SARS-CoV-2 Treatment: Current Perceptions and Perspectives

authors:

avatar Denis Andreevich Shoichet ORCID 1 , * , avatar Ivan Pavlovich Polovikov ORCID 1 , avatar Alena Sergeevna Sergeeva ORCID 1 , avatar Irina Dmitrievna Bulgakova ORCID 1 , 2

I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), Moscow, Russia
Federal State Budgetary Scientific Institution "Research Institute of Vaccines and Serums them. I.I. Mechnikov", Moscow, Russia

How To Cite? Andreevich Shoichet D, Pavlovich Polovikov I, Sergeevna Sergeeva A, Dmitrievna Bulgakova I. SARS-CoV-2 Treatment: Current Perceptions and Perspectives. J Microbiota. 2025;2(1):e157269. https://doi.org/10.5812/jmb-157269.

Abstract

Context:

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was first identified in Wuhan, China. From the outset, this highly contagious and dangerous illness posed significant risks and challenges for global healthcare systems and populations. Scientists and healthcare professionals worldwide have been striving to develop effective treatments for severe acute respiratory SARS-CoV-2 infection, employing both pre-existing and innovative approaches. However, a specific drug targeted explicitly against COVID-19 remains elusive. Despite the reduced prevalence of severe cases and the predominance of the SARS-CoV-2 Omicron variant, COVID-19 continues to pose significant risks, particularly for polymorbid patient groups. This review focuses on the potential antiviral effects of promising new drugs, including ensitrelvir, clazakizumab, upamostat, and others.

Evidence Acquisition:

An analytical review was conducted using the Scopus, PubMed, EMBASE, ScienceDirect, and Google Scholar databases for publications related to promising drugs against SARS-CoV-2, their mechanisms of action, and potential therapeutic effects up to November 2024. Selection criteria included: (1) Free-text availability; (2) English language; (3) relevance to the publication's theme a total of 104 articles were initially selected. Of these, 27 articles were excluded for not meeting the free-text criterion, 14 were excluded due to language incompatibility, and 28 were excluded for thematic irrelevance. Ultimately, data from 35 articles were analyzed and summarized for this review.

Results:

Thirty-five scientific studies were reviewed to describe current paradigms and emerging concepts in SARS-CoV-2 treatment. Current drugs with proven efficacy include: (1) Combined monoclonal antibodies targeting the S-protein (casirivimab/imdevimab); (2) monoclonal antibody preparations against the S-protein (pemivibart; vilobelimab); (3) viral replication inhibitors (molnupiravir); (4) protease inhibitors (nirmatrelvir/ritonavir); (5) immunosuppressants (dexamethasone; tocilizumab). The review also highlights drugs under development targeting traditional pathways [e.g., viral protease, interleukin-6 (IL-6)] and those aimed at novel mechanisms (e.g., antigalectin-3; adhesion blockers).

Conclusions:

Although numerous drug effects and potential drug combinations for COVID-19 have been described, limited clinical trials and research focus on identifying novel treatment approaches. Comprehensive investigations are needed to assess and evaluate the risks of different treatment strategies, minimizing potential short- and long-term complications for patients. Such advancements will pave the way for more optimal and effective medical treatment of COVID-19.

1. Context

In 2019, cases of severe acute respiratory syndrome were first reported in Wuhan, Hubei province, China. The illness spread rapidly worldwide, resulting in serious health consequences for the global population and healthcare systems (1). By November 2024, over 776 million people had been diagnosed with the coronavirus infection, commonly known as coronavirus disease 2019 (COVID-19), which is caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus.

1.1. Morphology of Virion

Syndrome coronavirus 2 is an enveloped virus with a positive ribonucleic acid (RNA) strand (+ssRNA) belonging to the Coronaviridae family of viruses (2). The virus contains four structural proteins (Figure 1): Spike (S) protein—a glycoprotein spike, envelope (E) protein, membrane (M) protein, and nucleocapsid (N) protein (3).

