1
|
Peng CC, Humeniuk R, Raut A, Kwan A, Mak L, Stacom C, Xiao D, Chen S, Davies S, Madera S, Koullias Y, Lichtman A, Llewellyn J, Amini E, Winter H, Caro L. Clinical Evaluation of Drug-Drug Interactions with Obeldesivir, an Orally Administered Antiviral Agent. Clin Pharmacol Ther 2025. [PMID: 39887351 DOI: 10.1002/cpt.3575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025]
Abstract
Obeldesivir is an oral nucleoside analog prodrug inhibitor of SARS-CoV-2 RNA-dependent RNA polymerase and other viral polymerases. Here, two Phase I studies evaluated potential drug-drug interactions between obeldesivir and substrates or inhibitors of cytochrome P450 and drug transporters in healthy participants. When obeldesivir was tested as a precipitant, pharmacokinetic parameter point estimates for midazolam (CYP3A4 inhibition/induction), caffeine (CYP1A2 inhibition), and metformin (organic cation transporter 1 inhibition) exposures were within 80-125% no-effect bounds representing the interval within which a systemic exposure change does not warrant clinical action based on EMA/FDA guidance. Dabigatran (P-glycoprotein substrate) and pitavastatin (organic anion transporting polypeptide 1B1/1B3 substrate) exposures decreased by approximately 25% and 30%, respectively, with obeldesivir coadministration; these were considered not clinically relevant, as these exposure changes are not associated with dose changes or precautions in the US prescribing information for these drugs. When obeldesivir was evaluated as an object, exposures of GS-441524, the parent nucleoside monophosphate metabolite of obeldesivir, were within the 80-125% no-effect bounds for ritonavir (P-glycoprotein inhibition) and cyclosporin A (breast cancer resistance protein inhibition) coadministration. Famotidine (gastric acid suppression) coadministration decreased GS-441524 exposure by approximately 26%; this was within the range of exposures observed in previous Phase III studies and was considered not clinically relevant. Obeldesivir was well tolerated, and adverse events were mild to moderate. These findings indicate that obeldesivir has low potential for drug-drug interactions. Obeldesivir remains a promising treatment against a broad spectrum of viruses given its antiviral activity and favorable safety profile.
Collapse
Affiliation(s)
- Chi-Chi Peng
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Anuja Raut
- Gilead Sciences, Inc., Foster City, California, USA
| | - Anna Kwan
- Gilead Sciences, Inc., Foster City, California, USA
| | - Lily Mak
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Deqing Xiao
- Gilead Sciences, Inc., Foster City, California, USA
| | | | | | | | | | | | | | - Elham Amini
- Gilead Sciences, Inc., Foster City, California, USA
| | - Helen Winter
- Gilead Sciences, Inc., Foster City, California, USA
| | | |
Collapse
|
2
|
Nagano Y, Aoki H, David J, Kondo NI, Aizawa M, Sumita T, Hamada Y, Yamamoto Y, Yamada K. Remdesivir-induced severe hypoglycemia in an elderly man without diabetes: a case report. J Pharm Health Care Sci 2025; 11:5. [PMID: 39871315 PMCID: PMC11770970 DOI: 10.1186/s40780-024-00406-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/23/2024] [Indexed: 01/29/2025] Open
Abstract
BACKGROUND Remdesivir is recommended to treat hospitalized patients with coronavirus disease 2019 (COVID-19). Remdesivir is known to affect glucose metabolism in individuals with and without diabetes. However, little is known about the possibility of hypoglycemia associated with remdesivir. Our case is the first report demonstrating the development of severe hypoglycemia following remdesivir treatment in an elderly man without diabetes. CASE PRESENTATION A 73-year-old man developed COVID-19 following surgery for sigmoid volvulus. The patient's medical history included surgery for posterior correction of scoliosis, Chiari malformation type I, and syringomyelia. There was no history of diabetes mellitus. The patient was started on remdesivir (200 mg on day 1, followed by 100 mg intravenously daily until day 9). On day 7, following remdesivir administration, the patient developed severe hypoglycemia with a blood glucose (BG) level of 25 mg/dL. On day 8 and 9 he repeatedly developed severe hypoglycemia following administration of remdesivir. Considering the timing of administration, the patient's hypoglycemia could have been caused by remdesivir. Therefore, his treatment with remdesivir was discontinued. After discontinuation, his BG levels normalized. The Naranjo algorithm, a tool for evaluating the causality of adverse drug reactions, classified the event as "Probable" (6 points). CONCLUSIONS Remdesivir may have caused hypoglycemia in this case. Health care professionals should be aware of its potential effects on glucose metabolism and the risk of hypoglycemia when treating patients with remdesivir.
