J Korean Med Sci. 2024 Apr 15;39(14):e134. English.
Published online Apr 09, 2024.
© 2024 The Korean Academy of Medical Sciences.
Review

Review of COVID-19 Therapeutics by Mechanism: From Discovery to Approval

Hee Sun Choi,1,* A Young Choi,1,* Jeffrey B. Kopp,2 Cheryl A. Winkler,3 and Sung Kweon Cho1,3
    • 1Department of Pharmacology, Ajou University School of Medicine, Suwon, Korea.
    • 2Kidney Disease Section, Kidney Diseases, Branch, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA.
    • 3Basic Research Laboratory, Center for Cancer Research, National Cancer Institute, Frederick, MD, USA.
Received January 11, 2024; Accepted March 18, 2024.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (https://creativecommons.org/licenses/by-nc/4.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

The global research and pharmaceutical community rapidly mobilized to develop treatments for coronavirus disease 2019 (COVID-19). Existing treatments have been repurposed and new drugs have emerged. Here we summarize mechanisms and clinical trials of COVID-19 therapeutics approved or in development. Two reviewers, working independently, reviewed published data for approved COVID-19 vaccines and drugs, as well as developmental pipelines, using databases from the following organizations: United States Food and Drug Administration (US-FDA), European Medicines Agency (EMA), Japanese Pharmaceutical and Medical Devices Agency (PMDA), and ClinicalTrials.gov. In all, 387 drugs were found for initial review. After removing unrelated trials and drugs, 66 drugs were selected, including 17 approved drugs and 49 drugs under development. These drugs were classified into six categories: 1) drugs targeting the viral life cycle 2) Anti-severe acute respiratory syndrome coronavirus 2 Monoclonal Antibodies, 3) immunomodulators, 4) anti-coagulants, 5) COVID-19-induced neuropathy drugs, and 6) other therapeutics. Among the 49 drugs under development are the following: 6 drugs targeting the viral life cycle, 12 immunosuppression drugs, 2 immunostimulants, 2 HIF-PHD targeting drugs, 3 GM-CSF targeting drugs, 5 anti-coagulants, 2 COVID-19-induced neuropathy drugs, and 17 others. This review provides insight into mechanisms of action, properties, and indications for COVID-19 medications.

Graphical Abstract

Keywords
COVID-19; SARS-CoV-2; Clinical Trials; Review

INTRODUCTION

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), was first identified in Wuhan, China in December 2019 and rapidly spread around the world.1 As of December 24, 2023, a total of 773,119,173 people were confirmed to have been infected with COVID-19 worldwide. Of these, 6,990,067 patients died, with a mortality rate of 1%.2

Because the COVID-19 pandemic was an unanticipated and unprecedented worldwide crisis, there were no established treatments or vaccine. The National Institutes of Health (NIH) announced the accelerating COVID-19 therapeutic interventions and vaccines (ACTIV) public-private partnership to develop a coordinated research strategy for prioritizing and speeding the development of the most promising treatments and vaccines on April 17, 2020.3 Developing a strategy for COVID-19 treatment took a multi-disciplinary approach that emphasized preventing virus replication and relieving symptoms and complications. Following this initiative, most major research organizations around the world have launched studies and clinical trials for repurposing known drugs and for discovering new ones.

Currently, there are several approved drugs for COVID-19. As of May 2023, World Health Organization (WHO) declares end to COVID-19 as a global health emergency thanks to improved clinical management and wide-spread acquired immunity by natural infection or vaccines or both; however, certain populations, which include the very young, the elderly and those who are immunocompromised, are still experiencing severe covid and death. There are many on-going clinical trials to develop drugs effective against new variants, to prepare for the next new strain of SARS-CoV2, to shorten the dosing period, or to reduce drug-drug interactions and side effects.

Herein, we summarize mechanisms and clinical trials of COVID-19 therapeutics that have been approved or are under development to illustrate the road we have taken and what the world is preparing for in the next phase.

METHODS

Two independent reviewers reviewed data on approved COVID-19 drugs and developmental pipelines using the following databases: US-FDA, European Medicines Agency (EMA), Japanese Pharmaceutical and Medical Devices Agency (PMDA), and ClinicalTrials.gov. Mechanisms of action and clinical trials of treatments were searched via PubMed, medRxiv, public institution websites, pharmaceutical companies’ sites, and ClinicalTrials.gov. Ambiguous points in data collection and summarization were resolved through discussion among reviewers. We investigated updated information on treatments through March 2023. Fig. 1 illustrates the selection process for the reviews. Most trials used a primary endpoint assessed by an 8-point National Institute of Allergy and Infectious Disease (NIAID) ordinal scale of COVID-19 severity4 or a 0 to 10-point WHO clinical progression scale.5

Fig. 1
Flow chart showing the review process for selection of publications to be reviewed.
COVID-19 = coronavirus disease 2019, FDA = US Food and Drug Administration, EMA = European Medicines Agency, PMDA = Japanese Pharmaceutical and Medical Devices Agency, HIF-PHD = hypoxia-inducible factor prolyl hydroxylase, GM-CSF = granulocyte-macrophage colony-stimulating factor.

We identified COVID-19 treatments in databases using keywords such as ‘COVID-19,’ ‘SARS-CoV-2,’ and ‘Coronavirus.’ A total of 387 drugs were identified, of which 248 were less relevant to COVID-19 treatment. After removing these, the remaining 139 drugs were classified based on whether they were approved or are under development. Table 1 summarizes European Use Authorization (EUA), US Food and Drug Administration (FDA) and European Medicines Agency (EMA) approved for drugs approved within specific countries.

Table 1
Approved medications for COVID-19 (N = 18)

The key elements of treatment for COVID consist of antiviral therapy, immunomodulation, prevention of blood-coagulation, and hypoxic damage control. We classified drugs into six categories, which included 1) Anti-SARS-CoV-2 monoclonal antibodies (Table 2), 2) viral life cycle inhibitors (Table 3), 3) immunomodulators (Tables 4, 5, 6), 4), anti-coagulants (Table 7), 5) drugs for treating COVID-19 induced neuropathy (Table 8), and 6) drugs targeting other mechanisms (Tables 9 and 10). During the review process, we also identified and classified drugs in a list of withdrawn COVID-19 medications: 1) the pipeline was discontinued for further development, 2) clinical trials failed to meet the primary end-point, 3) clinical trials were not active for more than a year, 4) drug was no longer listed in the company’s pipeline, 5) the most recent clinical trial was terminated, and 6) exploratory clinical trials without further development (Supplementary Table 1).

Table 3
COVID-19 drugs under development targeting viral life cycle (N = 6)

Table 4
Immunosuppressive drugs under development for COVID-19 treatment (N = 12)

Table 5
Immunostimulants under development for COVID-19 legend (N = 2)

Table 6
Drugs under development targeting GM-CSF (N = 3)

Table 7
Drugs under development for preventing blood coagulation in COVID-19 (N = 5)

Table 8
Drugs under developing for treating COVID-19-induced neuropathy (N = 2)

Table 9
Drugs under development targeting the HIF-PHD pathway (N = 2)

RESULTS

The NIH ACTIV public-private partnership classifies treatments based on severity of illness.6 We classified treatments based on the mechanism of action. This review included all drugs from ACTIV trials. We further investigated developing pipelines, worldwide. Each drug was reviewed for status of approval, indication for treatment, dose information, route of administration, and reported side effects.

  • Part 1. Approved COVID-19 Therapeutics

  • Part 2. Anti-SARS-CoV-2 Monoclonal Antibodies

  • Part 3. Developing COVID 19 drugs targeting the viral life cycle

  • Part 4. Developing COVID 19 immunomodulators

  • Part 5. Developing anti-coagulants in COVID-19

  • Part 6. Developing drugs for COVID-19-induced neuropathy

  • Part 7. Other mechanism drug for COVID-19 therapeutics under development

PART 1. APPROVED COVID-19 THERAPEUTICS

Approved anti-viral agents

Paxlovid (nirmatrelvir/ritonavir)

Paxlovid consists of two molecules. Nirmatrelvir is a peptidomimetic inhibitor of the SARS-CoV-2 main protease (Mpro), also referred to as 3C-like protease (3CLpro) or nsp5 protease. A peptidomimetic is a molecule whose structure mimics that of a natural peptide. Inhibition of SARS-CoV-2 Mpro renders the protein incapable of processing polyprotein precursors, thus preventing viral replication. In contrast, ritonavir is a protease inhibitor that boosts the activity of nirmatrelvir by inhibiting cytochrome P450 3A (CYP3A), the enzyme required for metabolizing nirmatrelvir. When administered together with nirmatrelvir, ritonavir increases plasma concentrations of nirmatrelvir. Paxlovid received an EUA from the FDA on December 22, 2021 for treating adults and pediatric patients with mild-to-moderate COVID-19 who are at high risk for progressing to severe COVID-19.7 Paxlovid received final approval from the FDA for treating mild to moderate COVID-19 on March 17, 2023. EU approval was received on January 28, 2022. It is not authorized for initiation of treatment in patients requiring hospitalization due to severe or critical COVID-19, nor is it authorized for use as pre-exposure or as post-exposure prophylaxis.

The recommended Paxlovid dosage is 300 mg nirmatrelvir (two 150 mg tablets) with 100 mg ritonavir (one 100 mg tablet) taken together orally every 12 hours for 5 days. Paxlovid should be administered as soon as possible after a diagnosis of COVID-19. It should be also be administered within 5 days of symptom onset.8 It cannot be used for over 5 days.9 Paxlovid is not recommended for those with severe renal impairment (estimated glomerular filtration rate < 30 mL/min/1.73 m2).8 In Korean clinical studies, Paxlovid demonstrated safety and a reduction in viral load against the omicron variant.10 Moreover, it lowered the requirement for supplemental oxygen.11

Lagevrio (molnupiravir)

Molnupiravir is a prodrug with antiviral activity against SARS-CoV-2. It is metabolized to a cytidine nucleoside analog, forming a pharmacologically active ribonucleoside triphosphate (NHC-TP). NHC-TP incorporation into SARS-CoV-2 RNA by viral RNA polymerase (nsp12) introduces copying errors during viral RNA replication. This mechanism of action is known as ‘viral error catastrophe’ and the drug was originally developed at Emory University12 to treat influenza.

Molnupiravir received an EUA from the FDA on December 23, 2021 to treat mild-to-moderate COVID-19 in adults who are at high risk for progression to severe COVID-19 and for whom alternative COVID-19 treatment options authorized by FDA are not accessible or clinically appropriate.13 However, EMA did not approve the drug for treating COVID-19 in adults until February 23, 2023, with the main reason being that the clinical benefit of molnupiravir was limited for those with COVID-19 who were not receiving supplemental oxygen.14

The dose for adult patients is 800 mg (four 200 mg capsules) taken orally every 12 hours for 5 days. The dug is not authorized for use for longer than five consecutive days. It is not authorized for use in patients who are less than 18 years of age, nor for initiation of treatment in patients who are hospitalized due to COVID-19. Molnupiravir is also not authorized for pre-exposure or post-exposure prophylaxis.15

Azvudine

Azvudine is a thymus-homing nucleoside analogue drug that inhibits viral RNA-dependent RNA polymerase (RdRp).16 It was approved by the National Medical Products Administration (NMPA) of China (the equivalent of the US Food and Drug Administration) on July 25, 2022 for COVID-19 treatment.17 Although there are no specified dosage guidelines for COVID-19, the usual clinical dose of azvudine is 5 mg orally once daily.16

Veklury (remdesivir)

Remdesivir is an adenosine nucleotide prodrug that is metabolized within host cells to form a pharmacologically-active nucleoside triphosphate metabolite. Remdesivir triphosphate acts as an analog of adenosine triphosphate (ATP) and competes with the natural ATP substrate for incorporation into nascent RNA chains by SARS-CoV-2 RNA-dependent RNA polymerase. Remdesivir triphosphate also inhibits viral RNA synthesis through incorporation of the complementary natural nucleotide.

Remdesivir received FDA approval for adult and pediatric patients with COVID-19 requiring hospitalization18 on October 22, 2020 as the first approved treatment for COVID-19. FDA expanded its use for treating outpatients with mild-to-moderate COVID-19 on January 21, 2022. Remdesivir received EMA approval on July 3, 2020. According to the EMA, it is indicated for patients with pneumonia requiring supplemental oxygen.

The recommended dose is a 200 mg intravenous infusion on the first day and 100 mg intravenous infusion on the following day. Treatment duration is at least 5 days but not more than 10 days. Treatment duration is 3 days for patients who do not require supplemental oxygen. It must not be given as an intramuscular injection. The recommended dose and treatment duration vary for pediatric patients. Increased liver enzymes (transaminases) and a coagulation tendency are its common adverse effects, affecting more than 1 in 10 patients.19 In the treatment guidelines for mild to moderate COVID-19 patients in South Korea, Remdesivir is widely utilized.20 Additionally, hepatic injury and anemia have been identified as the most common side effects.21

It was reported that remdesivir is superior to placebo in shortening the time to recovery in adults who were hospitalized with SARS-CoV-2 infection and had no effect on all-cause mortality at up to day 28 in hospitalized adults with SARS-CoV-2 infection.22, 23

Mindvy (VV116)

Mindvy is an oral analogue of remdesivir. Mindvy is a nucleoside triphosphate analog drug that binds to the active site of RdRp and inhibits viral replication by creating an error-ridden and therefore defective genome.24, 25, 26 It was conditionally approved by NMPA for treating adult patients with COVID-19.25 It is also approved in Uzbekistan. A dose of 200 mg twice daily is recommended for COVID-19 treatment.26

2-DG (2-deoxy-D-glucose)

2-DG (2-deoxy-D-glucose) is a glucose analogue that competes with it and inhibits metabolism within host cells. It inhibits synthesis of viral molecules and energy production.27 2-DG received approval from the Drugs Controller General of India (DCGI) on May 1, 2021, for adjunctive treatment of moderate-to-severe COVID-19 illness. A clinically tolerable dose is 63 mg/kg. Further studies are underway to determine the optimal dose.28

Approved COVID-19 immunomodulators

Kineret (anakinra)

Anakinra is an immunmodulating, interleukin (IL)-1 type I receptor inhibitor that blocks both IL-1α and IL-1β.29 Anakinra has received FDA approved for treatment of three diseases: rheumatoid arthritis (RA), cryopyrin-associated periodic syndromes, and deficiency of IL-1 receptor antagonist.30 Anakinra received an EUA from the FDA on November 8, 2022 to treat hospitalized COVID-19 pneumonia adult patients requiring supplemental oxygen who are at high risk for progressing to severe respiratory failure and likely to have an elevated soluble urokinase plasminogen activator receptor (suPAR).31 EU approval was obtained on December 17, 2021.32 The recommended dose is 100 mg subcutaneous injection once daily for 10 days. Common side effects (occurring more than 10% of patients) are pain and redness at the injection site and increased total cholesterol levels.29

Artlegia (olokizumab)

Olokizumab is a monoclonal antibody targeting IL-6.24 It received approval from the Ministry of Health of the Russian Federation (Minzdrav) for COVID-19 pneumonia.33 The recommended dose is a single subcutaneous injection of 64 mg, administered in the thigh or anterior abdominal wall once every four weeks. It is not recommended for patients aged below 18 years of age.24

Actemra (tocilizumab)

Tocilizumab is an IL-6 receptor (IL-6R) inhibitor that binds membrane-bound IL-6 receptors.34 It has FDA approval for RA, giant cell arteritis, systemic sclerosis-associated interstitial lung disease, polyarticular juvenile idiopathic arthritis (PJIA), systemic juvenile idiopathic arthritis, cytokine release syndrome (CRS), and COVID-19.35

Tocilizumab received an EUA from the FDA on June 24, 2021 for hospitalized COVID-19 patients aged 2 years and older with systemic glucocorticoids who require supplemental oxygen or mechanical ventilation. On December 21, 2022, tocilizumab received Biologics License Application (BLA) 125276 for the same patients.36 EU approval of tocilizumab was obtained on December 7, 2021.37

The recommended dose of tocilizumab is a single 60-minute intravenous infusion at 8 mg/kg (maximum 800 mg). If any clinical improvements are found, one additional infusion can be administered after an interval of at least 8 hours. Patients with abnormal levels of liver enzymes, absolute neutrophil count (ANC), and platelet count are not recommended to receive tocilizumab.34

Ilsira (levilimab)

Levilimab is a monoclonal antibody targeting the IL-6 receptor. Levilimab was developed to prevent cytokine storm caused by COVID-19.38, 39 It received approval from the Ministry of Health of the Russian Federation (Minzdrav) on June 5, 2020 for COVID-19 treatment.38 Subcutaneous injection of Levilimab at 324 mg is generally recommended.40 It apparently has not been approved in the USA.