Morphology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)
Morphology of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)

The S-protein is presented as a spike on the surface of the viral particle and is responsible for binding the virus to angiotensin-converting enzyme 2 (ACE2) receptors (4). These receptors are abundant on epithelial cells of the respiratory tract, such as type 2 alveolocytes (5, 6). The ACE2 receptors are also found on cells in the upper esophagus, ileal enterocytes, myocardial cells, proximal tubule cells of the kidneys, and bladder cells (7), leading to a wide range of possible virus-organ interactions.

Among the primary clinical manifestations of COVID-19, particularly the Omicron variant, are fever, cough, shortness of breath, and dyspnea. Although SARS-CoV-2 can enter various organs, it predominantly affects the respiratory system and blood vessels. Other systems, including the gastrointestinal (GI) tract, cardiovascular, hepatobiliary, renal, and central nervous systems, may also be impacted, though less commonly (8). These effects may result from mechanisms such as viral toxicity, ischemic injury caused by vasculitis, thrombosis or thromboinflammation, impaired immune regulation, and dysregulation of the renin-angiotensin-aldosterone system (9).

As a result, SARS-CoV-2 can trigger a wide range of pathological processes. This raises critical concerns regarding effective antiviral and systemic treatment for patients diagnosed with coronavirus infection, as the extensive inflammatory and infectious process affects the entire organism, potentially leading to unfavorable outcomes or even death (10).

Currently, several commonly used treatment strategies exist for COVID-19. However, mutations, the emergence of novel strains, and the increasing resilience of the virus pose significant challenges. These developments reduce the effectiveness of standard treatment regimens, potentially rendering them inadequate (11, 12). This underscores the urgent need to develop new drugs and treatment approaches for SARS-CoV-2 to enhance the efficacy of medications and therapeutic plans. A summary of medicines in development is provided in Table 1.

Table 1.

List of Medicines in Development Against Syndrome Coronavirus 2

DrugsMechanism of Action
EnsitrelvirInhibitor of SARS-CoV-2 3CL protease
ClazakizumabMonoclonal antibody with a high affinity for human interleukin-6
UpamostatBlock the attachment of the virus to the cell surface
CK0802Suppress intense immune responses
Antigalectin-3Decrease in the secretion of cytokines IL-1, IL-6, and TNF-α by macrophages and dendritic cells
LufotrelvirInhibitor of SARS-CoV-2 3CL protease

2. Ensitrelvir

Ensitrelvir (Xocova), a newly synthesized oral inhibitor of the SARS-CoV-2 3CL protease (Figure 2), has been used to treat patients with mild to moderate COVID-19 or asymptomatic SARS-CoV-2 infection (13, 14). Clinical trials demonstrated that a 5-day course of ensitrelvir rapidly reduced viral titer and viral RNA levels between days 2 and 6 in the study population. Minor side effects included decreases in high density lipids (HDL) and triglyceride levels and increases in total bilirubin and iron levels. Importantly, these changes were asymptomatic and resolved without additional treatment.

Life cycle of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mechanisms of action of promising preparations against SARS-CoV-2
Life cycle of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and mechanisms of action of promising preparations against SARS-CoV-2

A potential limitation of the study is that participants had received at least one dose of a COVID-19 vaccine, making it challenging to assess the drug's standalone efficacy. However, this reflects current global realities, as a significant portion of the population has already been vaccinated against SARS-CoV-2 (15).

3. Clazakizumab

Clazakizumab is a genetically engineered humanized monoclonal antibody with a high affinity for human interleukin-6 (IL-6) (16). As a direct ligand inhibitor that does not bind to circulating soluble receptors, clazakizumab may outperform IL-6R inhibitors, potentially offering greater benefits to patients with severe disease complicated by hyperinflammation (Figure 2).

