins J, The COVID genomics UK COG-UK consortium, peacock TP, Barclay WS et al. Reduced neutralisation of the Delta B. medRxiv Sheikh A, McMenamin J, Taylor B, Robertson C, Public Health Scotland and the EAVE II Collaborators. SARS-CoV-2 Delta VOC in Scotland: demographics, risk of hospital admission, and vaccine effectiveness.

World Health Organization WHO. COVID weekly epidemiological update, 1 June Geneva: WHO; Increased transmissibility and global spread of SARS-CoV-2 variants of concern as at June Singh TU, Parida S, Lingaraju MC, Kesavan M, Kumar D, Singh RK. Drug repurposing approach to fight COVID Pharmacol Rep.

Pawar AY. Combating devastating COVID by drug repurposing. Int J Antimicrob Agents. Consortium WHOST, Pan H, Peto R, Henao-Restrepo AM, Preziosi MP, Sathiyamoorthy V, Abdool Karim Q, Alejandria MM, Hernandez Garcia C, Kieny MP et al.

Repurposed Antiviral Drugs for Covid - Interim WHO solidarity trial results. Del Rio C, Collins LF, Malani P. Long-term Health Consequences of COVID Wrapp D, Wang N, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, Graham BS, McLellan JS.

Cryo-EM structure of the nCoV spike in the prefusion conformation. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, function, and antigenicity of the SARS-CoV-2 Spike glycoprotein. Structure, function, and evolution of coronavirus spike proteins.

Annu Rev Virol. Huang Y, Yang C, Xu XF, Xu W, Liu SW. Structural and functional properties of SARS-CoV-2 spike protein: potential antivirus drug development for COVID Acta Pharmacol Sin. Article PubMed PubMed Central CAS Google Scholar.

Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor.

Martinez MA. Compounds with Therapeutic Potential against Novel Respiratory Coronavirus. Antimicrob Agents Chemother. Willis VC, Arriaga Y, Weeraratne D, Reyes F, Jackson GP. A narrative review of emerging therapeutics for COVID Mayo Clin Proc Innov Qual Outcomes.

Peng HT, Rhind SG, Beckett A. Convalescent Plasma for the Prevention and Treatment of COVID A systematic review and quantitative analysis.

JMIR Public Health Surveill. Wooding DJ, Bach H. Treatment of COVID with convalescent plasma: lessons from past coronavirus outbreaks. Clin Microbiol Infect. Wang Y, Huo P, Dai R, Lv X, Yuan S, Zhang Y, Guo Y, Li R, Yu Q, Zhu K. Convalescent plasma may be a possible treatment for COVID A systematic review.

Int Immunopharmacol. Renn A, Fu Y, Hu X, Hall MD, Simeonov A. Fruitful neutralizing antibody pipeline brings hope to defeat SARS-Cov Trends Pharmacol Sci. Jiang S, Zhang X, Yang Y, Hotez PJ, Du L. Neutralizing antibodies for the treatment of COVID Nat Biomed Eng.

Seyedpour S, Khodaei B, Loghman AH, Seyedpour N, Kisomi MF, Balibegloo M, Nezamabadi SS, Gholami B, Saghazadeh A, Rezaei N. Targeted therapy strategies against SARS-CoV-2 cell entry mechanisms: a systematic review of in vitro and in vivo studies.

Xiaojie S, Yu L, Lei Y, Guang Y, Min Q. Neutralizing antibodies targeting SARS-CoV-2 spike protein. Stem Cell Res. Case JB, Winkler ES, Errico JM, Diamond MS. On the road to ending the COVID pandemic: are we there yet? Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, Musser BJ, Soo Y, Rofail D, Im J, et al.

REGN-COV2, a neutralizing antibody cocktail, in outpatients with Covid Gottlieb RL, Nirula A, Chen P, Boscia J, Heller B, Morris J, Huhn G, Cardona J, Mocherla B, Stosor V, et al. Effect of bamlanivimab as monotherapy or in combination with etesevimab on viral load in patients with mild to moderate COVID a randomized clinical trial.