Collapse
Affiliation(s)
- Yasunori Nagano
- Department of Pharmacy, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan.
| | - Hisae Aoki
- Department of Surgery, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Juri David
- Department of Surgery, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Naoko Iwahashi Kondo
- Department of Surgery, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Makimi Aizawa
- Department of Pharmacy, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Toshiyuki Sumita
- Department of Surgery, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Yuki Hamada
- Department of Pharmacy, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Yuki Yamamoto
- Department of Pharmacy, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| | - Kaoru Yamada
- Department of Internal Medicine, Sanraku Hospital, 2-5 Kandasurugadai, Chiyoda-ku, Tokyo, 1018326, Japan
| |
Collapse
|
3
|
Zhang H, Humeniuk R, Regan S, Koullias Y, Davies S, John A, Shen G, Xiao D, Hyland RH, Winter H, Kim A. Clinical Pharmacokinetics and Safety of Remdesivir in Phase I Participants with Varying Degrees of Renal Impairment. Clin Pharmacokinet 2025; 64:67-78. [PMID: 39562399 DOI: 10.1007/s40262-024-01453-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/31/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND AND OBJECTIVE Remdesivir is a nucleotide analog prodrug approved for the treatment of COVID-19. This study evaluated the pharmacokinetics and safety of remdesivir and its metabolites (GS-704277 and GS-441524) in participants with varying degrees of renal impairment. Results of this phase I study, along with those of a phase III study, contributed to an extension of indication for remdesivir in the USA and Europe for use in patients with COVID-19 with all stages of renal impairment, including those on dialysis, with no dose adjustment. METHODS This phase I, open-label, parallel-group study enrolled participants who had mild (n = 12), moderate (n = 11), or severe (n = 10) renal impairment or kidney failure (n = 6 with dialysis, n = 4 without dialysis). Healthy matched controls were enrolled as reference. Remdesivir was given as single intravenous doses of 100 mg (mild and moderate renal impairment), 40 mg (severe renal impairment, kidney failure predialysis), and 20 mg (kidney failure postdialysis and without dialysis). RESULTS Plasma pharmacokinetics of remdesivir were not affected by mild, moderate, or severe renal impairment or kidney failure. Geometric least squares mean ratios ranged from 0.8 to 1.2 for remdesivir area under the plasma concentration-time curve (AUC). GS-704277 AUC was up to 2.8-fold higher and GS-441524 AUC up to 7.9-fold higher in participants with renal impairment. Adverse events and laboratory abnormalities were consistent with the existing safety profile for remdesivir. CONCLUSIONS Observed pharmacokinetics for remdesivir and its metabolites in participants with renal impairment aligned with expected changes based on known routes of elimination. Remdesivir was generally safe and well tolerated in participants with renal impairment, and no new safety concerns were identified. These results, along with those from the phase III study in patients with COVID-19 with severely reduced kidney function, support the use of remdesivir in patients with any degree of renal impairment with no dose adjustments. TRIAL REGISTRATION EudraCT no. 2020-003441-10; 9 July 2020.
Collapse
Affiliation(s)
| | | | - Sean Regan
- Gilead Sciences, Inc., Foster City, CA, 94404, USA
| | | | | | - Amy John
- Gilead Sciences, Inc., Foster City, CA, 94404, USA
| | - Gong Shen
- Gilead Sciences, Inc., Foster City, CA, 94404, USA
| | - Deqing Xiao
- Gilead Sciences, Inc., Foster City, CA, 94404, USA
| | | | - Helen Winter
- Gilead Sciences, Inc., Foster City, CA, 94404, USA
| | - Aryun Kim
- Gilead Sciences, Inc., Foster City, CA, 94404, USA.