Clinical trial results suggest that adding clazakizumab to standard therapy improves the 28-day survival rate without mechanical ventilation and enhances overall survival at 28 and 60 days compared to a placebo. However, a potential drawback is the reported association between clazakizumab and hypoxemia, which has been observed as a side effect. This raises concerns about its safety, as the treatment may lose effectiveness if the patient develops severe hypoxemia, warranting further evaluation of its risk-benefit profile (17).

4. Upamostat

Current treatment strategies for COVID-19 primarily target the virus itself rather than host factors, which also play a role in viral spread and entry. For instance, transmembrane serine protease 2 (TMPRSS2) facilitates the cleavage of the SARS-CoV-2 spike protein (18, 19). Upamostat, a prodrug of WX-UK1, targets the activity of multiple serine proteases (20). By doing so, upamostat blocks viral attachment to the cell surface and inhibits viral uptake by host cells (Figure 2).

A recent outpatient study on upamostat in patients exhibiting two moderate or severe symptoms of COVID-19, but not requiring hospitalization, demonstrated that the drug is safe and well-tolerated. It showed high bioavailability when self-administered and was intracellularly converted to WX-UK1. The study reported no adverse effects on liver, kidney, or hematological parameters, and no bleeding was observed. These findings suggest a modest anticoagulant effect, which could be potentially beneficial for COVID-19 patients, as coagulopathy is one of the most severe complications of the disease (21).

5. CK0802

Regular T-cells have shown potential in the treatment of COVID-19 (22) (Figure 2), as they can mitigate excessive immune responses (23). A randomized, double-blind, placebo-controlled phase 1 RESOLVE clinical trial investigated the safety and early efficacy of CK0802, a cryopreserved allogeneic umbilical cord blood (UCB) Treg cell product, in patients with moderate to severe COVID-19 (24). The study confirmed the safety of this therapy in critically ill patients with acute respiratory distress syndrome (ARDS) associated with COVID-19; however, its efficacy remains uncertain, necessitating larger clinical trials.

This treatment approach holds promise due to its convenience. The use of cryopreserved cell therapy products allows for application in intensive care unit settings without requiring complex on-site recovery of cell products. The therapy involves the infusion of ready-to-use CryoStore CS50N bags, which can be thawed bedside, and the capability to store medication stocks on-site ensures immediate availability (25). Overall, this method appears highly advantageous and has the potential to revolutionize COVID-19 treatment paradigms.

6. Antigalectin-3

In COVID-19, the severity of the disease correlates with an increase in galectin-3 levels (26), a mammalian β-galactoside-binding lectin (27). Galectin-3 plays a role in regulating systemic and pulmonary pro-inflammatory cytokine profiles by promoting IL-18 production by macrophages, which activates NLRP3 inflammasomes (28) (Figure 2). The therapeutic effect of antigalectin-3 involves reducing the secretion of cytokines such as IL-1, IL-6, and tumor necrotic factor – alpha (TNF-α) by macrophages and dendritic cells (29), thereby mitigating inflammatory responses.

The investigational drug GB0139, an inhaled thiodigalactoside inhibitor of galectin-3, has demonstrated safety in COVID-19 patients. Clinical data have shown reductions in markers of inflammation, including C-reactive protein (CRP), neutrophil-to-lymphocyte ratio (NLR), and chemokine CXCL10, in patients treated with GB0139 compared to those receiving a placebo (30). These findings suggest that targeting galectin-3 could provide a promising anti-inflammatory strategy for managing COVID-19.

7. Lufotrelvir

PF-00835231 is a small-molecule protease inhibitor designed to target the 3CL protease of SARS-CoV-2, an essential enzyme responsible for viral replication, and is also implicated in the earlier SARS outbreak (31-33) (Figure 2). In a randomized, double-blind, placebo-controlled phase 1 clinical trial, single doses of its phosphate prodrug, lufotrelvir, administered via continuous infusion (50 mg to 700 mg over 24 hours), demonstrated safety (34).

Lufotrelvir has also shown potential as part of combination therapy, with a relatively low incidence of contraindications and side effects (35). A completed phase 1b study evaluating its safety and efficacy in hospitalized COVID-19 patients has produced promising results, paving the way for further exploration in future clinical trials.