Alzumab (itolizumab)

Itolizumab is a monoclonal antibody against CD6, a costimulatory molecule that stimulates T cell proliferation. The drug was developed to prevent CRS or acute respiratory distress syndrome (ARDS) due to COVID-19. It was approved for chronic plaque psoriasis in 2013 in India. Itolizumab also received restricted emergency use approval from the Drugs Controller General of India (DCGI) in July 2020 for treating COVID-19-related CRS or ARDS.41

The recommended dose of itolizumab is 1.6 mg/kg for the first dose, administered as an intravenous infusion. One week later, a second dose 0.8 mg/kg is given.42 Patients with hepatic or renal disease, those with infectious diseases, and pregnant or lactating women are not recommended to receive itolizumab.41

Olumiant (baricitinib)

Olumiant, also known as baricitinib, is a Janus kinase (JAK) inhibitor.43 Baricitinib received FDA approval for RA, alopecia areata, and COVID-19.44 It received an EUA from the FDA on November 19, 2020 for use in combination with remdesivir for COVID-19 patients requiring supplemental oxygen. On May 10, 2022, the FDA approved baricitinib for hospitalized COVID-19 adults requiring supplemental oxygen, non-invasive or invasive mechanical ventilation, or extracorporeal membrane oxygenation (ECMO).44 On October 27, 2022, FDA expanded the EUA for use of baricitinib in hospitalized adults and pediatric patients aged 2 to 18 years requiring supplemental oxygen.45 However, the EMA did not approve baricitinib for treating hospitalized COVID-19 patients until December 7, 2022.46

The recommended dose of baricitinib for pediatric patients 9 years and older and for adult patients is 4 mg. For pediatric patients aged 2 to 9 years, the recommended dose of baricitinib is 2 mg orally once daily. The recommended treatment duration is 14 days or until hospital discharge.43, 44, 45 In Korea, various immunomodulators, including baricitinib, were administered to patients requiring supplemental oxygen.47 Dosage modifications are recommended for patients with renal or hepatic impairment.43 Olumiant is not recommended for patients with an absolute lymphocyte count less than 200 cells/µL or an ANC less than 500 cells/µL.44 Known adverse events are serious venous thromboses including pulmonary embolism.43

According to the NIH COVID-19 treatment guidelines, non-hospitalized patients at risk of progressing to severe COVID-19 symptoms may be offered Paxlovid, remdesivir, or molnupiravir. For hospitalized patients, those not requiring supplemental oxygen may receive remdesivir, while those requiring supplemental oxygen may receive dexamethasone and remdesivir, and in more severe cases, baricitinib or tocilizumab may be considered.

PART 2. ANTI-SARS-COV-2 MONOCLONAL ANTIBODIES

Monoclonal antibodies blocking SARS-CoV-2 cell attachment are no longer recommended in treating COVID-19 due to an immune evasion mechanism.48 Approved therapeutic monoclonal antibodies have failed to neutralize either omicron subvariants such as BQ.1.1 or XBB. The FDA has updated drug labels to address the limitation of monoclonal antibodies blocking SARS-CoV-2 cell attachment. New therapeutic monoclonal antibodies are needed for future COVID pandemics. After WHO declares an end to COVID-19 as a global health emergency, it is likely there will be no new monoclonal antibodies at advanced stages of research.

Anti-SARS-CoV-2 monoclonal antibodies

Evusheld (tixagevimab/ cilgavimab)

Evusheld (previously known as AZD8895 and AZD1061) contains tixagevimab and cilgavimab. Tixagevimab and cilgavimab were designed to attach to the spike protein of SARS-CoV-2 (the virus that causes COVID-19) at two different sites. Evusheld hinders viral entry by attaching to the viral spike protein.49

On December 8, 2021, Evusheld receive Emergency Use Authorization (EUA) from the US Food and Drug Administration (FDA) for pre-exposure prophylaxis of COVID-19 in adults and pediatric individuals, and treatment of those with mild to moderate COVID-19 who are at high risk for developing severe disease. However, due to new SARS-CoV-2 variants, EUA of Evusheld is no loonger valid in the U.S..50, 51 Currently, Evusheld approval remains valid throughout the EU.

For pre-exposure prophylaxis of COVID-19, 150 mg of tixagevimab and 150 mg of cilgavimab, administered as two separate sequential intramuscular injections, are recommended. For treatment, 300 mg of tixagevimab and 300 mg of cilgavimab within 7 days of showing symptoms of COVID-19 administered as two separate sequential intramuscular injections are recommended. Tixagevimab and cilgavimab must be given as separate sequential intramuscular injections at different injection sites in two different muscles, preferably in gluteal muscles. These therapies are indicated for treating adults and adolescents aged 12 years or older not requiring supplemental oxygen who are at an increased risk of progressing to severe COVID-19. Systemic hypersensitivity (allergic) reactions and local reactions at the site of injection are the most common side effects.52

Regkirona (regdanvimab)

Regkirona, also known as CT-P59 is a recombinant human IgG1 monoclonal antibody that binds to the receptor binding domain (RBD) of the spike(s) protein of SARS-CoV-2, consequently blocking cellular entry and SARS-CoV-2 infection. Regkirona received approval first from the MFDS (Minister of Food and Drug Safety), of Korea on December 29, 2020. Regkirona received approval from EU on November 12, 2021. It is indicated for treating confirmed COVID-19 in adult patients who do not require supplemental oxygen but are at a high risk of progressing to severe COVID-19.

The recommended dosage of Regkirona is 40 mg/kg via a single dose by intravenous infusion. The maximum dosage is 8000 mg. Adverse allergic responses, including fever, dyspnea, tachycardia, bradycardia, and itching due to Regkirona use are uncommon, occurring in about 1 person per 100.53 With the emergence of the delta variant, Regkirona is no longer recommended.54

Ronapreve (casirivimab/imdevimab)

Ronapreve consists of two recombinant human monoclonal antibodies, casirivimab (IgG1κ) and imdevimab (IgG1λ), that target non-overlapping epitopes of the spike protein RBD of SARS-CoV-2. The drug prevents RBD binding to the human ACE2 receptor and thereby blocks viral entry into cells. Ronapreve received approval throughout the EU on November 12, 2021.

Ronapreve can be used for treating and preventing COVID-19. The recommended dosage for treatment is 600 mg for casirivimab and 600 mg for imdevimab, administered as a single intravenous infusion or subcutaneous injection. They should be given within seven days of the onset of COVID-19 symptoms. The recommended dosage for pre-exposure prophylaxis is 600 mg for casirivimab and 600 mg for imdevimab as an initial dose, administered as a single intravenous infusion or subcutaneous injection. Subsequent doses of 300 mg of casirivimab and 300 mg of imdevimab can be administered as a single intravenous infusion or as subcutaneous injections every four weeks, until prophylaxis is no longer required. There are no data on repeat dosing beyond 24 weeks (6 doses). The recommended dosage for post-exposure prophylaxis is 600 mg for casirivimab and 600 mg for imdevimab, administered as a single intravenous infusion or subcutaneous injection. Casirivimab combined with imdevimab should be given as soon as possible after exposure to persons with COVID-19.55 Ronapreve is not currently authorized for use in any U.S. after emergence of omicron variants.

Xevudy (sotrovimab)

Xevudy, also known as VIR-7831 and GSK4182136, is a human IgG1 monoclonal antibody that binds to a highly conserved epitope on the spike protein RBD of SARS-CoV-2. Xevudy received an EUA from the FDA on May 26, 2021 for treating mild-to-moderate COVID-19 in adults and in children (12 years of age and older weighing at least 40 kg) (+) and those who are at high risk for progression to severe COVID-19, including hospitalization and death. The EUA of Xevudy is not valid for the omicron variant.56, 57 Xevudy received approval throughout the EU on December 17, 2021.

The recommended dose is a single 500 mg intravenous infusion. Allergic reactions to Xevudy are common, affecting up to 1 in 10 people.58 Xevudy is no longer authorized to treat COVID-19 in any U.S. caused by the Omicron BA.2 sub-variants.

BRII-196/BRII-198 (amubarvimab/romlusevimab)

BRII-196/BRII-198 contains two monoclonal antibodies, amubarvimab (BRII-196) and romlusevimab (BRII-198). Each binds non-overlapping epitopes of the spike protein of SARS-CoV-2 and prevents viral entry into cells. BRII-196/BRII-198 received approval from NMPA of China in December 2021 for treating adult patients (18 years aged and older) with mild COVID-19. The recommended dose is 1,000 mg for amubarvimab and 1,000 mg for romlusevimab, administered as separate sequential intravenous infusions.59 BRII-196/BRII-198 is no longer recommended to use after the emergence of omicron variants.

PART 3. DEVELOPING COVID 19 DRUGS TARGETING THE VIRAL LIFE CYCLE

Fig. 2 illustrates approved drugs and drugs under development that target the COVID-19 viral life cycle.

Fig. 2
SARS-CoV-2 life cycle. Medications in black font are approved. Medications in blue font are under development.
ER = endoplasmic reticulum.

Nucala (meplazumab)

Nucala (meplazumab) is an IgG2 monoclonal antibody targeting CD147, a member of the immunoglobulin super-family with diverse functions related to cell differentiation, function, and migration. It is being studied for treating severe malaria and COVID-19 pneumonia by Jiangsu Pacific Meinuoke Pharmaceuticals in China.60 CD147 (basigin) is a receptor for SARS-CoV-2 entry into cells, and can facilitate viral entry even when host cells lack ACE2 receptors.61

Meplazumab is under development to treat hospitalized COVID-19 patients by targeting CD147. A phase 2/3 study is recruiting participants in China (NCT04586153) to evaluate the safety and efficacy of meplazumab in addition to the standard of care for treating hospitalized adults with COVID-19. A total of 276 subjects will be randomized and allocated 1:1 (138:138) to receive 0.2 mg/kg meplazumab or control. The primary outcome will be reduced mortality and improved pulmonary function, as assessed by comparing the proportion of subjects who are alive and discharged without needing supplemental oxygen from day 1 to day 29 after treatment.

Aplidin (Plitidepsin)

Plitidepsin is an inhibitor of eukaryotic-translation-elongation-factor-1-alpha (eEF1A). eEF1a blocks the translation of SARS-CoV-1open reading frames 1A and 1B (ORF1A and ORF1B). Plitidepsin was developed for treating multiple myeloma, leukemia, lymphoma, and melanoma by PharmaMar in Spain due to its anti-tumor properties.62, 63 PharmaMar is recruiting participants for a phase 2 study (NCT05705167) to evaluate the safety and efficacy of plitidepsin in immunosuppressed COVID-19 patients. Subjects will be randomized to receive 2.5 mg plitidepsin via intravenous infusion together with supportive care, or will receive supportive care only. The primary outcome will be mortality rate from day 1 to day 30.

Ebselen (SPI-1005)

Ebselen (SPI-1005) (Sound Pharmaceuticals, Seattle WA, USA is an oral Mpro inhibitor. It is being studied for treating Meniere disease, noise-induced hearing loss (NIHL), and COVID-19.64 It protects cells from oxidative damage.65 The main mechanism of action of this drug in treating COVID-19 ins Mpro inhibition.66, 67 A phase 2 study to evaluate the safety and efficacy of ebselen in COVID-19 patients is enrolling subjects now (NCT04483973). Subjects will receive of 400 or 800 mg ebselen orally, or placebo, twice daily for 14 days. The primary outcome measure is the number of subjects who manifest adverse events within 30 days.

Obeldesivir (GS-5245)

Obeldesivir is an oral version of remdesivir. Obeldesivir is a nucleoside analogue that inhibits RdRp. The compound was developed by Gilead Sciences, Foster City, California.68 Gilead Sciences is recruiting participants in the USA (NCT05603143) for a phase 3 study to evaluate the efficacy and safety of GS-5245 in patients with COVID-19 who have a high risk of developing severe illness. Participants will be randomized into GS-5245 or placebo groups. Participants in the GS-5245 group will be administered 350 mg GS-5245 orally twice daily without food, for 5 days. Participants in the placebo group will receive a placebo orally twice daily without food for 5 days. The primary outcome measure will be the proportion of COVID-19-related hospitalizations or all-cause death from the date of the first dose to day 29.

Sabizabulin

Sabizabulin is a microtubule disruptor that induces depolymerization of microtubules.69 Sabizabulin has been studied for treating COVID-19 and cancer by Veru (Miami, FL, USA). Veru applied for an EUA for sabizabulin, however, it was rejected on March 2, 2023.70

A phase 3 study to evaluate the efficacy and safety of sabizabulin in COVID-19 patients has been completed (NCT04842747); data are pending. The sabizabulin group received 9 mg of daily sabizabulin orally, together with the standard of care, until released from the hospital or for up to 21 days. The placebo group received a placebo capsule with the standard of care until released from the hospital or for up to 21 days. The primary outcome measure was the proportion of patients in the study who died within 60 days.

Ampligen (rintatolimod)

Ampligen (rintatolimod) is a Toll-like receptor 3 agonist. The compound consists of double-stranded RNA (dsRNA) developed by AIM ImmunoTech, Philadelphia, PA, USA. It has received FDA approval for treating severe chronic fatigue syndrome and pancreatic cancer. The route of administration is intravenous infusion. Rintatolimod binds Toll-like receptor 3 and thereby activates the ribonuclease L (RNase L), which degrades viral RNA. Furthermore, rintatolimod activates both innate and adaptive immune systems by stimulating production of various cytokines.71 It is currently being studied as post-COVID treatment.72

A phase 2 study of rintatolimod in patients with post-COVID patients with fatigue has been conducted (NCT05592418). Subjects were randomized into two groups. The treatment group received 100 to 400 mg rintatolimod twice weekly via intravenous infusion for 12 weeks. The control group received normal saline 40 to 160 mL twice weekly via intravenous infusion for 12 weeks. The primary outcome measure was PROMIS Fatigue Score change from baseline to week 13. Anticipated study completion was November 2023.

PART 4. DEVELOPING COVID 19 IMMUNOMODULATORS

Fig. 3 illustrates approved and under development immune modulators used in COVID-19 treatment.

Fig. 3
Immune modulators used in COVID-19 treatment. Medications written in black are approved and medications written in blue are under development. Gray boxes denote medications that are immunostimulants. Yellow boxes denote medications that are immunosuppressants.
JAK = Janus kinase, IL = interleukin.