| |
Collapse
|
4
|
Foti RS. Utility of physiologically based pharmacokinetic modeling in predicting and characterizing clinical drug interactions. Drug Metab Dispos 2025; 53:100021. [PMID: 39884811 DOI: 10.1124/dmd.123.001384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/09/2023] [Accepted: 01/02/2024] [Indexed: 02/01/2024] Open
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is a mechanistic dynamic modeling approach that can be used to predict or retrospectively describe changes in drug exposure due to drug-drug interactions (DDIs). With advancements in commercially available PBPK software, PBPK DDI modeling has become a mainstream approach from early drug discovery through to late-stage drug development and is often used to support regulatory packages for new drug applications. This Minireview will briefly describe the approaches to predicting DDI using PBPK and static modeling approaches, the basic model structures and features inherent to PBPK DDI models, and key examples where PBPK DDI models have been used to describe complex DDI mechanisms. Future directions aimed at using PBPK models to characterize transporter-mediated DDI, predict DDI in special populations, and assess the DDI potential of protein therapeutics will be discussed. A summary of the 209 PBPK DDI examples published to date in 2023 will be provided. Overall, current data and trends suggest a continued role for PBPK models in the characterization and prediction of DDI for therapeutic molecules. SIGNIFICANCE STATEMENT: Physiologically based pharmacokinetic (PBPK) models have been a key tool in the characterization of various pharmacokinetic phenomena, including drug-drug interactions. This Minireview will highlight recent advancements and publications around physiologically based pharmacokinetic drug-drug interaction modeling, an important area of drug discovery and development research in light of the increasing prevalence of polypharmacology in clinical settings.
Collapse
Affiliation(s)
- Robert S Foti
- Pharmacokinetics, Dynamics, Metabolism and Bioanalytics, Merck & Co, Inc, Boston, Massachusetts.
| |
Collapse
|
5
|
Tamilarasi W, Balamurugan BJ. New reverse sum Revan indices for physicochemical and pharmacokinetic properties of anti-filovirus drugs. Front Chem 2024; 12:1486933. [PMID: 39749221 PMCID: PMC11693449 DOI: 10.3389/fchem.2024.1486933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 11/21/2024] [Indexed: 01/04/2025] Open
Abstract
Ebola and Marburg viruses, biosafety level 4 pathogens, cause severe hemorrhaging and organ failure with high mortality. Although some FDA-approved vaccines or therapeutics like Ervebo for Zaire Ebola virus exist, still there is a lack of effective therapeutics that cover all filoviruses, including both Ebola and Marburg viruses. Therefore, some anti-filovirus drugs such as Pinocembrin, Favipiravir, Remdesivir and others are used to manage infections. In theoretical chemistry, a chemical molecule is converted into an isomorphic molecular graph, G ( V , E ) by considering atom set V as vertices and bond set E as edges. A topological index is a molecular descriptor derived from the molecular graph of a chemical compound that characterizes its topology. The relationship between a compound's chemical structure and its properties is investigated through the quantitative structure-property relationship (QSPR). This article introduces new reverse sum Revan degree based indices to explore the physicochemical and pharmacokinetic properties of anti-filovirus drugs via multilinear regression. The findings reveal a strong correlation between these proposed indices and the properties of anti-filovirus drugs when compared to reverse and Revan degree-based indices. Thus, reverse sum Revan indices offer valuable insights for analyzing the drugs properties used to treat Ebola and Marburg virus infections. Moreover, the multilinear regression (MLR) results through reverse sum Revan indices are compared with Artificial Neural Network (ANN) modelling technique and it provides the better prediction of the physicochemical and pharmacokinetic properties of anti-filovirus drugs.