8. Conclusions

The search for an optimal and most effective treatment strategy for COVID-19 is ongoing, with a primary focus on preventing and rapidly eliminating the virus from the body. Key criteria for such treatments include minimal side effects, efficacy across a range of patient severities (from mild to severe cases), and ease of access, storage, and administration. Efforts are also being made to test combination therapies with existing drugs to enhance their effects, accelerate action, and mitigate side effects.

While the efficacy of many new drugs against COVID-19 remains unproven in large-scale clinical trials, early research data provide a foundation for further testing. Additionally, the use of innovative drug delivery systems may play a role in simplifying treatment processes in the near future. In conclusion, the primary challenge lies in systematizing all available information to devise the most effective treatment strategy for managing this infection.

8.1. Limitations

Based on our analysis, it is important to note that the new treatment options are still undergoing pre-clinical and clinical research, and therefore, it cannot be stated with absolute certainty that they offer comprehensive treatment solutions.

Moreover, the development of these new treatment options is driven by the increasing emergence of therapy-resistant viral particles, which pose a significant new challenge to healthcare systems.

Additionally, there is growing evidence of adverse reactions associated with medications targeting cytokine storms, highlighting another critical factor that must be considered in the development of new treatments for COVID-19.

References

  • 1.

    Haileamlak A. The impact of COVID-19 on health and health systems. Ethiop J Health Sci. 2021;31(6):1073-4. [PubMed ID: 35392335]. [PubMed Central ID: PMC8968362]. https://doi.org/10.4314/ejhs.v31i6.1.

  • 2.

    V'Kovski P, Kratzel A, Steiner S, Stalder H, Thiel V. Coronavirus biology and replication: implications for SARS-CoV-2. Nat Rev Microbiol. 2021;19(3):155-70. [PubMed ID: 33116300]. [PubMed Central ID: PMC7592455]. https://doi.org/10.1038/s41579-020-00468-6.

  • 3.

    Kakavandi S, Zare I, VaezJalali M, Dadashi M, Azarian M, Akbari A, et al. Structural and non-structural proteins in SARS-CoV-2: potential aspects to COVID-19 treatment or prevention of progression of related diseases. Cell Commun Signal. 2023;21(1):110. [PubMed ID: 37189112]. [PubMed Central ID: PMC10183699]. https://doi.org/10.1186/s12964-023-01104-5.

  • 4.

    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-19. Acta Pharmacol Sin. 2020;41(9):1141-9. [PubMed ID: 32747721]. [PubMed Central ID: PMC7396720]. https://doi.org/10.1038/s41401-020-0485-4.

  • 5.

    Beyerstedt S, Casaro EB, Rangel EB. COVID-19: angiotensin-converting enzyme 2 (ACE2) expression and tissue susceptibility to SARS-CoV-2 infection. Eur J Clin Microbiol Infect Dis. 2021;40(5):905-19. [PubMed ID: 33389262]. [PubMed Central ID: PMC7778857]. https://doi.org/10.1007/s10096-020-04138-6.

  • 6.

    Bridges JP, Vladar EK, Huang H, Mason RJ. Respiratory epithelial cell responses to SARS-CoV-2 in COVID-19. Thorax. 2022;77(2):203-9. [PubMed ID: 34404754]. [PubMed Central ID: PMC9273148]. https://doi.org/10.1136/thoraxjnl-2021-217561.

  • 7.

    Cascella M, Rajnik M, Aleem A, Dulebohn SC, Di Napoli R. Features, Evaluation, and Treatment of Coronavirus (COVID-19). Treasure Island (FL); 2025.

  • 8.

    Zhang Y, Geng X, Tan Y, Li Q, Xu C, Xu J, et al. New understanding of the damage of SARS-CoV-2 infection outside the respiratory system. Biomed Pharmacother. 2020;127:110195. [PubMed ID: 32361161]. [PubMed Central ID: PMC7186209]. https://doi.org/10.1016/j.biopha.2020.110195.