Developing drugs for immunosuppression in COVID-19

Jaktinib

Jaktinib is a JAK inhibitor that has been used to prevent and treat cytokine storms in COVID-19 patients. Jaktinib inhibits JAK1, JAK2, JAK3, and TYK2 by blocking the JAK-STAT signaling pathway (suppressing the release of IL-2, IL-4, IL-6, IL-7, and IL-10).73

Jaktinib was developed by Suzhou Zelgen Biopharmaceuticals, Shanghai, China. The drug is being studied for treating myelofibrosis, severe alopecia areata, atopic dermatitis, acute graft versus host disease (GvHD), and COVID-19.74

A phase 2 study to evaluate the safety and efficacy of jaktinib in severe COVID-19 pneumonia patients is planned (NCT05686629) but is not recruiting subjects yet. The treatment group will receive two 50 mg jaktinib doses or placebo tablets twice daily. The primary outcome measures will be the proportion of subjects who show clinical improvement (a score of more than 2) in the NIAID Ordinal Scale (NIAID-OS) for COVID-19, a change in the value of NIAID-OS, the time for subjects to reach 1-3 points, and the time to discharge.

Ilaris (canakinumab)

Ilaris is an IL-1β blocker developed by Novartis, Basel, Switzerland.75 The drug is in development for autoinflammatory periodic fever syndromes and active Still disease. It is now being repurposed to alleviate clinical manifestations in patients with COVID-19.76, 77

Katia and colleagues reported on a single center, single arm study conducted in 20209 in 20 patients with COVID-related severe acute respiratory syndrome. Treatment with canakinumab was associated with reduced requirement for supplemental oxygen.

A phase 3 study to compare the efficacy and safety of canakinumab with standard of care in COVID-19-induced pneumonia and CRS patients has been completed (NCT04362813). The canakinumab group received a single intravenous dose of canakinumab, dosed according to body weight (450 mg for 40 to 60 kg, 600 mg for 60 to 80 kg, and 750 mg for more than 80 kg) in 250 mL of 5% dextrose on day 1. The control group received a single dose of 250 mL of 5% dextrose via intravenous infusion on day 1. All subjects received standard of care. The primary outcome measure was the number of surviving participants who did not need invasive mechanical ventilation from Day 3 to Day 29. Trial results are pending.

Reparixin

Reparixin is a non-competitive allosteric inhibitor of CXCR1 (keratinocyte-derived chemokine receptor) and CXCR2 (macrophage inflammatory protein 2 receptor). Reparaxin was developed by Dompe’ Pharmaceutical, Milan, Italy. By inhibiting CXCR1/2, it suppresses CXCL8 (IL-8)-induced neutrophil activation and reduces lung edema and neutrophil recruitment into the lung.78

Clinical studies evaluating the efficacy and safety of reparixin efficacy and safety in patients with breast cancer or with pancreatic transplantation have been completed.79

Due to its anti-inflammatory properties, reparixin might be a treatment option to prevent or attenuate CRS.80 Clinical trials to test the efficacy of reparixin for treating COVID-19 are currently underway. A phase 3 study is currently recruiting participants to evaluate the efficacy of reparixin in patients with community-acquired pneumonia, including COVID-19 (NCT05254990). Subjects will receive standard of care and two 600 mg reparixin or placebo tablets three times daily with 8 hours intervals (6 tablets daily) for up to 21 days. The primary outcome will be the proportion of subjects who die or need mechanical ventilation or extracorporeal membrane oxygenation within a 28 day period.

Taltz (ixekizumab)

Ixekizumab is a humanized IL-17A antagonist developed by Eli Lilly and Company (Indianapolis, IN, USA).81 In COVID-19 patients, overactivation of T lymphocytes, which is manifested by an increase in Th17 cell numbers, can lead to increased production of IL-17 and IL-22 cytokines. The use of an IL-17 blocking agent could be effective in treating patients with severe COVID-19.82 Ixekizumab was approved by the FDA for treating plaque psoriasis, psoriatic arthritis, ankylosing spondylitis, and non-radiographic axial spondyloarthritis.81

A phase 3 study aimed at finding an immune modulator of cytokines for subjects with severe COVID-19 was recently completed (NCT04724629). The trial name was Survival RTrial Using CytoKines in COVID-9 (STRUCK). The study drugs included an IL-17 inhibitor (ixekizumab), an IL-2 inhibitor (aldesleukin), and an IL-6 inhibitor (colchicine). The ixekizumab treatment group received 80 mg by subcutaneous injection weekly for four weeks or until hospital discharge. The control group received standard of care including mechanical ventilation or dexamethasone. The primary outcome measure was the proportion of subjects who showed clinical improvement of two points on a seven-category ordinal scale on day 21. Study results are pending.

Remicade (infliximab)

Infliximab blocks tumor necrosis factor (TNF) [formerly named TNF-α) that was developed by Janssen Biotech (Horsham, PA, USA).83 Infliximab received FDA approval for treating Crohn disease, colitis, and RA in 1998.84 Infliximab is now being repurposed to treat COVID-19 patients.

A phase 3 study (ACTIV-1 trial) sponsored by the National Center for Advancing Translational Sciences (NCATS), NIH sought to find an effective immune modulator for patients with COVID-19 and has been completed (NCT04593940). The study consisted of three arms (infliximab, abatacept, and cenicriviroc). Participants in each arm received either drug or placebo, together with standard of care. Participants in the infliximab arm received a single 5 mg/kg dose of infliximab on day 1. The primary outcome measure was the time to recover from COVID-19 up to 29 days. Results are pending.

Orencia (abatacept)

Orencia is a selective T cell co-stimulation modulator that competes with CD28 for binding CD80 and CD86 receptors, thus decreasing T cell proliferation and inhibiting the production of cytokines such as TNF, interferon-γ, and IL-2.85 Neutralizing inflammatory factors in CRS using abatacept could be an effective way to avoid severe inflammation in COVID-19.86

Abatacept was developed by Bristol-Myers Squibb (New York, NY, USA) and was approved by the FDA in 2005 for treating RA, PJIA, and psoriatic arthritis.85 There has been interest in repurposing abatacept to treat patients with COVID-19.

A phase 3 study, the ACTIV-1 IM Trial Immunomodulators for Treating COVID-19 (NCT04593940) was sponsored by National Center for Advancing Translational Sciences (NCATS), NIH and has been complete. This study consisted of three arms. Each arm was provided with a standard of care including remdesivir. The infliximab group received a single 5 mg/kg dose of infliximab via intravenous infusion on day 1. The abatacept arm received a single 10 mg/kg dose of abatacept up to 1,000 mg via intravenous infusion on day 1. The control group received cenicriviroc, a chemokine 2 and 5 receptor antagonist (450 mg on day 1 and 300 mg on days 2 to 29) or a matching placebo. The primary outcome measure was the time to recover from COVID-19 up to day 29. None of the three therapies significantly improved outcomes compared to placebo.

Vascepa (icosapent ethyl)

Vascepa is an ethyl ester of eicosapentaenoic acid (EPA) that reduces hepatic very low-density lipoprotein triglycerides (VLDL-TG) synthesis and enhances triglyceride clearance from circulating VLDL particles. The drug was developed by Amarin, Dublin, Ireland.87 Isopent ethyl (Vascepa) received FDA approval as an adjunct to maximally tolerated statin therapy and diet to reduce triglyceride levels.

EPA improves endothelial function, limits vascular inflammation and reactive oxygen species production, and reduces vascular thrombosis. Through these mechanisms, Vascepa might reduce inflammation and improve symptoms of patients with COVID-19.88

A phase 3 study of Vascepa for COVID-19 has been completed (NCT04460651). This study consisted of a prevention group (enrolling subjects at high risk of infection from COVID-19) and a treatment group (COVID-19-positive subjects). Subjects in the prevention group received 8 g oral Vascepa or placebo every 12 hours on days 1 to 3 and 4 g oral Vascepa or placebo every 12 hours for days 4 to 60. Subjects in the treatment group received 8 g oral Vascepa or placebo every 12 hours on days 1 to 3 and 4 g oral Vascepa or placebo every 12 hours for days 4 to 28. Primary outcome measures were COVID-19-positive subjects in the prevention group within 60 days and hospitalized or deceased subjects in the treatment group due to COVID-19 within 28 days. Results are pending.

NuSepin (HY209)

Nusepin is a fibroblast growth factor 19 (FGF-19) analog developed by Shaperon in South Korea. It targets cytokine storm in COVID-19 by suppressing pro-inflammatory cytokine production.89

A phase 2, 3 study to evaluate the safety and efficacy in COVID-19 pneumonia patients is recruiting participants (NCT05352347). Subjects in the treatment group will receive 0.2 mg/kg or 0.4 mg/kg NuSepin, twice daily by intravenous injection. Subjects in the control group will receive 100 mL normal saline twice daily. Primary outcome measures will be the time to improve more than 2 categories of the WHO 8-point ordinal scale and the time to discharge from the first dosing to day 29.

Ruconest (conestat alfa)

Ruconest (conestat alfa) is a C1 esterase inhibitor (C1INH) that promotes activation of complement. Conestat alfa was developed by Pharming Group, Leiden, Netherlands.90 Conestat alfa targets several proteases of the contact and complement systems. Target proteases of conestat alfa are involved in the activation of complement protein C1, kallikrein, factor XIIa, and factor XIa.90

Activation of the complement system plays a central role in the pathogenesis of acute lung injury. The kallikrein-kinin system is also involved in thrombo-inflammation. Targeting multiple inflammatory cascades with conestat alfa could lead to clinical improvement in patients with severe COVID-19.91 Conestat alfa received FDA approval for treating hereditary angioedema in 2014 and is under development for treating COVID-19.

A phase 2 study to evaluate the ability of conestat alfa to prevent severe disease progression in hospitalized COVID-19 patients has been completed (NCT04530136); results are pending. Participants in the conestat alfa to group received an intravenous injection of a dose of 50 U/kg (maximum dose of 4,200 U) every 12 hours for 4 days, for a total 8 injections. The control group received a standard of care. The primary outcome measure was the score of the WHO Ordinal Scale of disease severity on day 7.

MN-166 (ibudilast)

Ibudilast is an allosteric inhibitor developed by Kyorin Pharmaceutical, Tokyo, Japan.92 Ibudilast is a selective inhibitor of phosphodiesterases acts to macrophage function. PDE 3, 4, and 10.93 The use of PDE4 inhibitors has the potential to prevent cytokine storm in COVID-19.94 Additionally, the inhibitor can inhibit production of proinflammatory cytokines such as IL-1β, TNF, and IL-6, while increasing the production of anti-inflammatory cytokines (IL-10, IL-4).93

Ibudilast was approved by Japan in 1989 for treating bronchial asthma and improving dizziness secondary to chronic cerebral circulation impairment associated with sequelae of cerebral infarction.

Currently, ibudilast is being repurposed for use in the treatment of COVID-19. A phase 2 study to evaluate the efficacy and safety of ibudilast in COVID-19 patients is underway (NCT04429555). Participants receive MN-166 or placebo capsules twice daily for 7 days. Primary outcome measures are the proportion of subjects free from respiratory failure, the mean change from baseline in clinical status using the NIAID 8-point ordinal scale, the percentage of patients showing improvement in clinical status, and changes in cytokine levels at day 7.

CAP-1002

CAP-1002 refers to cardiosphere-derived cells (CDCs), which are stromal progenitor cells, derived from heart tissues by Capricor Therapeutics (Beverly Hills, CA, USA).95 Cardiospheres are cell clusters that form invitro when cardiac progenitor cells are cultured. CDCs modulate activity of macrophages and effector T cells, and affect expression pro-inflammatory cytokines, potential o contributing to the pathogenesis of COVID-19. Additionally, CDCs can decrease production of proinflammatory cytokines (IFNγ, TNF, IL-1β, IL-6) and increase plasma levels of IL-10.96

CAP-1002 has been granted orphan drug designation by the FDA for treating Duchenne muscular dystrophy. CAP-1002 is being repurposed to treat COVID-19. A phase 2 study to evaluate the safety and efficacy of CAP-1002 in COVID-19 patients has been completed (NCT04623671). The treatment group received CAP-1002 therapy by infusion of 150 million CDCs in 5% human serum albumin. The control group received a placebo infusion. The primary outcome measure was the number of deaths within 90 days. Results are pending.

GB-0139 (Nafomostat)

GB-0139 is an inhibitor of galectin-3 (Gal-3) developed by Galecto Biotech (Boston, MA, USA). Gal-3 activates signaling pathways, including in particular those involving three molecules: extra-cellular signal regulated kinase (ERK), protein kinase B (Akt), and JAK/signal transducer and activator of transcription (STAT1). Activation of pathways leads to the release of pro-inflammatory cytokines, including IL-6, IL-8, and TNF.

Nafomostat is a synthetic serine protease inhibitor that reduces plasma levels several pro-fibrotic mediators.97 GB-0139 being developed as treatment for idiopathic pulmonary fibrosis and for COVID-19.98 GB-0139 might reduce viral-induced lung injury in patients with COVID-19

A phase 1/2 study to evaluate nafomostat as a therapy for COVID-19 to mitigate lung damage in patients with COVID-19 is currently recruiting participants (NCT04473053). Subjects are randomized into a nafamostat group, a GB-0139 group, and a control group. The nafamostat group will receive nafamostat via continuous intravenous infusion at 0.2 mg/kg/hr for 7 days. The GB-0139 group will receive 10 mg inhaled GB-0139 twice daily for 2 days and then 10 mg once daily for the remaining 12 days. The control group will receive standard of care. Primary outcomes will be changes of vital signs (blood pressure, heart rate, temperature and respiratory rate) up to 16 days following GB-0139 administration.

Remestemcel-L

Remestemcel-L refers to mesenchymal stromal cells (MSCs) derived from the bone marrow of healthy donors. Cells are prepared by Mesoblast (Melbourne, Australia). MSCs prevent T lymphocytes from entering the cell cycle, thus inhibiting clonal expansion. Additionally, MSCs promote the secretion of IL-4, impeding the secretion of IFN-γ and enhancing the development of regulatory T lymphocytes.99 Remestemcel-L is currently under development as a drug for treating acute (GVHD), inflammatory bowel disease, and ARDS caused by COVID-19 infection.100

A pilot study showed that remestemcel-L improved clinical outcomes of COVID-19-associated multisystem inflammatory syndrome in children (MIS-C).101 The primary aim is to achieve 43% reduction in mortality at 30 days in patients with moderate-to-severe acute respiratory distress (ARDS). An expanded access study to evaluate the safety and efficacy of remestemcel-L cells for MIS-C is currently enrolling subjects (NCT04456439), including children and adolescents from 2 months to 17 years of age. Subjects will receive a total of 2 × 106 remestemcel-L over 5 days. Participants will receive an intravenous infusion of diphenhydramine 0.5–1 mg/kg (up to 50 mg) prior to infusion of remestemcel-L. Subjects who are not receiving glucocorticoids for other indications will receive an intravenous infusion of 0.5–1 mg/kg hydrocortisone (up to 50 mg) prior to infusion of remestemcel-L.

Developing immunostimulants in COVID-19

IMM-101

IMM-101 is an immunotherapeutic agent containing heat-killed Mycobacterium obuense that was developed by Immodulon Therapeutics in Uxbridge, UK. IMM-101 is very effective at inducing cytokines expressed by innate immune cells. Recognition of IMM-101 by dendritic cells can enhance antigen processing and presentation capacity, increase expression of co-stimulatory molecules, and promote secretion of anti-viral interferon-γ, perforin, and granzyme B.102 IMM-101 is currently being developed as a drug for treating pancreatic cancer, colorectal cancer and advanced melanoma, and colorectal cancer.103 Additionally, clinical trials are underway to investigate potential use in the treatment of COVID-19. A phase 3 study to evaluate the safety and efficacy of IMM-101 in cancer patients with severe respiratory and COVID-19 infections is enrolling subjects (NCT04442048). The treatment group will receive 1.0 mg IMM-101 on day 0 and 0.5 mg on day 14 and day 45. Subjects in the control group will not receive any active treatment. The primary endpoint is the rate of flu-like illness or confirmed viral/bacterial respiratory infection after treatment.