Chen RE, Zhang X, Case JB, Winkler ES, Liu Y, VanBlargan LA, Liu J, Errico JM, Xie X, Suryadevara N et al. Resistance of SARS-CoV-2 variants to neutralization by monoclonal and serum-derived polyclonal antibodies.

Kuzmina A, Khalaila Y, Voloshin O, Keren-Naus A, Boehm-Cohen L, Raviv Y, Shemer-Avni Y, Rosenberg E, Taube R. SARS-CoV-2 spike variants exhibit differential infectivity and neutralization resistance to convalescent or post-vaccination sera.

Cell Host Microbe. Wang P, Nair MS, Liu L, Iketani S, Luo Y, Guo Y, Wang M, Yu J, Zhang B, Kwong PD et al. Increased Resistance of SARS-CoV-2 Variants B. Weisblum Y, Schmidt F, Zhang F, DaSilva J, Poston D, Lorenzi JC, Muecksch F, Rutkowska M, Hoffmann HH, Michailidis E et al.

Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. Hoffmann M, Hofmann-Winkler H, Krüger N, Kempf A, Nehlmeier I, Graichen L, Sidarovich A, Moldenhauer AS, Winkler MS, Schulz S et al. SARS-CoV-2 variant B. bioRxiv, Accessed July 7 th , Accessed July 7thth, Tada T, Zhou H, Dcostaa BM, Samanovicb MI, Mulliganb MJ, Landaua NR.

The Spike Proteins of SARS-CoV-2 B. Rappazzo CG, Tse LV, Kaku CI, Wrapp D, Sakharkar M, Huang D, Deveau LM, Yockachonis TJ, Herbert AS, Battles MB, et al. Broad and potent activity against SARS-like viruses by an engineered human monoclonal antibody.

Wec AZ, Wrapp D, Herbert AS, Maurer DP, Haslwanter D, Sakharkar M, Jangra RK, Dieterle ME, Lilov A, Huang D, et al. Broad neutralization of SARS-related viruses by human monoclonal antibodies.

Starr TN, Czudnochowski N, Zatta F, Park YJ, Liu Z, Addetia A, Pinto D, Beltramello M, Hernandez P, Greaney AJ et al. Antibodies to the SARS-CoV-2 receptor-binding domain that maximize breadth and resistance to viral escape. Tortorici MA, Czudnochowski N, Starr TN, Marzi R, Walls AC, Zatta F, Bowen JE, Jaconi S, Iulio JD, Wang Z et al.

Structural basis for broad sarbecovirus neutralization by a human monoclonal antibody. Baum A, Fulton BO, Wloga E, Copin R, Pascal KE, Russo V, Giordano S, Lanza K, Negron N, Ni M, et al.

Antibody cocktail to SARS-CoV-2 spike protein prevents rapid mutational escape seen with individual antibodies. Pinto D, Park YJ, Beltramello M, Walls AC, Tortorici MA, Bianchi S, Jaconi S, Culap K, Zatta F, De Marco A, et al.

Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody. Liu H, Yuan M, Huang D, Bangaru S, Zhao F, Lee CD, Peng L, Barman S, Zhu X, Nemazee D et al.

A combination of cross-neutralizing antibodies synergizes to prevent SARS-CoV-2 and SARS-CoV pseudovirus infection. Sasisekharan R. P reparing for the Future - Nanobodies for Covid? Konwarh R: Nanobodies Prospects of expanding the gamut of neutralizing antibodies against the novel coronavirus, SARS-CoV Front Immunol.

Huo J, Le Bas A, Ruza RR, Duyvesteyn HME, Mikolajek H, Malinauskas T, Tan TK, Rijal P, Dumoux M, Ward PN, et al. Neutralizing nanobodies bind SARS-CoV-2 spike RBD and block interaction with ACE2.