  • 9.

    Bourgonje AR, Abdulle AE, Timens W, Hillebrands JL, Navis GJ, Gordijn SJ, et al. Angiotensin-converting enzyme 2 (ACE2), SARS-CoV-2 and the pathophysiology of coronavirus disease 2019 (COVID-19). J Pathol. 2020;251(3):228-48. [PubMed ID: 32418199]. [PubMed Central ID: PMC7276767]. https://doi.org/10.1002/path.5471.

  • 10.

    Wong RSY. Inflammation in COVID-19: from pathogenesis to treatment. Int J Clin Exp Pathol. 2021;14(7):831-44. [PubMed ID: 34367415]. [PubMed Central ID: PMC8339720].

  • 11.

    Su S, Wang Q, Jiang S. Facing the challenge of viral mutations in the age of pandemic: Developing highly potent, broad-spectrum, and safe COVID-19 vaccines and therapeutics. Clin Transl Med. 2021;11(1). e284. [PubMed ID: 33463059]. [PubMed Central ID: PMC7803365]. https://doi.org/10.1002/ctm2.284.

  • 12.

    Ridgway H, Chasapis CT, Kelaidonis K, Ligielli I, Moore GJ, Gadanec LK, et al. Understanding the Driving Forces That Trigger Mutations in SARS-CoV-2: Mutational Energetics and the Role of Arginine Blockers in COVID-19 Therapy. Viruses. 2022;14(5). [PubMed ID: 35632769]. [PubMed Central ID: PMC9143829]. https://doi.org/10.3390/v14051029.

  • 13.

    Kawashima S, Matsui Y, Adachi T, Morikawa Y, Inoue K, Takebayashi S, et al. Ensitrelvir is effective against SARS-CoV-2 3CL protease mutants circulating globally. Biochem Biophys Res Commun. 2023;645:132-6. [PubMed ID: 36689809]. [PubMed Central ID: PMC9839456]. https://doi.org/10.1016/j.bbrc.2023.01.040.

  • 14.

    Mukae H, Yotsuyanagi H, Ohmagari N, Doi Y, Sakaguchi H, Sonoyama T, et al. Efficacy and Safety of Ensitrelvir in Patients With Mild-to-Moderate Coronavirus Disease 2019: The Phase 2b Part of a Randomized, Placebo-Controlled, Phase 2/3 Study. Clin Infect Dis. 2023;76(8):1403-11. [PubMed ID: 36477182]. [PubMed Central ID: PMC10110269]. https://doi.org/10.1093/cid/ciac933.

  • 15.

    Mukae H, Yotsuyanagi H, Ohmagari N, Doi Y, Imamura T, Sonoyama T, et al. A Randomized Phase 2/3 Study of Ensitrelvir, a Novel Oral SARS-CoV-2 3C-Like Protease Inhibitor, in Japanese Patients with Mild-to-Moderate COVID-19 or Asymptomatic SARS-CoV-2 Infection: Results of the Phase 2a Part. Antimicrob Agents Chemother. 2022;66(10). e0069722. [PubMed ID: 36098519]. [PubMed Central ID: PMC9578433]. https://doi.org/10.1128/aac.00697-22.

  • 16.

    Eskandary F, Durr M, Budde K, Doberer K, Reindl-Schwaighofer R, Waiser J, et al. Clazakizumab in late antibody-mediated rejection: study protocol of a randomized controlled pilot trial. Trials. 2019;20(1):37. [PubMed ID: 30635033]. [PubMed Central ID: PMC6329051]. https://doi.org/10.1186/s13063-018-3158-6.

  • 17.

    Lonze BE, Spiegler P, Wesson RN, Alachkar N, Petkova E, Weldon EP, et al. A Randomized Double-Blinded Placebo Controlled Trial of Clazakizumab for the Treatment of COVID-19 Pneumonia With Hyperinflammation. Crit Care Med. 2022;50(9):1348-59. [PubMed ID: 35583232]. [PubMed Central ID: PMC9380150]. https://doi.org/10.1097/CCM.0000000000005591.