Sarconeos (BIO101)

The small molecule sarconeos (Ruvembri, BIO101) activates the Mas receptor and triggers the P13K/AKT/mTOR pathway and the AMPK/ACC pathway.104 The drug was developed by Biophytis (Paris, France). It is being developed as a drug for treating sarcopenia.104

Stimulating the Mas receptor may have beneficial effects on respiratory function, arterial oxygenation, and lung tissues in patients with severe COVID-19.105 Investigation is underway to explore the potential use of sarconeos in the treatment of COVID-19. A phase 2, 3 study to evaluate the efficacy and safety of sarconeos for preventing respiratory deterioration in hospitalized COVID-19 patients was terminated due to the lack of eligible patients (NCT04472728). However, Biophytis continues developing sarconeos for COVID-19.106

Developing drugs targeting granulocyte-macrophage colony-stimulating factor (GM-CSF)

GM-CSF belongs to the CSF superfamily. It is mainly produced by lymphocytes and innate lymphoid cells.107 GM-CSF can stimulate mature neutrophils to exhibit chemotaxis, enhance oxidative metabolism, enhance antibody-dependent phagocytosis and killing of microorganisms, and produce various regulatory proteins. GM-CSF deficiency can lead to alveolar proteinosis, caused by failure to clear surfactant and due to a shortage of pulmonary macrophages induced by GM-CSF.108

GM-CSF basal circulating levels are low under homeostatic conditions. Levels quickly increase during infection or inflammation. GM-CSF is produced by a variety of cells, including fibroblasts, macrophages, endothelial cells, T cells, neutrophils, and eosinophils, among others. Expression of GM-CSF can be induced by proinflammatory cytokines, including IL-1α, IL-1β, TNF, and IL-12, whereas expression is suppressed by IL-4, interferon-γ, and IL-10.109 During inflammation, elevated levels of systemic GM-CSF can dramatically increase monocyte and neutrophil production from bone marrow.107

Leukine (sargramostim)

Sargramostim is a GM-CSF manufactured by Partner Therapeutics (Lynnwood, WA, USA).110 It stimulates the division and differentiation of partially committed progenitor cells in granulocyte-macrophage pathways.110 Sargramostim is thought to offer benefits in the early phase of SARS-CoV-2 infection by preserving the function of alveolar macrophages.111

Sargramostim was approved by the FDA for treating acute myeloid leukemia and delayed neutrophil recovery or graft failure after autologous or allogeneic bone marrow transplantation. Investigations are being conducted to repurpose sargramostim for treating COVID-19.

A phase 4 study to evaluate the safety and efficacy of sargramostim in patients with COVID-19 and acute hypoxic respiratory failure has been completed (NCT04326920). The treatment group received 125 mcg inhaled sargramostim twice daily for 5 days in addition to the standard of care. If subjects in the treatment group require mechanical ventilation within 5 days, inhaled Leukine could be replaced by intravenous infusion of 125 mcg/m2 until day 5. After day 6, clinicians could choose to administer Leukine intravenously for an additional 5 days. The control group received standard of care for 5 days. If subjects in the control group required mechanical ventilation, the clinician could decide to administer an intravenous infusion of sargramostim 125 mcg/m2 once daily for 5 days from day 6 onwards. The primary outcome measure was mean improvement in oxygenation on day 6 or hospital discharge. Results are pending.

Plonmarlimab (TJ003234)

Plonmarlimab directly binds GM-CSF, blocking intracellular signaling. Downstream signaling from the GM-CSF receptor involves JAK2/signal transducer and activator of transcription 5 (STAT5), nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), ERK, and the phosphoinositide 3-kinase (PI3K)-Akt (protein kinase B) pathway. Therefore, inhibiting GM-CSF might represent a viable treatment option for preventing the CRS.112

Plonmarlimab is under development for treating CRS and COVID-19. It is manufactured by I-Mab Biopharma based in China.113

Phase 2 and 3 studies to evaluate the safety and efficacy of plonmarlimab (also known as TJ003234) in severe COVID-19 patients have been completed (NCT04341116). In those studies, subjects were randomized to receive an infusions of placebo or plonmarlimab at low, medium or high dose. The primary outcome measure was the proportion of subjects who were alive and free from mechanical ventilation among subjects who were free of mechanical ventilation at baseline for 30 days. Plonmarlimab reduced mortality in the phase 2/3 trial.

Lenzilumab

Lenzilumab is a GM-CSF monoclonal antibody developed by Humanigen based in Burlingame, CA, USA. By binding to GM-CSF, lenzilumab prevent binding to the GM-CSF receptor, thereby alleviating CRS. This GM-CSF neutralization can reduce IL-6, MCP-1, macrophage inflammatory protein 1 alpha (MIP-1α), IP-10, VEGF, and TNFα levels. Consequently, lenzilumab exhibits potential immunomodulatory activity.114 It is indicated for treating refractory chronic myelomonocytic leukemia, acute GvHD, COVID-19, among other indications.115

A phase 2 study, which is the ACTIV-5 Big Effect Trial (BET), sponsored by NIAID, NIH for COVID-19 patients, has been completed (NCT04583969). The experimental group received 200-mg intravenous remdesivir on Day 1, followed by a 100-mg once-daily intravenous maintenance dose up to a 10-day course while hospitalized; this group also received 600-mg intravenous lenzilumab infusion every 8 hours starting on Day 1, for a total of 3 doses. The control group received 200 mg of intravenous remdesivir on Day 1, followed by a 100 mg once-daily intravenous maintenance dose and 600-mg intravenous placebo infusion every 8 hours starting on Day 1. The primary outcome measures were the number of surviving participants and ventilation-free subjects at 29 days.

PART 5. DEVELOPING ANTI-COAGULANTS IN COVID-19

Fig. 4 illustrates drugs under development that can prevent blood coagulation.

Fig. 4
Blood coagulation and anticoagulation pathways and related drugs. Therapies are shown in white boxes.
SYK = spleen tyrosine kinase.

Heparin

Heparin is a factor Xa and thrombin inhibitor. Heparin is isolated from porcine tissues and is available in several forms. These include Lovenox (enoxaparin sodium), by Sanofi Aventis, France,116 Fragmin (dalteparin sodium) by Pfizer, USA,117 and Innohep (tinzaparin sodium) by Leo Pharmaceutical, Denmark.118 Routes of administration are subcutaneous and intravenous injections. Heparin acts by inhibiting factor Xa and thrombin, which promotes blood coagulation.119 Many COVID-19 patients manifest coagulopathies, venous thromboembolism (VTE).120

The ACTIV-4A clinical trial comparing anticoagulation treatment strategies has been completed (NCT04505774). This trial tested four approaches: therapeutic and prophylactic doses of heparin, therapeutic or prophylactic doses of heparin combined with a P2Y12 inhibitor, and the standard of care with crizanlizumab (used for veno-occlusive disease). The primary outcome measure was the number of days that a patient was alive and free of organ support through the first 21 days after trial entry. Results are pending.

Xarelto (rivaroxaban)

Rivaroxaban is a factor Xa inhibitor developed by Bayer, Leverkeusen, Germany.121 It received FDA approval for treating deep vein thrombosis (DVT), pulmonary embolism (PE), and VTE. Its mechanism of action is by inhibiting factor Xa to suppress thrombin formation and the coagulation pathway.122 Xarelto is expected to reduce thrombotic disease progression in severe COVID-19 patients.123

A phase 3 study of rivaroxaban to reduce clinical progression of COVID-19 is currently active, in a not recruiting status (NCT04324463). In that study, the treatment group will be receive 2.5 mg oral tablets of Rivaroxaban twice daily for 28 days and the control group will receive the standard of care. The primary outcome measure is the time to the first occurrence of specified outcomes within the first 45 days after randomization; these outcomes include 1) requirement for high flow oxygen or mechanical ventilation and 2) major thrombosis or death.

Anpocogin (rNAPc2 [recombinant Nematode Anticoagulant Protein c2])

rNAPc2 is a factor Xa inhibitor developed by ARCA Biopharma, Westminster, CO. It is administered by subcutaneous injection.124 Anpocogin binds factor Xa to form the FXa-rNAPc2 complex. This complex inhibits tissue factor-factor VIIa (TF/FVIIa), which is necessary for blood coagulation.125 Therefore, rNAPc2 drug inhibits the extrinsic pathway. It may be effective in the treatment of COVID-19-induced blood coagulopathy.

A phase 2, 3 study of rNAPc2 compared to heparin, for treating hospitalized COVID-19 patients with high D-dimer levels has been completed (NCT04655586). The higher dose, group was given 7.5 μg/kg rNAPc2 via subcutaneous injection on Day 1 and 5 μg/kg via subcutaneous administration on Days 3 and 5. The lower dose group was given 5 ug/kg rNAPc2 via subcutaneous injection on Day 1 and 3 ug/kg via subcutaneous administration on Days 3 and 5. The control group was given standard-of-care heparin following local institutional guidelines (therapeutic or prophylactic regimen). Primary outcome measures were proportional change in D-dimer level from baseline up to Day 8 (phase 2), the number of major or non-major clinically relevant bleeding events within 8 days compared to heparin (phase 2, 3), and the time to recovery within 30 days (phase 3). Treatment with rNAPc2 was well tolerated without excess bleeding or serious adverse events. There was no significant reduction in the primary efficacy end point, D-dimer levels, from baseline to day 8 with rNAPc2 compared with heparin.126

Tavalisse (fostamatinib disodium hexahydrate)

Tavalisse is a tyrosine kinase inhibitor that blocks enzymatic activity of spleen tyrosine kinase (SYK). The drug was developed by Rigel Pharmaceuticals, South San Francisco.127 Fostamatinib received FDA approval for chronic immune thrombocytopenia in 2018. The route of administration is oral supplementation.127 The major metabolite of fostamatinib is R406.

Fostamatinib inhibits signal transduction of B-cell receptor and Fc-activating receptors which bind to antibodies; the drug activates kinases,including SYK.127, 128 Activated SYK stimulates the the inflammatory pathway that promotes the release of neutrophil extracellular traps (NETs). High levels of NETs induce immunothrombosis in COVID-19 patients.129 Fostamatinib also suppresses the activation of platelets induced by COVID-19 plasma. Therefore, fostamatinib might be effective prophylaxis for COVID-19 patients at high thrombotic risk.130

A phase 2, 3 (ACTIV-4) trial (NCT04924660 is randomizing subjects to either of two angiotensin antagonists, TXA127 or TRV027, fostamatinib, or placebo. The fostamatinib arm will receive 100-150 mg oral fostamatinib twice daily for 14 days (28 doses). The primary outcome measure is the number of oxygen-free days up to 28 days following randomization.

Defitelio (defibrotide sodium)

Defitelio enhances enzymatic activity of plasmin to hydrolyze fibrin clots. It was developed by Jazz Pharmaceuticals, Ireland. It received FDA approval for treating hepatic veno-occlusive disease on 2016. The route of administration is intravenous infusion.131 Its mechanism of action is to suppress synthesis of proinflammatory cytokines such as IL-6, inhibiting generation of reactive oxygen species, and to restore nitric oxide generation. Defibrotide sodium has other mechanisms which involve anti-inflammatory and antioxidant pathways. Therefore, defibrotide sodium could be beneficial for treating COVID-19 patients with stimulated platelet aggregation and endothelial cell damage by increasing the expression of pro-coagulant factors.132

A phase 2 study to evaluate the efficacy and safety of defibrotide in COVID-19-related pneumonia is recruiting participants (NCT04652115). Cohort 1 consists of hospitalized patients with COVID-19 pneumonia needing supplemental oxygen or mechanical ventilation without use of anticoagulants who require no more than one vasopressor. Cohort 2 consists of hospitalized COVID-19 pneumonia patients in an intensive care unit (ICU) who are at elevated risk of hemorrhage and/or hypotension. The former is defined as a requirement for therapeutic dose anticoagulation for active thrombosis, ECMO, or continuous renal replacement therapy and the latter is defined as a requirement for two vasopressive agents to maintain hemodynamic stability). Each subject will receive an FDA-approved dose of 6.25 mg/kg defibrotide via intravenous infusion every 6 hours for 21 days.

For the cohort 2 study, a 6+6 dose de-escalation design will be applied. If 2 of 6 dose-limiting toxicities (DLTs) (grade 3/4 hemorrhage and significant new hypotension) are experienced in the first 6 subjects, subjects will receive a reduced dose of defibrotide at 10 mg/kg/d CIVI. If there are 0 or 1 DLTs in the first six subjects at the FDA-approved dose, another six subjects will be enrolled at the same dose. The primary outcome measure is the rate of a special adverse event such as bleeding or hypotension within 21 days. Results are pending.

PART 6. DEVELOPING DRUGS FOR COVID-19-INDUCED NEUROPATHY

Cerebrolysin (FPF-1070)

Cerebrolysin is a peptide-based analog of brain-derived neurotrophic factors and developed by EVER Neuro Pharma (Unterach, Austria). It is approved for treatment of stroke and dementia in 44 countries, including Austria, China, Germany, Russia, and South Korea. Cerebrolysin increases the growth of new neurons, supports existing neurons, and protects neurons against amyloid beta toxicity.133 Due to its neurotrophic and neuroprotective properties, cerebrolysin might be useful to treat post-COVID patients with anosmia or ageusia (NCT04830943).

A phase 4 pilot study of cerebrolysin for COVID-19-induced anosmia and ageusia has been completed (NCT04830943). The cerebrolysin group received 5 mL cerebrolysin (215.2 mg/mL cerebrolysin) via intramuscular injection once daily, five times per week. The treatment lasted at least 8 weeks (total of 40 injections) and a maximum of 24 weeks. The control group received olfactory training twice daily for 8 weeks. Primary outcome measures were a smell and taste questionnaire and odor, taste, and flavor identification testing. Results are pending.

NA-831 (traneurocin)

Transneurocin is an oral neuroprotective drug developed by Biomed Industries (San Jose, CA). It was originally studied for Alzheimer disease. Now it is being developed for treating COVID-19-induced neuropathy. Transneurocin is currently known to enhance neurogenesis. More detailed mechanisms are being investigated. This drug is being studied for use with other antiviral and anti-inflammatory drugs for COVID-19 patients.134, 135, 136

A phase 2, 3 study to evaluate four Traneurocin combinations for COVID-19 infection hospitalized participants is recruiting participants (NCT04452565). Arm 1 will receive 30 mg oral Traneurocin twice on day 1 and then will receive drug once a day from day 2 to day 5. Arm 2 will have oral administration of 60 mg Traneurocin and 400 mg atazanavir, twice on day 1 and then 30 mg and 200 mg respectively, once a day from day 2 to day 5. Arm 3 will be orally administered Traneurocin and dexamethasone (60 mg and 8 mg respectively) twice on day 1 and then 30 mg and 4 mg, once a day from day 2 to day 5. Arm 4 subjects receive oral Atazanavir and dexamethasone (400 mg and 8 mg respectively) twice on day 1 and 200 mg and 4 mg once a day from day 2 to day 5. The primary outcome measure will be the time to neurologic recovery, assessed up to 36 days after treatment initiation.