Collapse
Affiliation(s)
| | - B. J. Balamurugan
- Department of Mathematics, School of Advanced Sciences, Vellore Institute of Technology, Chennai, Tamil Nadu, India
| |
Collapse
|
6
|
Sise ME, Santos JR, Goldman JD, Tuttle KR, Teixeira JP, Seibert AF, Koullias Y, Llewellyn J, Regan S, Zhao Y, Huang H, Hyland RH, Osinusi A, Winter H, Humeniuk R, Hulter HN, Gottlieb RL, Fusco DN, Birne R, Stancampiano FF, Libertin CR, Small CB, Plate M, McPhail MJ. Efficacy and Safety of Remdesivir in People With Impaired Kidney Function Hospitalized for COVID-19 Pneumonia: A Randomized Clinical Trial. Clin Infect Dis 2024; 79:1172-1181. [PMID: 38913574 PMCID: PMC11581693 DOI: 10.1093/cid/ciae333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 06/06/2024] [Accepted: 06/18/2024] [Indexed: 06/26/2024] Open
Abstract
BACKGROUND Few antiviral therapies have been studied in patients with coronavirus disease 2019 (COVID-19) and kidney impairment. Herein, the efficacy, safety, and pharmacokinetics of remdesivir, its metabolites, and sulfobutylether-β-cyclodextrin excipient were evaluated in hospitalized patients with COVID-19 and severe kidney impairment. METHODS In REDPINE, a phase 3, randomized, double-blind, placebo-controlled study, participants aged ≥12 years hospitalized for COVID-19 pneumonia with acute kidney injury, chronic kidney disease, or kidney failure were randomized 2:1 to receive intravenous remdesivir (200 mg on day 1; 100 mg daily up to day 5) or placebo (enrollment from March 2021 to March 2022). The primary efficacy end point was the composite of the all-cause mortality rate or invasive mechanical ventilation rate through day 29. Safety was evaluated through day 60. RESULTS Although enrollment concluded early, 243 participants were enrolled and treated (remdesivir, n = 163; placebo, n = 80). At baseline, 90 participants (37.0%) had acute kidney injury (remdesivir, n = 60; placebo, n = 30), 64 (26.3%) had chronic kidney disease (remdesivir, n = 44; placebo, n = 20), and 89 (36.6%) had kidney failure (remdesivir, n = 59; placebo, n = 30); and 31 (12.8%) were vaccinated against COVID-19. Composite all-cause mortality or invasive mechanical ventilation rates through day 29 were 29.4% and 32.5% in the remdesivir and placebo group, respectively (P = .61). Treatment-emergent adverse events were reported in 80.4% for remdesivir versus 77.5% for placebo, and serious adverse events in 50.3% versus 50.0%, respectively. Pharmacokinetic plasma exposure to remdesivir was not affected by kidney function. CONCLUSIONS Although the study was underpowered, no significant difference in efficacy was observed between treatment groups. REDPINE demonstrated that remdesivir is safe in patients with COVID-19 and severe kidney impairment. CLINICAL TRIALS REGISTRATION EudraCT 2020-005416-22; Clinical Trials.gov NCT04745351.
Collapse
Affiliation(s)
- Meghan E Sise
- Division of Nephrology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jose Ramon Santos
- Fight Infections Foundation, Service of Infectious Diseases, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Jason D Goldman
- Swedish Center for Research and Innovation, Providence Swedish Medical Center, Seattle, Washington, USA
- Division of Allergy and Infectious Diseases, University of Washington, Seattle, Washington, USA
| | - Katherine R Tuttle
- Providence Medical Research Center, Providence Inland Northwest Health, Spokane, Washington, USA
| | - J Pedro Teixeira
- Divisions of Nephrology and Pulmonary, Critical Care, and Sleep Medicine, University of New Mexico Hospital, Albuquerque, New Mexico, USA
| | - Allan F Seibert
- Pulmonary Associates Research, Ascension Providence, Mobile, Alabama, USA
| | | | | | - Sean Regan
- Gilead Sciences, Foster City, California, USA
| | - Yang Zhao
- Gilead Sciences, Foster City, California, USA
| | | | | | - Anu Osinusi
- Gilead Sciences, Foster City, California, USA
| | | | | | - Henry N Hulter
- Department of Medicine, University of California, San Francisco, California, USA
| | - Robert L Gottlieb
- Department of Internal Medicine, Baylor University Medical Center, Dallas, Texas, USA
- Baylor Scott & White Research Institute, Dallas, Texas, USA
| | - Dahlene N Fusco
- Department of Medicine, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Rita Birne
- Department of Nephrology, Centro Hospitalar de Lisboa Ocidental, Lisbon, Portugal
- NOVA Medical School, Lisbon, Portugal
| | - Fernando F Stancampiano
- Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Jacksonville, Florida, USA
| | - Claudia R Libertin
- Department of Internal Medicine, Mayo Clinic College of Medicine and Science, Jacksonville, Florida, USA
| | - Catherine B Small
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Markus Plate
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Mark J McPhail