  • 18.

    Posadas-Sanchez R, Fragoso JM, Sanchez-Munoz F, Rojas-Velasco G, Ramirez-Bello J, Lopez-Reyes A, et al. Association of the Transmembrane Serine Protease-2 (TMPRSS2) Polymorphisms with COVID-19. Viruses. 2022;14(9). [PubMed ID: 36146782]. [PubMed Central ID: PMC9505830]. https://doi.org/10.3390/v14091976.

  • 19.

    Jackson CB, Farzan M, Chen B, Choe H. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1):3-20. [PubMed ID: 34611326]. [PubMed Central ID: PMC8491763]. https://doi.org/10.1038/s41580-021-00418-x.

  • 20.

    Oldenberg E, Schar CR, Lange EL, Plasse TF, Abramson DT, Levitt ML, et al. Abstract B055: New potential therapeutic applications of WX-UK1 as a specific and potent inhibitor of human trypsin-2 and human trypsin-3. Mol Cancer Ther. 2018;17(1_Supplement):B055. https://doi.org/10.1158/1535-7163.Targ-17-b055.

  • 21.

    Plasse TF, Delgado B, Potts J, Abramson D, Fehrmann C, Fathi R, et al. A randomized, placebo-controlled pilot study of upamostat, a host-directed serine protease inhibitor, for outpatient treatment of COVID-19. Int J Infect Dis. 2023;128:148-56. [PubMed ID: 36549549]. [PubMed Central ID: PMC9762116]. https://doi.org/10.1016/j.ijid.2022.12.003.

  • 22.

    Papadopoulou A, Karavalakis G, Papadopoulou E, Xochelli A, Bousiou Z, Vogiatzoglou A, et al. SARS-CoV-2-specific T cell therapy for severe COVID-19: a randomized phase 1/2 trial. Nat Med. 2023;29(8):2019-29. [PubMed ID: 37460756]. https://doi.org/10.1038/s41591-023-02480-8.

  • 23.

    Sojka DK, Huang YH, Fowell DJ. Mechanisms of regulatory T-cell suppression - a diverse arsenal for a moving target. Immunol. 2008;124(1):13-22. [PubMed ID: 18346152]. [PubMed Central ID: PMC2434375]. https://doi.org/10.1111/j.1365-2567.2008.02813.x.

  • 24.

    Gladstone DE, Howard C, Lyu M, Mock J, Adams D, Gibbs K, et al. Randomized, Multi-Center, Double-Blinded, Placebo Controlled Safety and Early Efficacy Trial of Cryopreserved Cord Blood Derived T-Regulatory Cell Infusions (CK0802) in the Treatment of COVID-19 Induced ARDS. (RESOLVE Trial). Blood. 2021;138(Supplement 1):828. https://doi.org/10.1182/blood-2021-153616.

  • 25.

    Gladstone DE, D'Alessio F, Howard C, Lyu MA, Mock JR, Gibbs KW, et al. Randomized, double-blinded, placebo-controlled trial of allogeneic cord blood T-regulatory cells for treatment of COVID-19 ARDS. Blood Adv. 2023;7(13):3075-9. [PubMed ID: 36961352]. [PubMed Central ID: PMC10043947]. https://doi.org/10.1182/bloodadvances.2022009619.

  • 26.

    Gajovic N, Markovic SS, Jurisevic M, Jovanovic M, Arsenijevic N, Mijailovic Z, et al. Galectin-3 as an important prognostic marker for COVID-19 severity. Sci Rep. 2023;13(1):1460. [PubMed ID: 36702907]. [PubMed Central ID: PMC9878495]. https://doi.org/10.1038/s41598-023-28797-5.

  • 27.