PART 7. OTHER MECHANISM DRUG FOR COVID-19 THERAPEUTICS UNDER DEVELOPMENT

Drugs targeting the hypoxia-inducible factor (HIF)-prolyl hydroxylase (PHD) (HIF-PHD) pathway

Tissue hypoxia is a common feature at sites of immunity and inflammation, resulting in elevated expression and activity of HIF. In response to hypoxia, specific cells increase their production of erythropoietin through activation of HIF signaling. This process counteracts a systemic decrease in pO2 and increases the oxygen-carrying capacity of the blood through amplified erythrocyte production.137 HIF is the most well-characterized substrate of HIF prolyl hydroxylase (HIF-PHD). HIF has two subunits: an α-subunit that quickly degrades in normoxia and a stable β-subunit. The HIF α-subunit is regulated by post-transcriptional modifications such as phosphorylation, acetylation, and hydroxylation. Hydroxylation (the main regulator of HIFα) is controlled by HIF-PHD belonging to the family of α-ketoglutarate-dependent non-heme iron dioxygenases.138

Under normoxic conditions, HIF α-subunits are hydroxylated on proline residues by three distinct enzymes (PHD1, PHD2 and PHD3), leading to HIFα ubiquitination and subsequent degradation. Hydroxylation of HIF inhibits binding of a transcriptional co-activator to prevent formation of a functional transcriptional complex.137 Under hypoxic conditions, PHD enzymatic activity is reduced and consequently, HIF-α is degraded at lower rate. HIF-α can then be translocated to the nucleus, where it heterodimerizes with HIF-β and activates gene transcription. As a result, HIF facilitates oxygen delivery and cellular adaptation to hypoxia by regulating cellular processes such as angiogenesis, mitochondrial biogenesis, anaerobic glucose metabolism, and others.139 Fig. 5 illustrates drugs that are currently under development modulating the HIF-PHD pathway.

Fig. 5
Hypoxia-inducible factor-prolyl hydroxylase (HIF-PHD) pathway and its related drugs.
HIF = hypoxia-inducible factor, PHD = prolyl hydroxylase, VEGF = vascular endothelial growth factor.

Avastin (bevacizumab)

Bevacizumab was developed by Roche in Basel, Switzerland.140 This monoclonal antibody recognizes vascular endothelial growth factor-A (VEGF-A) and prevents the interaction of VEGF-A with the VEGF receptor. Consequentially, bevacizumab inhibits the activation of the VEGF signaling pathways and represses angiogenesis.141 In patients with COVID-19, ARDS induces tissue hypoxia, which leads to increased VEGF expression through activation of the HIF PHD-1 pathway. In these patients, VEGF contributes to plasma extravasation and pulmonary edema, further exacerbating tissue hypoxia and lung inflammation. Therefore, blocking VEGF with Avastin (bevacizumab) can improve oxygen perfusion and elicit anti-inflammatory response in patients with COVID-19.142

Avastin (bevacizumab) received FDA approval in 2004 for treating metastatic colorectal cancer. Currently, it is being repurposed for treating COVID-19. A phase 3 study to evaluate the efficacy and safety of bevacizumab for patients with COVID-19 is recruiting participants (NCT04305106). The bevacizumab group will receive a single 7.5 mg/kg dose of bevacizumab via intravenous infusion with a standard of care if necessary. The control group will receive a single dose of placebo (inactive excipient) at 7.5 mg/kg via intravenous infusion, together with a standard of care. The primary outcome measures are the time to achieve a clinical improvement of two points on a seven-category ordinal scale and discharge from the hospital within 28 days.

Vadadustat (AKB-6548)

Vadadustat is a structural analog of α-ketoglutarate, a Krebs cycle intermediate that inhibits HIF-PHD. Vladadustat was developed by Akebia Therapeutics (Cambridge, MA, USA).143 The drug inhibits PHD catalysis and stabilizes HIF-2α to a greater extent than does HIF-1α. Vadadustat increases hemoglobin levels, increases total iron binding capacity, and lowers plasma ferritin and hepcidin levels. Vadadustat was developed for treating anemia due to chronic kidney disease.143 Currently, it is being studied for its potential application in the treatment of COVID-19-associated hypoxemia.139

A phase 2 study of vadadustat for preventing ARDS in COVID-19 patients is currently active but recruitment has not begun (NCT04478071). Subjects will be randomized to receive 900 mg vadadustat or placebo, taken orally once daily for 14 days. The primary outcome measure will be the number of subjects who are classified as 8 (deceased), 7 (hospitalized, on invasive mechanical ventilation or ECMO), or 6 (hospitalized, on non-invasive ventilation or high flow oxygen devices) by the NIAID ordinal scale on day 14.144

Trimodulin (BT588)

Trimodulin is an immunoglobulin concentrate containing IgG (~56%), IgA (~21%), and IgM (~23%). It was developed by Biotest, Germany. Initially, trimodulin was developed to treat patients with severe community-acquired pneumonia (sCAP). Trimodulint is currently under clinical development for treating patients with either sCAP or severe COVID-19.145

As immunoglobulin preparations have long been used as therapy for inflammatory diseases, the anti-inflammatory effects of trimodulin might be beneficial for patients with severe COVID-19.146

A phase 3 study to evaluate the efficacy and safety of trimodulin as an adjunctive treatment together with standard of care for patients with COVID-19 is recruiting participants (NCT05531149). Subjects will receive trimodulin or human albumin 1% (placebo) via intravenous infusion for five consecutive days. Primary outcome measures will be percentage of clinical deterioration rate and the percentage of mortality rate between days 6 to 29.

Octagam

Octagam is an intravenous human immune globulin preparation that supplies both opsonic and neutralizing IgG antibodies. It was developed by Octapharma in Lachen, Switzerland. The name derives from the company’s expertise in developing factor 8 preparations to treat hemophilia).147 Intravenous immunoglobulin (IVIG) enhances pro-inflammatory cell activation and suppresses T cell and B cell proliferation. As severe cytokine storm is associated with high death rates in severe COVID-19, use of IVIG might be effective.148 Octagam received FDA approval for treating primary humoral immunodeficiency in 2004.147 It is currently being repurposed for use in the treatment of COVID-19.

A phase 2 study to evaluate the safety and efficacy of Octagam to reduce all-cause mortality in COVID-19 has been completed (NCT04480424). The treatment groups received intravenous infusions of Octagam (500 mg/kg for 4 days or 400 mg/kg for 5 days), together with standard of care. The control group received a standard of care. The primary outcome measure was all-cause mortality rate up to 29 days. Octagam treatment was associated with improved functional status,

Opaganib

Opaganib is an oral sphingosine kinase 2 (SK2) inhibitor developed by RedHill Biopharma (Tel Aviv, Israel). Opaganib inhibits host SK2 but not viral SK2. Thus, it is less affected by viral mutations.149 This drug is being studied for treating cancer and infectious diseases including COVID-19. Its mechanism of action is through inhibiting production of sphingosine 1-phosphate (S1P). S1P promotes TNF signaling, which promotes cytokine production. Opaganib also inhibits dihydroceramide desaturase (DES1) and glucosylceramide (GCS). DES1 inhibition promotes autophagy to inhibit viral replication, while GCS inhibition can also reduce the viral entry into cells. Hence, opaganib might be effective for treating COVID-19 through multiple mechanisms.150

A phase 2/3 study to evaluate the efficacy and safety of opaganib in hospitalized patients with severe COVID-19 has been completed (NCT04467840). The opaganib group received 500 mg opaganib orally every 12 hours with the standard of care. The medication can be administered as a liquid suspension via nasogastric tube. The control group received a placebo orally every 12 hours, together with standard of care. All participants received drug or placebo for 14 days. The primary outcome measure was comparing the proportion of patients no longer requiring supplemental oxygen for at least 24 hours in a time frame of 14 days. In a phase 2/3 study of 475 patients, treatment with opaganib was associated with a 62% decrease mortality in patients with moderate to severe COVID-19.

ARADKOA (Tafenoquine)

Tafenoquine is a potential TMPRSS2 and Mpro inhibitor developed by 60 Degrees Pharmaceuticals (Washington, D.C., USA). It received FDA approval for prophylaxis of malaria. It inhibits hematin polymerization and induce parasite death.151 It has potential to inhibit TMPRSS2 and Mpro. Inhibiting TMPRSS2 can reduce virus entry. Mpro is involved in the correct cleavage of viral polyproteins translated from the viral genome. Inhibiting Mpro can reduce viral replication.152 Therefore, tafenoquine may be effective in the treatment of COVID-19.

A phase 2 study of tafenoquine treatment in patients with mild to moderate COVID-19 been completed (NCT04533347). Subjects took two 100 mg tafenoquine tablets or two placebo tablets orally for 10 days. The primary outcome measure was the proportion of patients with clinical recovery of COVID-19 symptoms on Day 14. Results are pending.

Luvox (fluvoxamine)

Fluvoxamine is an inhalant selective serotonin reuptake inhibitor developed by Solvay in Houston, TX. It received FDA approval for treating obsessive-compulsive disorders in 1994.153 Its mechanisms of action are diverse and include reducing histamine release from mast cells, showing lysosomotrophic effects (i.e., increasing lysosomal pH) by interfering with viral trafficking, interfering with lysosomal membrane binding of acid sphingomyelinase, affecting inositol-requiring enzyme 1α-mediated inflammation, and increasing melatonin levels.154 Due to these anti-inflammatory and antiviral properties, Fluvoxamine might be effective for treating COVID-19.

A phase 3, ACTIV-6 trial to evaluate the effectiveness of repurposed medications in COVID-19 patients is recruiting participants (NCT04885530). This study consists of six subgroups (fluvoxamine and ivermectin will each have two subgroups with different doses, together with two placebo group. Each subgroup participant will receive either a study drug (fluvoxamine, ivermectin, fluticasone, montelukast) or placebo. Fluvoxamine subgroups are ‘arm B’ and ‘arm E’. Arm B participants will self-administer 50 mg fluvoxamine or placebo orally twice a day for 10 days. Arm E participants will orally self-administer 50 mg fluvoxamine or placebo twice on day 1, and 100 mg fluvoxamine or placebo twice a day for 12 days. Primary outcome measures are number of hospitalizations, deaths and symptoms reported by subjects, all within a time frame of up to 28 days.

Pyramax (artesunate/pyronaridine)

Pyramax is an oral antimalarial drug repurposed for COVID-19 by Shin Poong Pharmaceutical, Seoul, South Korea. Pyronaridine inhibits hematin formation to protect the heme group that is toxic to malaria. Artesunate produces free radicals with antimalarial activity.155, 156 In treating COVID-19, artesunate can act as a viral infection inhibitor at the cellular post-entry level.157 Pyronaridine modulates appropriate levels of antiviral interferons, including interferon-1β. Moreover, pyronaridine can lower high levels of CXCL1 and CCL3 involved in inflammation responses.158 Therefore, Pyramax has potential as an effective COVID-19 treatment. A phase 3 study to evaluate the efficacy and safety of Pyramax in COVID-19 patients is currently recruiting participants (NCT05084911). The Pyramax group will receive a pyronaridine-artesunate (180/60 mg) tablet for three days and the control group will receive a placebo. The primary outcome measure will be the proportion of patients who require hospitalization or have died due to COVID-19 infection up to 29 days.

RVX-208 (Apabetalone)

Apabetalone is an inhibitor of bromodomain and extra-terminal (BET) proteins. It specifically targets bromodomain 2 (BD2). It was shown by Resverlogix (Calgary, Aberta, Canada)159 to be a potential treatment for cardiovascular disease, pulmonary arterial hypertension, chronic kidney disease, and COVID-19. It was developed with a cardiovascular indication and was granted a Breakthrough Therapy Designation by the FDA.160

The mechanism of action is to prevent interactions between acetylated lysine on histone tails and transcription factors. In particular, apabetalone can suppress gene expression of vascular inflammation mediators whose genes are dependent for transcription on BET domain and extra terminal domain protein (BET) proteins. Apabetalone prevents BRD4 associations with key transcriptional enhancers and promoters.159

In addition, ACE2 expression is regulated by BET proteins. Inhibitors of BET proteins downregulate ACE2 expression and limit SARS-CoV-2 replication in vitro.160 Due to its antiviral and anti-inflammatory effects, apabetalone is a promising candidate for treating COVID-19.161

A phase 2, 3 study to evaluate the safety and efficacy of apabetalone in COVID-19 patients is recruiting participants (NCT04894266). The treatment group will receive 100 mg apabetalone orally twice daily together with standard of care, and the control group will receive standard of care only. The primary outcome measure is the WHO Ordinal Scale for Clinical Improvement change on day 14.

ANA001 (Niclosamide)

Niclosamide is an inhaled preparation of salicylanilide, a derivative of salicylic acid belonging to a large group of lipophilic compounds. Niclosamide was developed by NeuroBo Pharmaceuticals, Cambridge, MA, USA.162, 163 Plausible mechanisms of action of niclosamide against COVID-19 involve four cellular processes: 1) endosomal pH neutralization to prevent viral replication, 2) promotion of autophagy through inhibition of S-phase kinase-associated protein 2 (SKP2), 3) decreased mucus plugging through inhibition of TMEM16A (calcium activated chloride channel, CaCC), and 4) prevention of syncytia formation by ion channel inhibition.163

Additionally, niclosamide has anti-inflammatory effects by inhibiting pro-inflammatory cytokines, mainly TNF. Furthermore, it can inhibit the progression of inflammatory airway diseases by suppressing the release of IL-8 and intracellular Ca+2 signaling.164 Due to these anti-inflammatory and antiviral effects, it may be effective for treating patients with COVID-19.

A phase 4 study of niclosamide to treat patients with COVID-19 has been completed (NCT05087381) in Thailand. One treatment group received 500 mg niclosamide orally by tablet twice daily for 14 days. A second treatment group received the same dose for 14 days but in combination with one 8 mg tablet of bromhexine twice daily for 10 days. The control group received supportive care. Primary outcome measures were hospital admission or mortality related to COVID-19 within 28 days, the time taken to self-report recovery until the final day of participation, and the progression to severe COVID-19 disease until the final study day. Study results are not available.

(pamapimod/pioglitazone)

KIN001 is a combination of pamapimod, a selective inhibitor of p38 mitogen-activated protein kinase alpha (p38 MAPKα), and pioglitazone, a peroxisome proliferator-activated receptor γ (PPARγ) agonist. The drug was developed by Kinarus Therapeutics in Switzerland for treating wet age-related macular degeneration (Wet-AMD) and idiopathic pulmonary fibrosis. The route of administration is oral inhalation.165

The p38 MAPK pathway plays a crucial role in viral infection. Viral p38 MAPK activation can promote endocytosis of viral receptors, including ACE2. Downstream effectors of this pathway include transcription factors and RNA binding proteins that regulate cytokine production, cell proliferation, cell differentiation, and apoptosis, as well as development. PPARs are transcription factors involved in insulin responses. Studies have suggested that pioglitazone, a glucose-lowering medication, can improve manifestations of COVID-19 patients with type 2 diabetes. Therefore, KIN001, the combination of a p38 MAPK inhibitor with pioglitazone, might have synergistic antiviral activity against SARS-CoV-2 with anti-inflammatory effects.166

A phase 2 study to evaluate the efficacy and safety of KIN001 (pamapimod/pioglitazone) in non-hospitalized COVID-19 patients is recruiting participants (NCT05659459). The treatment group will receive 75 mg pamapimod and 5 mg pioglitazone orally twice daily for 14 days. The control group will receive matching tablets of KIN001 orally twice daily for 14 days. The primary outcome measure is the evaluation of COVID-19 symptoms within 28 days.

Xpovio (selinexor)

Selinexor is an oral exportin 1 (XPO1) blocker developed by Karyopharm Therapeutics, Newton, MA. In 2019, selinexor received FDA approval for treating multiple myeloma and diffuse large B-cell lymphoma.167 In COVID-19, XPO1 inhibitors can transiently inhibit expression of ACE2 and its interaction with SARS-CoV-2. Selinexor also inhibits transport of the tumor suppressor protein p53. This results in nuclear accumulation of p53, and induction of apoptosis and cell-cycle arrest. Therefore, inhibition of the p53 pathway might induce apoptosis of SARS-CoV-2. Moreover, selinexor prevents release of pro-inflammatory cytokines and reduce oxidative and inflammatory disorders by activating nuclear factor erythroid 2-related factor-2 (Nrf2) and peroxisome proliferator-activated receptor (PPAR)-γ.168

A phase 2 study to evaluate the efficacy and safety of selinexor (KPT-330) in patients with severe COVID-19 has been completed (NCT04349098). In that study, subjects received standard of care together with 20 mg of oral selinexor or a matching placebo on Days 1, 3, and 5 of each week for up to two weeks. If subjects could tolerate the therapy and experienced clinical benefit, dosing could continue for an additional 2 weeks (28 days). The primary outcome measure was the proportion of subjects showing clinical improvement of at least two points on the 8-point ordinal scale within 14 days. Selinexor did not reduce mortality, reduce ICU admission rate or recovery time, compared to placebo.