Nat Struct Mol Biol. Chi X, Liu X, Wang C, Zhang X, Li X, Hou J, Ren L, Jin Q, Wang J, Yang W. Humanized single domain antibodies neutralize SARS-CoV-2 by targeting the spike receptor binding domain.

Nat Commun. Koenig PA, Das H, Liu H, Kummerer BM, Gohr FN, Jenster LM, Schiffelers LDJ, Tesfamariam YM, Uchima M, Wuerth JD et al. Structure-guided multivalent nanobodies block SARS-CoV-2 infection and suppress mutational escape. Xiang Y, Nambulli S, Xiao Z, Liu H, Sang Z, Duprex WP, Schneidman-Duhovny D, Zhang C, Shi Y.

Versatile and multivalent nanobodies efficiently neutralize SARS-CoV CAS PubMed PubMed Central Google Scholar. Xu J, Xu K, Jung S, Conte A, Lieberman J, Muecksch F, Cetrulo Lorenzi JC, Park S, Wang Z, Tessarollo L et al. Multimeric nanobodies from camelid engineered mice and llamas potently neutralize SARS-CoV-2 variants.

Schoof M, Faust B, Saunders RA, Sangwan S, Rezelj V, Hoppe N, Boone M, Billesbolle CB, Puchades C, Azumaya CM, et al. An ultrapotent synthetic nanobody neutralizes SARS-CoV-2 by stabilizing inactive Spike. Bracken CJ, Lim SA, Solomon P, Rettko NJ, Nguyen DP, Zha BS, Schaefer K, Byrnes JR, Zhou J, Lui I, et al.

Bi-paratopic and multivalent VH domains block ACE2 binding and neutralize SARS-CoV Nat Chem Biol. Sun D, Sang Z, Kim YJ, Xiang Y, Cohen T, Belford AK, Huet A, Conway JF, Sun J, Taylor DJ et al.

Potent neutralizing nanobodies resist convergent circulating variants of SARS-CoV-2 by targeting novel and conserved epitopes. Mast FD, Fridy PC, Ketaren NE, Wang J, Jacobs EY, Olivier JP, Sanyal T, Molloy KR, Schmidt F, Rutkowska M et al. Nanobody repertoires for exposing vulnerabilities of SARS-CoV Pymm P, Adair A, Chan LJ, Cooney JP, Mordant FL, Allison CC, Lopez E, Haycroft ER, O'Neill MT, Tan LL et al.

Nanobody cocktails potently neutralize SARS-CoV-2 DG NY variant and protect mice. LW, RL, YG, YL, XD, RX, YZ, and FY drafted the manuscript.

TY, SJ, and FY revised and edited the manuscript. The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Albulescu, I. Suramin inhibits Zika virus replication by interfering with virus attachment and release of infectious particles. Antiviral Res. doi: PubMed Abstract CrossRef Full Text Google Scholar. Arbuckle, J. mBio 8:e CrossRef Full Text Google Scholar. Baell, J. New substructure filters for removal of pan assay interference compounds PAINS from screening libraries and for their exclusion in bioassays.

Barr, K. Working with Zika and Usutu Viruses in vitro. PLoS Negl. Barros, J. Acute Zika virus infection in an endemic area shows modest proinflammatory systemic immunoactivation and cytokine-symptom associations.

Bavari, S. Lipid raft microdomains: a gateway for compartmentalized trafficking of Ebola and Marburg viruses. Bernatchez, J. Development and validation of a phenotypic high-content imaging assay for assessing the antiviral activity of small-molecule inhibitors targeting Zika virus.

Agents Chemother. Brugger, B. The HIV lipidome: a raft with an unusual composition. Bullard-Feibelman, K. The FDA-approved drug sofosbuvir inhibits Zika virus infection. Byrd, C. A novel inhibitor of dengue virus replication that targets the capsid protein.