    De Biasi S, Meschiari M, Gibellini L, Bellinazzi C, Borella R, Fidanza L, et al. Marked T cell activation, senescence, exhaustion and skewing towards TH17 in patients with COVID-19 pneumonia. Nat Commun. 2020;11(1):3434. [PubMed ID: 32632085]. [PubMed Central ID: PMC7338513]. https://doi.org/10.1038/s41467-020-17292-4.

  • 28.

    Chen YJ, Wang SF, Weng IC, Hong MH, Lo TH, Jan JT, et al. Galectin-3 Enhances Avian H5N1 Influenza A Virus-Induced Pulmonary Inflammation by Promoting NLRP3 Inflammasome Activation. Am J Pathol. 2018;188(4):1031-42. [PubMed ID: 29366678]. https://doi.org/10.1016/j.ajpath.2017.12.014.

  • 29.

    Oka N, Nakahara S, Takenaka Y, Fukumori T, Hogan V, Kanayama HO, et al. Galectin-3 inhibits tumor necrosis factor-related apoptosis-inducing ligand-induced apoptosis by activating Akt in human bladder carcinoma cells. Cancer Res. 2005;65(17):7546-53. [PubMed ID: 16140916]. https://doi.org/10.1158/0008-5472.CAN-05-1197.

  • 30.

    Gaughan EE, Quinn TM, Mills A, Bruce AM, Antonelli J, MacKinnon AC, et al. An Inhaled Galectin-3 Inhibitor in COVID-19 Pneumonitis: A Phase Ib/IIa Randomized Controlled Clinical Trial (DEFINE). Am J Respir Crit Care Med. 2023;207(2):138-49. [PubMed ID: 35972987]. [PubMed Central ID: PMC9893334]. https://doi.org/10.1164/rccm.202203-0477OC.

  • 31.

    Cheruvu N, van Duijn E, Spigt PA, Barbu IM, Toussi SS, Schildknegt K, et al. The Metabolism of Lufotrelvir, a Prodrug Investigated for the Treatment of SARS-COV2 in Humans Following Intravenous Administration. Drug Metab Dispos. 2023;51(10):1419-27. [PubMed ID: 37429728]. https://doi.org/10.1124/dmd.123.001416.

  • 32.

    Hoffman RL, Kania RS, Brothers MA, Davies JF, Ferre RA, Gajiwala KS, et al. Discovery of Ketone-Based Covalent Inhibitors of Coronavirus 3CL Proteases for the Potential Therapeutic Treatment of COVID-19. J Med Chem. 2020;63(21):12725-47. [PubMed ID: 33054210]. https://doi.org/10.1021/acs.jmedchem.0c01063.

  • 33.

    Allais C, Bernhardson D, Brown AR, Chinigo GM, Desrosiers JN, DiRico KJ, et al. Early Clinical Development of Lufotrelvir as a Potential Therapy for COVID-19. Org Process Res Dev. 2023;12(12). [PubMed ID: 37552749]. https://doi.org/10.1021/acs.oprd.2c00375.

  • 34.

    Zhu T, Pawlak S, Toussi SS, Hackman F, Thompson K, Song W, et al. Safety, Tolerability, and Pharmacokinetics of Intravenous Doses of PF-07304814, a Phosphate Prodrug Protease Inhibitor for the Treatment of SARS-CoV-2, in Healthy Adult Participants. Clin Pharmacol Drug Dev. 2022;11(12):1382-93. [PubMed ID: 36285536]. [PubMed Central ID: PMC9874748]. https://doi.org/10.1002/cpdd.1174.

  • 35.

    Robinson P, Toussi SS, Aggarwal S, Bergman A, Zhu T, Hackman F, et al. Safety, Tolerability, and Pharmacokinetics of Single and Multiple Ascending Intravenous Infusions of PF-07304814 (Lufotrelvir) in Participants Hospitalized With COVID-19. Open Forum Infect Dis. 2023;10(8):ofad355. [PubMed ID: 37559753]. [PubMed Central ID: PMC10407246]. https://doi.org/10.1093/ofid/ofad355.