- Institute of Liver Studies, King's College Hospital, London, United Kingdom
| |
Collapse
|
7
|
Anoshchenko O, Abdelghany M, Lichtman A, Duan R, Chen H, Shaik NA, Peng CC, Yue Q, Subramanian R, Hyland RH, Davies S, Castellanos K, Mak L, Shen G, Xiao D, Caro L, Winter H, Llewellyn J, Humeniuk R. Pharmacokinetics, Mass Balance, Safety, and Tolerability of Obeldesivir in Healthy Participants. Clin Pharmacol Ther 2024; 116:1231-1239. [PMID: 38940465 DOI: 10.1002/cpt.3337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 05/27/2024] [Indexed: 06/29/2024]
Abstract
There is an unmet need for safe and efficacious oral therapies for COVID-19 with low potential for drug-drug interactions. Obeldesivir is an orally administered nucleoside prodrug that has shown antiviral potency in nonclinical studies against SARS-CoV-2 and its circulating variants. Obeldesivir is metabolized to the active nucleoside triphosphate (GS-443902), which acts as an inhibitor of the SARS-CoV-2 RNA-dependent RNA polymerase, thereby inhibiting viral RNA synthesis. Here, we report the safety, tolerability, and pharmacokinetics from a first-in-human, randomized, placebo-controlled, phase I study following oral administration of obeldesivir and a phase I, open-label absorption, distribution, metabolism, and excretion study following oral administration of [14C]-obeldesivir. Overall, obeldesivir was safe and well tolerated at single and multiple doses between 100 and 1,600 mg, with low potential for QT prolongation as assessed by QT-concentration analysis. The exposures to GS-441524 increased dose proportionally in the 100-900-mg dose range. GS-441524 accumulated by 35% after twice-daily and 12% after once-daily dosing for 5 days. Dose-proportional increases in the intracellular concentration of GS-443902 were also observed in peripheral blood mononuclar cells. Plasma exposure of GS-441524 was not significantly altered by food intake. Following oral administration of [14C]-obeldesivir (500 mg; 100 μCi), the mean cumulative [14C]-dose recovery was 90.7% with 58.5% in urine and 32.2% in feces. GS-441524 was the predominant plasma component (90% of 14C-area under the concentration-time curve) and was primarily eliminated via renal excretion. Collectively, data from these studies support selection of the obeldesivir 350 mg twice-daily dosing regimen for further evaluation in phase III studies for COVID-19.
Collapse
Affiliation(s)
| | | | | | - Ran Duan
- Gilead Sciences, Inc., Foster City, California, USA
| | - Henry Chen
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Chi-Chi Peng
- Gilead Sciences, Inc., Foster City, California, USA
| | - Qin Yue
- Gilead Sciences, Inc., Foster City, California, USA
| | | | | | | | | | - Lily Mak
- Gilead Sciences, Inc., Foster City, California, USA
| | - Gong Shen
- Gilead Sciences, Inc., Foster City, California, USA
| | - Deqing Xiao
- Gilead Sciences, Inc., Foster City, California, USA
| | | | - Helen Winter
- Gilead Sciences, Inc., Foster City, California, USA
| | | | | |
Collapse
|
8
|
Brooks KM, Baltrusaitis K, Clarke DF, Nachman S, Jao J, Purswani MU, Agwu A, Beneri C, Deville JG, Powis KM, Stek AM, Eke AC, Shapiro DE, Capparelli E, Greene E, George K, Yin DE, Jean-Philippe P, Chakhtoura N, Bone F, Bacon K, Johnston B, Reding C, Kersey K, Humeniuk R, Best BM, Mirochnick M, Momper JD. Pharmacokinetics and Safety of Remdesivir in Pregnant and Nonpregnant Women With COVID-19: Results From IMPAACT 2032. J Infect Dis 2024; 230:878-888. [PMID: 38839047 PMCID: PMC11481345 DOI: 10.1093/infdis/jiae298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 04/15/2024] [Accepted: 06/03/2024] [Indexed: 06/07/2024] Open
Abstract
BACKGROUND Pregnant people with coronavirus disease 2019 (COVID-19) experience higher risk for severe disease and adverse pregnancy outcomes, but no pharmacokinetic (PK) data exist to support dosing of COVID-19 therapeutics during pregnancy. We report PK and safety data for intravenous remdesivir in pregnancy. METHODS IMPAACT 2032 was a phase 4 prospective, open-label, nonrandomized opportunistic study of hospitalized pregnant and nonpregnant women receiving intravenous remdesivir as part of clinical care. Intensive PK sampling was performed on infusion days 3, 4, or 5 with collection of plasma and peripheral blood mononuclear cells (PBMCs). Safety data were recorded from first infusion through 4 weeks after last infusion and at delivery. Geometric mean ratios (GMR) (90% confidence intervals [CI]) of PK parameters between pregnant and nonpregnant women were calculated. RESULTS Fifty-three participants initiated remdesivir (25 pregnant; median gestational age, 27.6 weeks; interquartile range, 24.9-31.0 weeks). Plasma exposures of remdesivir, its 2 major metabolites (GS-704277 and