Alinia (Nitazoxanide)

Nitazoxanide is an oral medication consisting of a nitrothiazole moiety and salicylamide moiety. It was developed by Romark Laboratories (Tampa, FL, USA).169 Upon administration, the drug is rapidly deacetylated to tizoxanide (TIZ), its active metabolite.170 Alinia received FDA approval in 2002 for treating diarrhea caused by Giardia lamblia or Cryptosporidium parvum as an anti-parasitic drug with potent antiviral activity.

Nitazoxanide’s mechanism of action is to inhibit viral entry and viral replication, and to suppress pro-inflammatory responses. Nitazoxanide inhibits ACE2 receptor/TMPRSS2 interactions by inhibiting protein disulfide isomerase. It also inhibits SARS-CoV-2 fusion and entry by suppressing intracellular signaling, including via MAPK/ERK, PI3K/Akt/mTOR, and Wnt/β-catenin.

Further, nitazoxanide inhibits viral replication by inhibiting enzymatic activity of the 3-chymotrypsin-like protease (3cLpro) and (papain-like proteae (pLpro). Nitazoxanide also stimulates immune responses of host cells by activating Type-I interferon secretion and curbing proinflammatory responses. Due to these mechanisms, nitazoxanide offers potential for repurposing as therapy for COVID-19.170

A phase 3 study to evaluate the efficacy and safety of nitazoxanide in patients with non-critical COVID-19 illness is recruiting participants (NCT04423861). The treatment group will receive a 600 mg tablet of nitazoxanide twice daily for 7 days and the control group will receive placebo twice daily for 7 days. The primary outcome measure will be the need for mechanical ventilation by evaluation of change in acute respiratory syndrome up to 14 days.

VirX/enovid (Nitric oxide nasal spray)

Nitric oxide nasal spray of SaNOtize is approved as a medical device in Thailand, Singapore, Hong Kong, Nepal, South Africa, Germany (branded as VirX), Israel, Indonesia and Bahrain (branded as Enovid). The drug releases nitric oxide.171 Tandon et al.172 have reported that NO has immediate virucidal action against SARS-CoV-2 and other respiratory pathogens. Mechanistically, NO can quickly alter structural integrity of viral proteins, including spike protein and viral protease, through reduced palmitoylation and reduced nitrosylation. In addition, NO is a systemic vasodilator endogenously produced in the endothelium. Inhaled NO induces bronchodilation, thereby improving oxygen delivery to the alveoli. Studies have indicated that NO as a therapeutic agent improves ventilation perfusion in patients with severe ARDS associated with COVID-19.172, 173

A phase 3 study to evaluate the efficacy and safety of nitric oxide nasal spray (NONS) for preventing COVID-19 infection is ongoing (NCT05109611). The treatment group will receive a nasal spray with nitric oxide-releasing solution up to 3 times daily for 28 days. The control group will receive a nasal spray with isotonic saline up to 3 times daily for 28 days. The primary outcome measure is assessing whether subjects were infected with SARS-CoV-2 through a COVID-19 test within 28 days.

Hesperco (hesperidin)

Hesperidin is a bioflavonoid formulation with antioxidant properties that was developed by Valeo Pharma in Kirkland, Quebec, Canada.174 The drug received a Natural Product License approval from Health Canada, authorizing its sale in Canada. Hesperidin is a potential medicine against COVID-19. Some studies have suggested that hesperidin may disrupt the interaction of ACE2 with the viral RBD. Hesperidin also has potent antioxidant properties that make it serve as an effective agent against superoxide and hydroxyl radicals. It also inhibits nitric oxide production. Therefore, Hesperidin might contribute to strategies for the prevention of COVID-19.175

A phase 2 study to evaluate the efficacy and safety of hesperidin in COVID-19 patients has been completed. Subjects were randomized to receive 1,000 mg hesperidin or matching placebo capsules once daily for 14 days. The primary outcome measure was the number of subjects with COVID-19 symptoms (fever, cough, shortness of breath, or anosmia) on days 3, 7, 10, and 14. Results are pending.

Lipitor (atorvastatin calcium)

Atorvostatin is an oral, selective, competitor inhibitor of 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) reductase developed by Viatris in Canonsburg, PA. It received FDA approval as an adjunct to reduce low-density lipoprotein.176 HMG-CoA reductase inhibitors (statins) and have anti-inflammatory and antithrombotic effects. Statins reduce inflammation by inhibiting the NF-kB pathway and reduce the risk of thrombotic events through its profibrinolytic activities. Therefore, statin treatment might alleviate inflammation and improve respiratory status in patients with COVID-19.177

A phase 3 study to evaluate the effects of atorvastatin in Long COVID-19 neurological symptoms patients is recruiting participants (NCT04904536). The treatment group will receive six months of daily oral atorvastatin (40 mg), together with standard of care for 18 months. The control group will receive standard of care for 18 months. The primary outcome measure will be subject speed on the oral Symbol Digit Modalities Test after 18 months.

RA 101495 (Zilucoplan)

Zilucoplan is a macrocyclic peptide that binds complement factor C5 and allosterically inhibits cleavage of C5 into C5a and C5b. It was developed by Union Chemique Belgium (UCB)178 for treating generalized myasthenia gravis and paroxysmal nocturnal hemoglobinurias.179 Its mechanism of action is by suppressing inflammation and coagulation. The anaphylatoxin C5a attracts inflammatory cells into tissues. C5b can induce further drive the complement cascade reaction by promoting assembly of the membrane attack complex, which kills damaged cells and opsonized pathogens. C5b can also trigger the formation of microthrombi on endothelial cells.

De Leeuw et al.180 have shown that increased complement system activation is related to a worse clinical outcomes in COVID-19 patients. As a result, complement blockade has emerged as a potential therapy for COVID-19. Zilucoplan may benefit COVID-19 patients with respiratory failure and signs of systemic inflammation.180

A phase 3 study to evaluate the efficacy and safety of three candidate COVID-19 drugs (Apremilast, Lanadelumab, Zilucoplan) in COVID-19 patients has been completed (NCT04590586). In that study, subjects were randomized between candidate drug or placebo with standard of care. The zilucoplan treatment group received standard of care and a subcutaneous injection of 32.4 mg zilucoplan once daily until discharge or up to 14 days. The zilucoplan control group received standard of care and a subcutaneous injection of zilucoplan matching placebo once daily until discharge or up to 14 days. The primary outcome measure was the time to discharge without re-hospitalization before day 29. This study was terminated without statistical analysis.

Vilobelimab

Vilobelimab is a chimeric monoclonal IgG4 antibody that binds human complement split product, factor C5a. The drug was developed by InflaRx in Jena Germany181 for treating pyoderma gangrenosum, cutaneous squamous cell carcinoma, and critical COVID-19.182 The antibody acts by suppressing inflammation and coagulation. The potent anaphylatoxin C5a recruits monocytes and neutrophils to infection sites and activates these cells, causing tissue damage by enzyme release and free radical formation. C5a also induces the release of tissue factor from endothelial cells and neutrophils, which can activate the coagulation system. Therefore, C5a inhibition can inhibit organ damage induced by neutrophils and improve microangiopathy and microthrombosis.181

A phase 2, 3 study of vilobelimab (IFX-1) to treat severe COVID-19 pneumonia has been completed (NCT04333420). Subjects received 800 mg vilobelimab or placebo via intravenous infusion for a maximum of six doses (days 1, 2, 4, 8, 15, and 22) with standard of care.183 The primary outcome measure was mortality within 28 days. Results are pending.

Lyfaquin (centhaquine)

Centhaquine is an α-2B adrenergic receptor agonist developed by Pharmazz (Willobroodk, IL, USA). Additionally, centhaquine enhances tissue blood perfusion by stimulating central α-2A adrenergic receptors.184 As a result, centhaquine improves tissue perfusion and oxygenation in patients with ARDS.185

The drug is approved in India and indicated as a resuscitative agent for treating patients with hypovolemic shock.184 Currently, research is being conducted to repurpose centhaquine as a treatment for COVID-19 patients.

A phase 2 study to evaluate the safety and efficacy of centhaquine in COVID-19 patients with ARDS is on-going (NCT05241067). In that study, subjects are randomized to a treatment group and a control group. The treatment group will receive an intravenous infusion of 0.01 mg/kg centhaquine with standard of care. An additional dose of centhaquine is administered if oxygenation is required or SBP remains or falls below or becomes equal to, 90 mmHg; but not until 24 hours after the previous dose. The control group receives an intravenous infusion of normal saline with standard of care. The primary outcome measure is clinical improvement of moderate to severe ARDS in subjects within 28 days. Phase 3 trials have been approved by the FDA and are in the planning stage.

CONCLUSION

COVID-19 has had a devastating impact on human society globally. It has caused an estimated three million deaths and has overwhelmed health care systems in many countries. Thanks to rapid vaccine development and drug development, the COVID-19 pandemic appears to be abating. However, long COVID remains a puzzling and distressing problem. The NIH is currently preparing clinical trials aimed at investigating how best to treat this entity. Several studies are planned to investigate whether nirmatrelvir (Paxlovid) is effective in long COVID patients and how to address neuronal problems associated with long COVID. It is impossible to predict the next pandemic, with a coronavirus or other pathogen. However, the cumulative knowledge of treatment strategies gained in this pandemic, are essential as we prepare to face the next pandemic threat.

Treatment of COVID-19 has evolved rapidly since the start of the pandemic. Antiviral agents are currently available for patients with mild-to-moderate risk of disease progression. Immunomodulators are available for hospitalized patients. However. currently approved monoclonal antibodies may lose efficacy as new viral variants emerge, requiring the development of new antibodies. Immunomodulators are approved to prevent cytokine storms. Combining antiviral treatment with immunomodulation is crucial for treating patients with severe COVID-19. With antiviral and anti-inflammatory properties, JAK inhibitors showed a potential treatment for severe ARDS. Anti-coagulation is important for reducing the risk of thrombosis.

In summary, here we reviewed approved drugs and drugs in development for COVID-19 and discussed their mechanisms of action. This background is intended to help clinicians and researchers. It must be borne in mind that the virus evolves rapidly, and the field must and will do so as well.

SUPPLEMENTARY MATERIAL

Supplementary Table 1

Withdrawn drug list

Click here to view.(239K, doc)

Notes

Funding:This work was supported by a new faculty research fund of Ajou University School of Medicine. It was also supported by a grant (RS-2023-00212830) of the Basic Science Research Program through the National Research Foundation (NRF) funded by the Ministry of Science, ICT & Future Planning, Republic of Korea. The project has been supported in part the National Institutes of Health and the National Cancer Institute Intramural Research Program (CAW) and under contract 75N91019D00024.

Disclosure:The authors have no conflicts of interest to disclose.

Data Availability Statement:Raw data are available from the corresponding author upon reasonable request.

Author Contributions:

  • Conceptualization: Cho SK.

  • Data curation: Choi HS, Choi AY.

  • Supervision: Cho SK, Kopp JB, Winkler CA.

  • Writing - original draft: Cho SK, Choi HS, Choi AY.

  • Writing - review & editing: Cho SK, Kopp JB, Winkler CA.

ACKNOWLEDGMENTS

We appreciate Young Sup Cho (MD, PhD) and Hyekyung Son (MD, PhD) for their helpful advice.