Cai, L. Viral polymerase inhibitors T and T are potential inhibitors of Zika virus replication. Calland, N. Hepatology 55, — Carneiro, B. The green tea molecule EGCG inhibits Zika virus entry. Virology , — Chazal, N. Virus entry, assembly, budding, and membrane rafts.

Chen, L. Antiviral activity of peptide inhibitors derived from the protein E stem against Japanese encephalitis and Zika viruses.

Chen, Y. Dengue virus infectivity depends on envelope protein binding to target cell heparan sulfate. Cheng, F. Suppression of Zika virus infection and replication in endothelial cells and astrocytes by PKA inhibitor PKI J Virol e Cho, N.

ACS Chem. Coronado, M. Cruz-Oliveira, C. Receptors and routes of dengue virus entry into the host cells. FEMS Microbiol. Dai, L. Structures of the Zika Virus envelope protein and its complex with a flavivirus broadly protective antibody. Cell Host. Microbe 19, — De Clercq, E. Approved antiviral drugs over the Past 50 Years.

de la Cruz, L. Binding of low molecular weight inhibitors promotes large conformational changes in the dengue virus NS2B-NS3 protease: fold analysis by pseudocontact shifts.

Di Croce, L. Transcriptional regulation by Polycomb group proteins. Dick, G. Zika virus. Isolations and serological specificity.

Duan, D. Colloidal aggregation and the in vitro activity of traditional Chinese medicines. Ekins, S. Open drug discovery for the Zika virus. FRes Erbel, P.

Structural basis for the activation of flaviviral NS3 proteases from dengue and West Nile virus. Estoppey, D. The natural product cavinafungin selectively interferes with Zika and Dengue virus replication by inhibition of the host signal peptidase. Cell Rep. Eyer, L. Nucleoside inhibitors of Zika virus.

Fernando, S. An approach for Zika virus inhibition using homology structure of the envelope protein. Fonseca, B. Structure-activity analysis of niclosamide reveals potential role for cytoplasmic pH in control of mammalian target of rapamycin complex 1 mTORC1 signaling.

Fu, B. ZMPSTE24 defends against influenza and other pathogenic viruses. Germi, R. Heparan sulfate-mediated binding of infectious dengue virus type 2 and yellow fever virus. Hamel, R. Biology of Zika virus infection in human skin cells. Han, Y. Investigational drugs for the treatment of Zika virus infection: a preclinical and clinical update.

Expert Opin. Drugs 27, — Hasan, S. A human antibody against Zika virus crosslinks the E protein to prevent infection. Heinz, F.

The antigenic structure of Zika Virus and its relation to other flaviviruses: implications for infection and immunoprophylaxis. Isaacs, C. Epigallocatechin gallate inactivates clinical isolates of herpes simplex virus. Jackman, J. Therapeutic treatment of Zika virus infection using a brain-penetrating antiviral peptide.

Ji, R. TAM receptors support neural stem cell survival, proliferation and neuronal differentiation. PLoS One 9:e Jin, J.

The inhibitory effect of sodium baicalin on oseltamivir-resistant influenza A virus via reduction of neuraminidase activity. Jurgeit, A. Niclosamide is a proton carrier and targets acidic endosomes with broad antiviral effects.

PLoS Pathog. Kim, J. Linker histone H1. Kim, Y. NMR analysis of a novel enzymatically active unlinked dengue NS2B-NS3 protease complex.

Kirby, W. Clinical and laboratory studies of novobiocin, a new antibiotic. AMA Arch. Google Scholar. Kuivanen, S.

Obatoclax, saliphenylhalamide and gemcitabine inhibit Zika virus infection in vitro and differentially affect cellular signaling, transcription and metabolism. Kumar, A. Hydroxychloroquine inhibits Zika virus NS2B-NS3 protease. ACS Omega 3, — Lee, H.