GS-441524), and the free remdesivir fraction were similar between pregnant and nonpregnant participants. Concentrations of the active triphosphate (GS-443902) in PBMCs increased 2.04-fold (90% CI, 1.35-3.03) with each additional infusion in nonpregnant versus pregnant participants. Three adverse events in nonpregnant participants were related to treatment (1 grade 3; 2 grade 2 resulting in treatment discontinuation). There were no treatment-related adverse pregnancy outcomes or congenital anomalies detected. CONCLUSIONS Plasma remdesivir PK parameters were comparable between pregnant and nonpregnant women, and no safety concerns were identified based on our limited data. These findings suggest no dose adjustments are indicated for intravenous remdesivir during pregnancy. CLINICAL TRIALS REGISTRATION NCT04582266.
Collapse
Affiliation(s)
- Kristina M Brooks
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Kristin Baltrusaitis
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Diana F Clarke
- Section of Pediatric Infectious Diseases, Boston Medical Center, Boston, Massachusetts, USA
| | - Sharon Nachman
- Division of Pediatric Infectious Diseases, Stony Brook Children's Hospital, Stony Brook, New York, USA
| | - Jennifer Jao
- Division of Pediatric Infectious Diseases, Division of Adult Infectious Diseases, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Murli U Purswani
- Division of Pediatric Infectious Diseases, BronxCare Health System, Bronx, New York, USA
| | - Allison Agwu
- Department of Pediatric Infectious Disease, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Christy Beneri
- Division of Pediatric Infectious Diseases, Stony Brook Children's Hospital, Stony Brook, New York, USA
| | - Jaime G Deville
- Division of Infectious Diseases, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, USA
| | - Kathleen M Powis
- Departments of Medicine and Pediatrics, Massachusetts General Hospital, Boston, Massachusetts, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Alice M Stek
- Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Ahizechukwu C Eke
- Division of Maternal Fetal Medicine, Department of Gynecology and Obstetrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Division of Clinical Pharmacology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - David E Shapiro
- Center for Biostatistics in AIDS Research, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Edmund Capparelli
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
- Pediatrics Department, School of Medicine-Rady Children's Hospital San Diego, University of California San Diego, San Diego, California, USA
| | | | - Kathleen George
- IMPAACT Operations Center, FHI 360, Durham North Carolina, USA
| | - Dwight E Yin
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Patrick Jean-Philippe
- Division of AIDS, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Nahida Chakhtoura
- National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Frederic Bone
- Frontier Science and Technology Research Foundation, Inc, Amherst, New York, USA
| | - Kira Bacon
- Frontier Science and Technology Research Foundation, Inc, Amherst, New York, USA
| | - Benjamin Johnston
- Frontier Science and Technology Research Foundation, Inc, Amherst, New York, USA
| | - Christina Reding
- Frontier Science and Technology Research Foundation, Inc, Amherst, New York, USA
| | | | | | - Brookie M Best
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
- Pediatrics Department, School of Medicine-Rady Children's Hospital San Diego, University of California San Diego, San Diego, California, USA
| | - Mark Mirochnick
- Division of Neonatalogy, Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| | - Jeremiah D Momper
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
9
|
Kim M, Mahmood M, Estes LL, Wilson JW, Martin NJ, Marcus JE, Mittal A, O'Connell CR, Shah A. A narrative review on antimicrobial dosing in adult critically ill patients on extracorporeal membrane oxygenation. Crit Care 2024; 28:326. [PMID: 39367501 PMCID: PMC11453026 DOI: 10.1186/s13054-024-05101-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 09/14/2024] [Indexed: 10/06/2024] Open
Abstract
The optimal dosing strategy of antimicrobial agents in critically ill patients receiving extracorporeal membrane oxygenation (ECMO) is unknown. We conducted comprehensive review of existing literature on effect of ECMO on pharmacokinetics and pharmacodynamics of antimicrobials, including antibacterials, antifungals, and antivirals that are commonly used in critically ill patients. We aim to provide practical guidance to clinicians on empiric dosing strategy for these patients. Finally, we discuss importance of therapeutic drug monitoring, limitations of current literature, and future research directions.