References

    1. Centers for Disease Control and Prevention. Basics of COVID-19. [Updated 2021]. [Accessed March 19, 2023].
    1. World Health Organization. WHO COVID-19 dashboard. [Updated 2023]. [Accessed December 24, 2023].
    1. National Institutes of Health. Accelerating COVID-19 therapeutic interventions and vaccines (ACTIV). [Updated 2023]. [Accessed March 19, 2023].
    1. Goletti D, Cantini F. Baricitinib therapy in Covid-19 pneumonia - an unmet need fulfilled. N Engl J Med 2021;384(9):867–869.
    1. Marshall JC, Murthy S, Diaz J, Adhikari NK, Angus DC, Arabi YM, et al. A minimal common outcome measure set for COVID-19 clinical research. Lancet Infect Dis 2020;20(8):e192–e197.
    1. LaVange L, Adam SJ, Currier JS, Higgs ES, Reineck LA, Hughes EA, et al. Accelerating COVID-19 therapeutic interventions and vaccines (ACTIV): designing master protocols for evaluation of candidate COVID-19 therapeutics. Ann Intern Med 2021;174(9):1293–1300.
    1. U.S. Food and Drug Administration. Coronavirus (COVID-19) update: FDA authorizes first oral antiviral for treatment of COVID-19. [Updated 2021]. [Accessed March 13, 2023].
    1. Pfizer. Paxlovid: EPAR - Product information. [Updated 2022]. [Accessed January 8, 2023].
    1. U.S. Food and Drug Administration. Paxlovid LOA 02012023. [Updated 2023]. [Accessed March 13, 2023].
    1. Lee E, Park S, Choi JP, Kim MK, Yang E, Ham SY, et al. Short-term effectiveness of oral nirmatrelvir/ritonavir against the SARS-CoV-2 omicron variant and culture-positive viral shedding. J Korean Med Sci 2023;38(8):e59
    1. Kim MK, Lee KS, Ham SY, Choi YY, Lee E, Lee S, et al. Real-world effectiveness of nirmatrelvir-ritonavir and its acceptability in high-risk COVID-19 patients. J Korean Med Sci 2023;38(35):e272
    1. Masyeni S, Iqhrammullah M, Frediansyah A, Nainu F, Tallei T, Emran TB, et al. Molnupiravir: a lethal mutagenic drug against rapidly mutating severe acute respiratory syndrome coronavirus 2-a narrative review. J Med Virol 2022;94(7):3006–3016.
    1. U.S. Food and Drug Administration. Coronavirus (COVID-19) update: FDA authorizes additional oral antiviral for treatment of COVID-19 in certain adults. [Updated 2021]. [Accessed March 16, 2023].
    1. European Medicines Agency. Refusal of the marketing authorisation for Lagevrio (molnupiravir). [Updated 2023]. [Accessed March 16, 2023].
    1. Merck Sharp & Dohme LLC. Fact sheet for healthcare providers: emergency use authorization for Lagevrio™ (Molnupiravir) capsules [fact sheet]. [Updated 2022]. [Accessed January 8, 2023].
    1. Zhang JL, Li YH, Wang LL, Liu HQ, Lu SY, Liu Y, et al. Azvudine is a thymus-homing anti-SARS-CoV-2 drug effective in treating COVID-19 patients. Signal Transduct Target Ther 2021;6(1):414.
    1. Yu B, Chang J. The first Chinese oral anti-COVID-19 drug Azvudine launched. Innovation (Camb) 2022;3(6):100321
    1. U.S. Food and Drug Administration. FDA approves first treatment for COVID-19. [Updated 2020]. [Accessed March 16, 2023].
    1. Gilead Sciences Ireland UC. Veklury: EPAR - product information. [Updated 2022]. [Accessed January 8, 2023].
    1. Jeon J, Chin B. Treatment options for patients with mild-to-moderate coronavirus disease 2019 in Korea. J Korean Med Sci 2022;37(48):e352
    1. Kang H, Kang CK, Im JH, Cho Y, Kang DY, Lee JY. Adverse drug events associated with remdesivir in real-world hospitalized patients with COVID-19, including vulnerable populations: a retrospective multicenter study. J Korean Med Sci 2023;38(44):e346
    1. Ansems K, Grundeis F, Dahms K, Mikolajewska A, Thieme V, Piechotta V, et al. Remdesivir for the treatment of COVID-19. Cochrane Database Syst Rev 2021;8(8):CD014962
    1. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, et al. Remdesivir for the treatment of Covid-19 - final report. N Engl J Med 2020;383(19):1813–1826.
    1. R-Pharm. Artlegia (olokizumab) [package insert]. [Updated 2022]. [Accessed March 18, 2023].
    1. Qian HJ, Wang Y, Zhang MQ, Xie YC, Wu QQ, Liang LY, et al. Safety, tolerability, and pharmacokinetics of VV116, an oral nucleoside analog against SARS-CoV-2, in Chinese healthy subjects. Acta Pharmacol Sin 2022;43(12):3130–3138.
    1. Delhi P. DCGI approves anti-COVID drug developed by DRDO for emergency use. [Updated 2021]. [Accessed March 16, 2023].
    1. Sahu KK, Kumar R. Role of 2-deoxy-D-glucose (2-DG) in COVID-19 disease: a potential game-changer. J Family Med Prim Care 2021;10(10):3548–3552.
    1. European Medicines Agency. EMA recommends approval for use of Kineret in adults with COVID-19. [Updated 2021]. [Accessed March 16, 2023].
    1. Swedish Orphan Biovitrum AB. KINERET® (anakinra) [package insert]. [Updated 2020]. [Accessed June 17, 2023].
    1. Swedish Orphan Biovitrum AB. Kineret: EPAR - product information. [Updated 2022]. [Accessed January 8, 2023].
    1. U.S. Food and Drug Administration. Kineret LOA 11082022. [Updated 2022]. [Accessed March 16, 2023].
    1. R-PHARM. Synthesizing and developing molecules. [Updated 2023]. [Accessed March 16, 2023].
    1. European Medicines Agency. EMA recommends approval for use of RoActemra in adults with severe COVID-19. [Updated 2021]. [Accessed March 16, 2023].
    1. Genentech, Inc. ACTEMRA® (tocilizumab) [package insert]. [Updated 2022]. [Accessed June 17, 2023].
    1. Roche Pharma AG. RoActemra: EPAR - product information. [Updated October 2022]. [Accessed January 8, 2023].
    1. U.S. Food and Drug Administration. Genentech Actemra tocilizumab LOA 12212022. [Updated 2022]. [Accessed January 8, 2023].
    1. Biocad. A drug against COVID-19 complications is registered in Russia. [Updated 2020]. [Accessed March 16, 2023].
    1. Lomakin NV, Bakirov BA, Protsenko DN, Mazurov VI, Musaev GH, Moiseeva OM, et al. The efficacy and safety of levilimab in severely ill COVID-19 patients not requiring mechanical ventilation: results of a multicenter randomized double-blind placebo-controlled phase III CORONA clinical study. Inflamm Res 2021;70(10-12):1233–1246.
    1. Biocad. A Clinical trial of the efficacy and safety of levilimab (BCD-089) in patients with severe COVID-19. [Updated 2020]. [Accessed March 16, 2023].
    1. Atal S, Fatima Z, Balakrishnan S. Approval of itolizumab for COVID-19: a premature decision or need of the hour? BioDrugs 2020;34(6):705–711.
    1. Loganathan S, Athalye SN, Joshi SR. Itolizumab, an anti-CD6 monoclonal antibody, as a potential treatment for COVID-19 complications. Expert Opin Biol Ther 2020;20(9):1025–1031.
    1. European Medicines Agency. Withdrawal of application to change the marketing authorisation for Olumiant (baricitinib). [Updated 2022]. [Accessed March 16, 2023].
    1. Eli Lilly and Company. Olumiant (baricitinib) [package insert]. [Updated 2022]. [Accessed June 17, 2023].
    1. Eli Lilly and Company. Fact sheet for healthcare providers emergency use authorization (EUA) of baricitinib [fact sheet]. [Updated 2022]. [Accessed January 8, 2023].
    1. U.S. Food and Drug Administration. Baricitinib EUA Letter of Authorization 10272022. [Updated 2022]. [Accessed March 16, 2023].
    1. Park DH, Kang CK, Choe PG, Kim NJ, Park WB, Oh MD. How we have treated severe to critically ill patients with coronavirus disease 2019 in Korea. J Korean Med Sci 2022;37(49):e353
    1. Imai M, Ito M, Kiso M, Yamayoshi S, Uraki R, Fukushi S, et al. Efficacy of antiviral agents against omicron subvariants BQ.1.1 and XBB. N Engl J Med 2023;388(1):89–91.
    1. European Medicines Agency. Evusheld (tixagevimab/cilgavimab) an overview of Evusheld and why it is authorised in the EU. [Updated 2022]. [Accessed March 24, 2023].
    1. U.S. Food and Drug Administration. FDA announces Evusheld is not currently authorized for emergency use in the U.S. [Updated 2023]. [Accessed March 24, 2023].
    1. U.S. Food and Drug Administration. Evusheld Letter of Authorization 01262023. [Updated 2023]. [Accessed March 24, 2023].
    1. AstraZeneca AB. Evusheld: EPAR - product information. [Updated 2022]. [Accessed January 8, 2023].
    1. NUVISAN GmbH. Regkirona. EPAR - product information. [Updated 2022]. [Accessed January 8, 2023].
    1. Planas D, Saunders N, Maes P, Guivel-Benhassine F, Planchais C, Buchrieser J, et al. Considerable escape of SARS-CoV-2 Omicron to antibody neutralization. Nature 2022;602(7898):671–675.
    1. Roche Pharma AG. Ronapreve: EPAR - product information. [Updated 2022]. [Accessed January 8, 2023].
    1. U.S. Food and Drug Administration. Coronavirus (COVID-19) update: FDA authorizes additional monoclonal antibody for treatment of COVID-19. [Updated 2021]. [Accessed March 13, 2023].
    1. U.S. Food and Drug Administration. FDA updates Sotrovimab emergency use authorization. [Updated 2022]. [Accessed March 13, 2023].
    1. GlaxoSmithKline. Xevudy: EPAR - product information. [Updated 2022]. [Accessed January 8, 2023].
    1. Hoy SM. Amubarvimab/romlusevimab: first approval. Drugs 2022;82(12):1327–1331.
    1. Bian H, Zheng ZH, Wei D, Wen A, Zhang Z, Lian JQ, et al. Safety and efficacy of meplazumab in healthy volunteers and COVID-19 patients: a randomized phase 1 and an exploratory phase 2 trial. Signal Transduct Target Ther 2021;6(1):194.
    1. Wang K, Chen W, Zhang Z, Deng Y, Lian JQ, Du P, et al. CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct Target Ther 2020;5(1):283.
    1. Papapanou M, Papoutsi E, Giannakas T, Katsaounou P. Plitidepsin: mechanisms and clinical profile of a promising antiviral agent against COVID-19. J Pers Med 2021;11(7):668.
    1. White KM, Rosales R, Yildiz S, Kehrer T, Miorin L, Moreno E, et al. Plitidepsin has potent preclinical efficacy against SARS-CoV-2 by targeting the host protein eEF1A. Science 2021;371(6532):926–931.
    1. Sound Pharmaceuticals. Our product pipeline. [Updated 2023]. [Accessed June 17, 2023].
    1. Azad GK, Tomar RS. Ebselen, a promising antioxidant drug: mechanisms of action and targets of biological pathways. Mol Biol Rep 2014;41(8):4865–4879.
    1. Haritha CV, Sharun K, Jose B. Ebselen, a new candidate therapeutic against SARS-CoV-2. Int J Surg 2020;84:53–56.
    1. Amporndanai K, Meng X, Shang W, Jin Z, Rogers M, Zhao Y, et al. Inhibition mechanism of SARS-CoV-2 main protease by ebselen and its derivatives. Nat Commun 2021;12(1):3061.
    1. Sendi P, Razonable RR, Nelson SB, Soriano A, Gandhi RT. First-generation oral antivirals against SARS-CoV-2. Clin Microbiol Infect 2022;28(9):1230–1235.
    1. Barnette KG, Gordon MS, Rodriguez D, Bird TG, Skolnick A, Schnaus M, et al. Oral sabizabulin for high-risk, hospitalized adults with Covid-19: interim analysis. NEJM Evid 2022;1(9):a2200145
    1. Veru Inc. Veru provides FDA update on request for emergency use authorization for sabizabulin for hospitalized COVID-19 patients at high risk for ARDS. [Updated 2023]. [Accessed March 13, 2023].
    1. Mustafa DA, Saida L, Latifi D, Wismans LV, de Koning W, Zeneyedpour L, et al. Rintatolimod induces antiviral activities in human pancreatic cancer cells: opening for an anti-COVID-19 opportunity in cancer patients? Cancers (Basel) 2021;13(12):2896.
    1. AIM ImmunoTech. AIM ImmunoTech announces central IRB approval of phase 2 study protocol evaluating Ampligen® for the treatment of post-COVID conditions. [Updated 2023]. [Accessed March 13, 2023].
    1. Biopharmaceuticals. Pipeline. [Updated 2023]. [Accessed March 18, 2023].
    1. Meng X, Ling Y, Zhang L, Zhang Q, Dong P, Zhu T, et al. Potential for jaktinib hydrochloride to treat cytokine storms in patients with COVID-19. Biosci Trends 2020;14(3):161–167.
    1. Novartis Pharmaceuticals. ILARIS®(canakinumab) [package insert]. [Updated 2020]. [Accessed March 18, 2023].
    1. Mai W, Liao Y. Targeting IL-1β in the treatment of atherosclerosis. Front Immunol 2020;11:589654
    1. Generali D, Bosio G, Malberti F, Cuzzoli A, Testa S, Romanini L, et al. Canakinumab as treatment for COVID-19-related pneumonia: a prospective case-control study. Int J Infect Dis 2021;104:433–440.
    1. Zarbock A, Allegretti M, Ley K. Therapeutic inhibition of CXCR2 by Reparixin attenuates acute lung injury in mice. Br J Pharmacol 2008;155(3):357–364.
    1. Dompe. Research and development pipeline. [Updated 2020]. [Accessed March 19, 2023].
    1. Landoni G, Piemonti L, Monforte AD, Grossi P, Zangrillo A, Bucci E, et al. A multicenter phase 2 randomized controlled study on the efficacy and safety of reparixin in the treatment of hospitalized patients with COVID-19 pneumonia. Infect Dis Ther 2022;11(4):1559–1574.
    1. Eli Lilly and Company. TALTZ (ixekizumab) [package insert]. [Updated 2022]. [Accessed March 19, 2023].
    1. Bulat V, Situm M, Azdajic MD, Likic R. Potential role of IL-17 blocking agents in the treatment of severe COVID-19? Br J Clin Pharmacol 2021;87(3):1578–1581.
    1. Maini RN, Feldmann M. How does infliximab work in rheumatoid arthritis? Arthritis Res 2002;4 Suppl 2:S22–S28.
    1. Janssen Biotech. INFLIXIMAB [package insert]. [Updated 2021]. [Accessed March 19, 2023].
    1. Bristol-Myers Squibb Company. ORENCIA (abatacept) [package insert]. [Updated 2021]. [Accessed March 19, 2023].
    1. Yang D, Li H, Chen Y, Ren W, Dong M, Li C, et al. Immunomodulatory mechanisms of abatacept: a therapeutic strategy for COVID-19. Front Med (Lausanne) 2022;9:951115
    1. Amarin Pharmaceuticals. VASCEPA® (icosapent ethyl) [package insert]. [Updated 2021]. [Accessed March 19, 2023].
    1. Kosmopoulos A, Bhatt DL, Meglis G, Verma R, Pan Y, Quan A, et al. A randomized trial of icosapent ethyl in ambulatory patients with COVID-19. iScience 2021;24(9):103040
    1. Shaperon. Pipeline - NuSepin®. [Updated 2023]. [Accessed March 19, 2023].
    1. Pharming Group. RUCONEST® [package insert]. [Updated 2014]. [Accessed March 19, 2023].
    1. Urwyler P, Moser S, Charitos P, Heijnen IA, Rudin M, Sommer G, et al. Treatment of COVID-19 with conestat alfa, a regulator of the complement, contact activation and kallikrein-kinin system. Front Immunol 2020;11:2072.
    1. Kyorin Pharmaceutical. KETAS® [package insert]. [Updated 2023]. [Accessed March 19, 2023].
    1. Oskarsson B, Maragakis N, Bedlack RS, Goyal N, Meyer JA, Genge A, et al. MN-166 (ibudilast) in amyotrophic lateral sclerosis in a Phase IIb/III study: COMBAT-ALS study design. Neurodegener Dis Manag 2021;11(6):431–443.
    1. Crocetti L, Floresta G, Cilibrizzi A, Giovannoni MP. An overview of PDE4 inhibitors in clinical trials: 2010 to early 2022. Molecules 2022;27(15):4964.
    1. Capricor Therapeutics, Inc. Product pipeline - CAP-1002. [Updated 2023]. [Accessed March 19, 2023].
    1. Singh S, Chakravarty T, Chen P, Akhmerov A, Falk J, Friedman O, et al. Allogeneic cardiosphere-derived cells (CAP-1002) in critically ill COVID-19 patients: compassionate-use case series. Basic Res Cardiol 2020;115(4):36.