Lei, J. Crystal structure of Zika virus NS2B-NS3 protease in complex with a boronate inhibitor. Science , — Lemke, G. Immunobiology of the TAM receptors. Li, C. Immunity 46, — Chloroquine, a FDA-approved Drug, Prevents Zika virus infection and its associated congenital microcephaly in mice.

EBioMedicine 24, — Li, Y. Structural dynamics of Zika virus NS2B-NS3 protease binding to dipeptide inhibitors. Structure Li, Z. Existing drugs as broad-spectrum and potent inhibitors for Zika virus by targeting NS2B-NS3 interaction.

Cell Res. Li, F. A scorpion venom peptide Ev37 restricts viral late entry by alkalizing acidic organelles. Structural insights into the inhibition of Zika virus NS2B-NS3 protease by a small-molecule inhibitor.

Erythrosin B is a potent and broad-spectrum orthosteric inhibitor of the flavivirus NS2B-NS3 protease. Lim, H. Inhibitory effect of flavonoids against NS2B-NS3 protease of ZIKA virus and their structure activity relationship.

Liu, T. Mechanism of thioesterase-catalyzed chain release in the biosynthesis of the polyether antibiotic nanchangmycin. Lorizate, M. Comparative lipidomics analysis of HIV-1 particles and their producer cell membrane in different cell lines.

Cell Microbiol. Lozach, P. Dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin DC-SIGN -mediated enhancement of dengue virus infection is independent of DC-SIGN internalization signals.

Lu, G. Margueron, R. The Polycomb complex PRC2 and its mark in life. Nature , — McGovern, S. A common mechanism underlying promiscuous inhibitors from virtual and high-throughput screening. Meertens, L. The TIM and TAM families of phosphatidylserine receptors mediate dengue virus entry.

Microbe 12, — Axl mediates ZIKA virus entry in human glial cells and modulates innate immune responses. Merino-Ramos, T. Antiviral activity of nordihydroguaiaretic acid and its derivative Tetra-O-methyl nordihydroguaiaretic acid against west nile virus and Zika virus. Miner, J. The TAM receptor Mertk protects against neuroinvasive viral infection by maintaining blood-brain barrier integrity.

Moghaddam, E. Baicalin, a metabolite of baicalein with antiviral activity against dengue virus. Mottin, M. The A-Z of Zika drug discovery. Drug Discov. Today 23, — Mounce, B.

Curcumin inhibits Zika and chikungunya virus infection by inhibiting cell binding. Musso, D. Zika Virus. Nance, C. Preclinical development of the green tea catechin, epigallocatechin gallate, as an HIV-1 therapy.

Allergy Clin. Navarro-Sanchez, E. Dendritic-cell-specific ICAM3-grabbing non-integrin is essential for the productive infection of human dendritic cells by mosquito-cell-derived dengue viruses. EMBO Rep. Nitsche, C. De novo discovery of nonstandard macrocyclic peptides as noncompetitive inhibitors of the Zika virus NS2B-NS3 protease.

ACS Med. Peptide-boronic acid inhibitors of flaviviral proteases: medicinal chemistry and structural biology. Nowakowski, T. Expression analysis highlights AXL as a Candidate Zika virus entry receptor in neural stem cells.

Cell Stem Cell 18, — Nyboe Andersen, A. Individualized versus conventional ovarian stimulation for in vitro fertilization: a multicenter, randomized, controlled, assessor-blinded, phase 3 noninferiority trial. Olsen, D. A 7-deaza-adenosine analog is a potent and selective inhibitor of hepatitis C virus replication with excellent pharmacokinetic properties.

Oo, A. Deciphering the potential of baicalin as an antiviral agent for Chikungunya virus infection. Baicalein and baicalin as Zika virus inhibitors. Pal, P. In silico screening of small molecule modulators of Zika virus proteins. Noida: IEEE. Passioura, T. Selection-based discovery of druglike macrocyclic peptides.

Perera-Lecoin, M. Flavivirus entry receptors: an update. Viruses 6, 69— Petersen, L. Phoo, W. Structure of the NS2B-NS3 protease from Zika virus after self-cleavage.