    1. Humphries DC, Mills R, Boz C, McHugh BJ, Hirani N, Rossi AG, et al. Galectin-3 inhibitor GB0139 protects against acute lung injury by inhibiting neutrophil recruitment and activation. Front Pharmacol 2022;13:949264
    1. Galecto. Pipeline. [Updated 2023]. [Accessed March 19, 2023].
    1. Daly A. Remestemcel-L, the first cellular therapy product for the treatment of graft-versus-host disease. Drugs Today (Barc) 2012;48(12):773–783.
    1. Mesoblast. Product candidates overview. [Updated 2023]. [Accessed March 19, 2023].
    1. Eckard AR, Borow KM, Mack EH, Burke E, Atz AM. Remestemcel-L therapy for COVID-19-associated multisystem inflammatory syndrome in children. Pediatrics 2021;147(5):e2020046573
    1. Kleen TO, Galdon AA, MacDonald AS, Dalgleish AG. Mitigating coronavirus induced dysfunctional immunity for at-risk populations in COVID-19: trained immunity, BCG and “New Old Friends”. Front Immunol 2020;11:2059.
    1. Immodulon. IMM-101 and cancer. [Updated 2022]. [Accessed March 18, 2023].
    1. Biophytis. Sarconeos (BIO101). [Updated 2023]. [Accessed March 18, 2023].
    1. Latil M, Camelo S, Veillet S, Lafont R, Dilda PJ. Developing new drugs that activate the protective arm of the renin-angiotensin system as a potential treatment for respiratory failure in COVID-19 patients. Drug Discov Today 2021;26(5):1311–1318.
    1. Biophytis. Biophytis announces positive final results of the phase 2-3 COVA study with Sarconeos (BIO101) in severe COVID-19 [Press release]. [Updated 2023]. [Accessed March 18, 2023].
    1. Mu X, Liu K, Li H, Wang FS, Xu R. Granulocyte-macrophage colony-stimulating factor: an immunotarget for sepsis and COVID-19. Cell Mol Immunol 2021;18(8):2057–2058.
    1. Metcalf D. The colony-stimulating factors and cancer. Nat Rev Cancer 2010;10(6):425–434.
    1. Ushach I, Zlotnik A. Biological role of granulocyte macrophage colony-stimulating factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) on cells of the myeloid lineage. J Leukoc Biol 2016;100(3):481–489.
    1. Partner Therapeutics. Leukine (sargramostim) [package insert]. [Updated 2022]. [Accessed March 18, 2023].
    1. McCormick TS, Hejal RB, Leal LO, Ghannoum MA. orchestrating the pulmonary response to infection. Front Pharmacol 2022;12:735443
    1. Bonaventura A, Vecchié A, Wang TS, Lee E, Cremer PC, Carey B, et al. Targeting GM-CSF in COVID-19 pneumonia: rationale and strategies. Front Immunol 2020;11:1625.
    1. Biopharma IM. Product pipeline. [Updated 2023]. [Accessed March 19, 2023].
    1. Temesgen Z, Assi M, Shweta FN, Vergidis P, Rizza SA, Bauer PR, et al. GM-CSF neutralization with lenzilumab in severe COVID-19 pneumonia: a case-cohort study. Mayo Clin Proc 2020;95(11):2382–2394.
    1. Humanigen. HGEN Lenzilumab Fact Sheet [Fact sheet]. [Updated 2021]. [Accessed March 18, 2023].
    1. Sanofi Company. LOVENOX (enoxaparin sodium) [package insert]. U.S. Food and Drug Administration website; [Updated 2021]. [Accessed June 17, 2023].
    1. Pfizer Labs. FRAGMIN® (dalteparin sodium) [package insert]. [Updated 2020]. [Accessed June 17, 2023].
    1. LEO Pharma Inc. Innohep (tinzaparin sodium injection) [package insert]. [Updated 2010]. [Accessed June 17, 2023].
    1. Warnock LB, Huang D. In: Heparin. Treasure Island, FL, USA: StatPearls Publishing; 2023.
    1. Mangiafico M, Caff A, Costanzo L. The role of heparin in COVID-19: an update after two years of pandemics. J Clin Med 2022;11(11):3099.
    1. Janssen Pharmaceuticals, Inc. XARELTO® (rivaroxaban) [package insert]. [Updated 2023]. [Accessed June 17, 2023].
    1. Capell WH, Barnathan ES, Piazza G, Spyropoulos AC, Hsia J, Bull S, et al. Rationale and design for the study of rivaroxaban to reduce thrombotic events, hospitalization and death in outpatients with COVID-19: The PREVENT-HD study. Am Heart J 2021;235:12–23.
    1. Hess CN, Capell WH, Bristow MR, Ruf W, Szarek M, Morrow DA, et al. Rationale and design of a study to assess the safety and efficacy of rNAPc2 in COVID-19: the Phase 2b ASPEN-COVID-19 trial. Am Heart J 2022;246:136–143.
    1. Lee AY, Vlasuk GP. Recombinant nematode anticoagulant protein c2 and other inhibitors targeting blood coagulation factor VIIa/tissue factor. J Intern Med 2003;254(4):313–321.
    1. Hess CN, Hsia J, Carroll IA, Nehler MR, Ruf W, Morrow DA, et al. Novel tissue factor inhibition for thromboprophylaxis in COVID-19: primary results of the ASPEN-COVID-19 trial. Arterioscler Thromb Vasc Biol 2023;43(8):1572–1582.
    1. Rigel Pharmaceuticals. TAVALISSE™ (fostamatinib disodium hexahydrate) [package insert]. [Updated 2018]. [Accessed March 19, 2023].
    1. Strich JR, Ramos-Benitez MJ, Randazzo D, Stein SR, Babyak A, Davey RT, et al. Fostamatinib inhibits neutrophils extracellular traps induced by COVID-19 patient plasma: a potential therapeutic. J Infect Dis 2021;223(6):981–984.
    1. Bournazos S, Gupta A, Ravetch JV. The role of IgG Fc receptors in antibody-dependent enhancement. Nat Rev Immunol 2020;20(10):633–643.
    1. Apostolidis SA, Sarkar A, Giannini HM, Goel RR, Mathew D, Suzuki A, et al. Signaling through FcγRIIA and the C5a-C5aR pathway mediate platelet hyperactivation in COVID-19. Front Immunol 2022;13:834988
    1. Jazz Pharmaceuticals. DEFITELIO (defibrotide sodium) [package insert]. [Updated 2016]. [Accessed March 19, 2023].
    1. Macciò A, La Nasa G, Oppi S, Madeddu C. Defibrotide has a role in COVID-19 therapy. Chest 2022;162(2):271–273.
    1. Berk C, Sabbagh MN. Successes and failures for drugs in late-stage development for Alzheimer’s disease. Drugs Aging 2013;30(10):783–792.
    1. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2019. Alzheimers Dement (N Y) 2019;5(1):272–293.
    1. Al-Horani RA, Kar S. Potential anti-SARS-CoV-2 therapeutics that target the post-entry stages of the viral life cycle: a comprehensive review. Viruses 2020;12(10):1092.
    1. Biomed Industries. Pipeline. [Updated 2021]. [Accessed June 17, 2023].
    1. Taylor CT, Scholz CC. The effect of HIF on metabolism and immunity. Nat Rev Nephrol 2022;18(9):573–587.
    1. Poloznikov AA, Nersisyan SA, Hushpulian DM, Kazakov EH, Tonevitsky AG, Kazakov SV, et al. HIF prolyl hydroxylase inhibitors for COVID-19 treatment: pros and cons. Front Pharmacol 2021;11:621054
    1. Haase VH. HIF-prolyl hydroxylases as therapeutic targets in erythropoiesis and iron metabolism. Hemodial Int 2017;21 Suppl 1:S110–S124.
    1. Genentech. AVASTIN [package insert]. [Updated 2022]. [Accessed March 17, 2023].
    1. Garcia J, Hurwitz HI, Sandler AB, Miles D, Coleman RL, Deurloo R, et al. Bevacizumab (Avastin®) in cancer treatment: a review of 15 years of clinical experience and future outlook. Cancer Treat Rev 2020;86:102017
    1. Pang J, Xu F, Aondio G, Li Y, Fumagalli A, Lu M, et al. Efficacy and tolerability of bevacizumab in patients with severe Covid-19. Nat Commun 2021;12(1):814.
    1. Akebia Therapeutics I. Research & development. [Updated 2022]. [Accessed March 17, 2023].
    1. Akebia Therapeutics. Akebia therapeutics announces initial findings from investigator-sponsored clinical study evaluating vadadustat for the prevention and treatment of acute respiratory distress syndrome (ARDS) in subjects with COVID-19 and hypoxemia (VSTAT) [Press release]. [Updated 2022]. [Accessed March 17, 2023].
    1. Biotest. Development pipeline - Trimodulin (BT-588, IgM Concentrate). [Updated 2023]. [Accessed March 18, 2023].
    1. Bohländer F, Riehl D, Weißmüller S, Gutscher M, Schüttrumpf J, Faust S. Immunomodulation: immunoglobulin preparations suppress hyperinflammation in a COVID-19 model via FcγRIIA and FcαRI. Front Immunol 2021;12:700429
    1. Octapharma. Octagam [package insert]. [Updated 2022]. [Accessed March 18, 2023].
    1. Xiang HR, Cheng X, Li Y, Luo WW, Zhang QZ, Peng WX. Efficacy of IVIG (intravenous immunoglobulin) for corona virus disease 2019 (COVID-19): a meta-analysis. Int Immunopharmacol 2021;96:107732
    1. RedHill Biopharma. RedHill biopharma reports that opaganib mechanism not impacted by viral spike-protein mutations, including omicron mutations. [Updated 2021]. [Accessed March 18, 2023].
    1. Smith CD, Maines LW, Keller SN, Katz Ben-Yair V, Fathi R, Plasse TF, et al. Recent progress in the development of opaganib for the treatment of Covid-19. Drug Des Devel Ther 2022;16:2199–2211.
    1. 60 Degrees Pharmaceuticals. ARAKODA™ (tafenoquine) [package insert]. [Updated 2018]. [Accessed June 17, 2023].
    1. Chen Y, Yang WH, Chen HF, Huang LM, Gao JY, Lin CW, et al. Tafenoquine and its derivatives as inhibitors for the severe acute respiratory syndrome coronavirus 2. J Biol Chem 2022;298(3):101658
    1. Pharmaceuticals AN. FLUVOXAMINE MALEATE [package insert]. [Updated 2021]. [Accessed March 18, 2023].
    1. Sukhatme VP, Reiersen AM, Vayttaden SJ, Sukhatme VV. Fluvoxamine: a review of its mechanism of action and its role in COVID-19. Front Pharmacol 2021;12:652688
    1. Croft SL, Duparc S, Arbe-Barnes SJ, Craft JC, Shin CS, Fleckenstein L, et al. Review of pyronaridine anti-malarial properties and product characteristics. Malar J 2012;11(1):270.
    1. Shin Poong Pharmaceutical. Pyramax. [Updated 2023]. [Accessed June 17, 2023].
    1. Zhou Y, Gilmore K, Ramirez S, Settels E, Gammeltoft KA, Pham LV, et al. In vitro efficacy of artemisinin-based treatments against SARS-CoV-2. Sci Rep 2021;11(1):14571.
    1. Puhl AC, Gomes GF, Damasceno S, Godoy AS, Noske GD, Nakamura AM, et al. Pyronaridine protects against SARS-CoV-2 infection in mouse. ACS Infect Dis 2022;8(6):1147–1160.
    1. Tsujikawa LM, Fu L, Das S, Halliday C, Rakai BD, Stotz SC, et al. Apabetalone (RVX-208) reduces vascular inflammation in vitro and in CVD patients by a BET-dependent epigenetic mechanism. Clin Epigenetics 2019;11(1):102.
    1. RESVERLOGIX. APABETALONE Fact Sheet [Fact sheet]. [Updated 2022]. [Accessed March 18, 2023].
    1. Gilham D, Smith AL, Fu L, Moore DY, Muralidharan A, Reid SP, et al. Bromodomain and extraterminal protein inhibitor, apabetalone (RVX-208), reduces ACE2 expression and attenuates SARS-CoV-2 infection in vitro. Biomedicines 2021;9(4):437.
    1. NeuroBo Pharmaceuticals, Inc. ANA001. [Updated 2023]. [Accessed March 18, 2023].
    1. Singh S, Weiss A, Goodman J, Fisk M, Kulkarni S, Lu I, et al. Niclosamide-A promising treatment for COVID-19. Br J Pharmacol 2022;179(13):3250–3267.
    1. Al-Kuraishy HM, Al-Gareeb AI, Alzahrani KJ, Alexiou A, Batiha GE. Niclosamide for Covid-19: bridging the gap. Mol Biol Rep 2021;48(12):8195–8202.
    1. Kinarus Therapeutics Holding AG. Therapeutic approach. [Updated 2023]. [Accessed March 18, 2023].
    1. Setz C, Große M, Auth J, Fröba M, Rauch P, Bausch A, et al. Synergistic antiviral activity of pamapimod and pioglitazone against SARS-CoV-2 and its variants of concern. Int J Mol Sci 2022;23(12):6830.
    1. Karyopharm Therapeutics. XPOVIO (selinexor) [package insert]. [Updated 2022]. [Accessed March 18, 2023].
    1. Mostafa-Hedeab G, Al-Kuraishy HM, Al-Gareeb AI, Welson NN, El-Saber Batiha G, Conte-Junior CA. Selinexor and COVID-19: the neglected warden. Front Pharmacol 2022;13:884228
    1. Romark. ALINIA® (nitazoxanide) [package insert]. [Updated 2022]. [Accessed March 18, 2023].
    1. Lokhande AS, Devarajan PV. A review on possible mechanistic insights of Nitazoxanide for repurposing in COVID-19. Eur J Pharmacol 2021;891:173748
    1. SaNOtize. VirX-enovid. [Updated 2023]. [Accessed March 18, 2023].
    1. Tandon M, Wu W, Moore K, Winchester S, Tu YP, Miller C, et al. Study Group. SARS-CoV-2 accelerated clearance using a novel nitric oxide nasal spray (NONS) treatment: a randomized trial. Lancet Reg Health Southeast Asia 2022;3:100036
    1. Alqahtani JS, Aldhahir AM, Al Ghamdi SS, AlBahrani S, AlDraiwiesh IA, Alqarni AA, et al. Inhaled nitric oxide for clinical management of COVID-19: a systematic review and meta-analysis. Int J Environ Res Public Health 2022;19(19):12803.
    1. Valeo Pharma. Valeo Pharma receives Hesperco™ natural product license approval from Health Canada. [Updated 2020]. [Accessed March 18, 2023].
    1. Bellavite P, Donzelli A. Hesperidin and SARS-CoV-2: new light on the healthy function of citrus fruits. Antioxidants 2020;9(8):742.
    1. Viatris Specialty LL. LIPITOR® (atorvastatin calcium) [package insert]. [Updated 2022]. [Accessed January 8, 2023].
    1. INSPIRATION-S Investigators. Atorvastatin versus placebo in patients with covid-19 in intensive care: randomized controlled trial. BMJ 2022;376:e068407
    1. Dalakas MC, Alexopoulos H, Spaeth PJ. Complement in neurological disorders and emerging complement-targeted therapeutics. Nat Rev Neurol 2020;16(11):601–617.
    1. UCB. Clinical studies index - Zilucoplan (RA101495). [Updated 2023]. [Accessed March 18, 2023].
    1. De Leeuw E, Van Damme KF, Declercq J, Bosteels C, Maes B, Tavernier SJ, et al. Efficacy and safety of the investigational complement C5 inhibitor zilucoplan in patients hospitalized with COVID-19: an open-label randomized controlled trial. Respir Res 2022;23(1):202.
    1. Vlaar AP, Lim EH, de Bruin S, Rückinger S, Pilz K, Brouwer MC, et al. The anti-C5a antibody vilobelimab efficiently inhibits C5a in patients with severe COVID-19. Clin Transl Sci 2022;15(4):854–858.
    1. InflaRx. Pipeline. [Updated 2023]. [Accessed March 18, 2023].
    1. Vlaar AP, Witzenrath M, van Paassen P, Heunks LM, Mourvillier B, de Bruin S, et al. Anti-C5a antibody (vilobelimab) therapy for critically ill, invasively mechanically ventilated patients with COVID-19 (PANAMO): a multicentre, double-blind, randomised, placebo-controlled, phase 3 trial. Lancet Respir Med 2022;10(12):1137–1146.
    1. Pharmazz. Lyfaquin (Centhaquine citrate) [product information]. [Updated 2020]. [Accessed March 19, 2023].
    1. Gulati A, Choudhuri R, Gupta A, Singh S, Ali SK, Sidhu GK, et al. A multicentric, randomized, controlled phase III study of centhaquine (Lyfaquin®) as a resuscitative agent in hypovolemic shock patients. Drugs 2021;81(9):1079–1100.
    1. Cao Z, Gao W, Bao H, Feng H, Mei S, Chen P, et al. VV116 versus nirmatrelvir-ritonavir for oral treatment of Covid-19. N Engl J Med 2023;388(5):406–417.
    1. Huang Z, Chavda VP, Vora LK, Gajjar N, Apostolopoulos V, Shah N, et al. 2-deoxy-D-glucose and its derivatives for the COVID-19 treatment: an update. Front Pharmacol 2022;13:899633
    1. Biocon, L. Efficacy and safety of itolizumab in COVID-19 complications. [Updated 2020]. [Accessed March 19, 2023].
    1. Samama MM. The mechanism of action of rivaroxaban--an oral, direct Factor Xa inhibitor--compared with other anticoagulants. Thromb Res 2011;127(6):497–504.
    1. McKeage K, Lyseng-Williamson KA. Fostamatinib in chronic immune thrombocytopenia: a profile of its use in the USA. Drugs Ther Perspect 2018;34(10):451–456.

Metrics
Share
Figures

1 / 5

Tables

1 / 10

PERMALINK