Structures of Zika virus NS2B-NS3 protease in complex with peptidomimetic inhibitors. Pierson, T. Flaviviruses: braking the entering. Pryke, K. A novel agonist of the TRIF pathway induces a cellular state refractory to replication of Zika, Chikungunya, and dengue viruses.

Puschnik, A. A small-molecule oligosaccharyltransferase inhibitor with pan-flaviviral activity. Ramharack, P.

Zika virus NS5 protein potential inhibitors: an enhanced in silico approach in drug discovery. Rausch, K. Screening bioactives reveals nanchangmycin as a broad spectrum antiviral active against Zika Virus.

Rocker, A. The molecular tweezer CLR01 inhibits Ebola and Zika virus infection. Rohini, K. Exploring the lead compounds for Zika Virus NS2B-NS3 protein: an e-pharmacophore-based approach. Roy, A. Solution conformations of Zika NS2B-NS3pro and its inhibition by natural products from edible plants.

PLoS One e Sahoo, M. Virtual screening for potential inhibitors of NS3 protein of Zika Virus. Genomics Inform. Shannon, A. Product release is rate-limiting for catalytic processing by the Dengue virus protease. Shi, Y.

Structural Biology of the Zika Virus. Trends Biochem. Shiryaev, S. Characterization of the Zika virus two-component NS2B-NS3 protease and structure-assisted identification of allosteric small-molecule antagonists. Singh, A. Discovery of potential Zika virus RNA polymerase inhibitors by docking-based virtual screening.

Song, J. NMR for the design of functional mimetics of protein-protein interactions: one key is in the building of bridges. Cell Biol.

PubMed Abstract Google Scholar. Antiviral effect of catechins in green tea on influenza virus. Spencer, J. Replication of Zika Virus in human prostate cells: a potential source of sexually transmitted virus.

Stephen, P. Structural insight into NS5 of Zika virus leading to the discovery of MTase inhibitors. Suroengrit, A. Halogenated chrysins inhibit dengue and Zika virus infectivity. Tong, X. Merimepodib, an IMPDH inhibitor, suppresses replication of Zika virus and other emerging viral pathogens. Tritsch, D.

Flavonoids: true or promiscuous inhibitors of enzyme? The case of deoxyxylulose phosphate reductoisomerase. Vanwalscappel, B. Toll-like receptor agonist R blocks Zika virus replication by inducing the antiviral protein viperin. Vasireddi, M.

A novel antiviral inhibits Zika virus infection while increasing intracellular glutathione biosynthesis in distinct cell culture models. Verma, S.

Identification of Potent, Selective, Cell-active inhibitors of the histone lysine methyltransferase EZH2. Wang, C. Nuclear import inhibitor N- 4-hydroxyphenyl retinamide targets Zika virus ZIKV nonstructural protein 5 to inhibit ZIKV infection.

Wang, S. Screening of FDA-approved drugs for inhibitors of Japanese encephalitis virus infection. Wang, X. Development of small-molecule viral inhibitors targeting various stages of the life cycle of emerging and re-emerging viruses.

Journal Anti-cancer mind-body practices Nanobiotechnology volume solutiknsArticle number: Cite this Virjs-inhibiting. Metrics details. Severe acute solutios syndrome coronavirus 2 SARS-CoV-2 solutione to COVID and Anti-cancer mind-body practices become a pandemic worldwide with mortality of millions. Nanotechnology can be used to deliver antiviral medicines or other types of viral reproduction-inhibiting medications. At various steps of viral infection, nanotechnology could suggest practical solutions for usage in the fight against viral infection. Nanotechnology-based approaches can help in the fight against SARS-CoV-2 infection.

Author: Gabei

0 thoughts on “Virus-inhibiting solutions

Leave a comment

Yours email will be published. Important fields a marked *

Design by ThemesDNA.com