Collapse
Affiliation(s)
- Myeongji Kim
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA.
| | - Maryam Mahmood
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| | - Lynn L Estes
- Department of Pharmacy, Mayo Clinic, Rochester, MN, USA
| | - John W Wilson
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Joseph E Marcus
- Department of Medicine, Brooke Army Medical Center, Joint Base San Antonio-Fort Sam Houston, Fort Sam Houston, TX, USA
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Ankit Mittal
- Department of Infectious Diseases, AIG Hospitals, Hyderabad, India
| | | | - Aditya Shah
- Division of Public Health, Infectious Diseases and Occupational Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
10
|
Jony MR, Ahn S. Drug-Drug Interactions between COVID-19 and Tuberculosis Medications: A Comprehensive Review of CYP450 and Transporter-Mediated Effects. Pharmaceuticals (Basel) 2024; 17:1035. [PMID: 39204140 PMCID: PMC11360778 DOI: 10.3390/ph17081035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 08/02/2024] [Accepted: 08/03/2024] [Indexed: 09/03/2024] Open
Abstract
Most medications undergo metabolism and elimination via CYP450 enzymes, while uptake and efflux transporters play vital roles in drug elimination from various organs. Interactions often occur when multiple drugs share CYP450-transporter-mediated metabolic pathways, necessitating a unique clinical care strategy to address the diverse types of CYP450 and transporter-mediated drug-drug interactions (DDI). The primary focus of this review is to record relevant mechanisms regarding DDI between COVID-19 and tuberculosis (TB) treatments, specifically through the influence of CYP450 enzymes and transporters on drug absorption, distribution, metabolism, elimination, and pharmacokinetics. This understanding empowers clinicians to prevent subtherapeutic and supratherapeutic drug levels of COVID medications when co-administered with TB drugs, thereby mitigating potential challenges and ensuring optimal treatment outcomes. A comprehensive analysis is presented, encompassing various illustrative instances of TB drugs that may impact COVID-19 clinical behavior, and vice versa. This review aims to provide valuable insights to healthcare providers, facilitating informed decision-making and enhancing patient safety while managing co-infections. Ultimately, this study contributes to the body of knowledge necessary to optimize therapeutic approaches and improve patient outcomes in the face of the growing challenges posed by infectious diseases.
Collapse
Affiliation(s)
- M. Rasheduzzaman Jony
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea;
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea
| | - Sangzin Ahn
- Department of Pharmacology and Pharmacogenomics Research Center, Inje University College of Medicine, Busan 47392, Republic of Korea;
- Center for Personalized Precision Medicine of Tuberculosis, Inje University College of Medicine, Busan 47392, Republic of Korea
| |
Collapse
|
11
|
Liu K, Li Z, Li L, Heyward S, Wang SR, He L, Wang H. Mechanistic Understanding of Dexamethasone-Mediated Protection against Remdesivir-Induced Hepatotoxicity. Mol Pharmacol 2024; 106:71-82. [PMID: 38769019 DOI: 10.1124/molpharm.124.000894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/24/2024] [Accepted: 05/01/2024] [Indexed: 05/22/2024] Open
Abstract
Remdesivir (RDV), a broad-spectrum antiviral agent, is often used together with dexamethasone (DEX) for hospitalized COVID-19 patients requiring respiratory support. Potential hepatic adverse drug reaction is a safety concern associated with the use of RDV. We previously reported that DEX cotreatment effectively mitigates RDV-induced hepatotoxicity and reduces elevated serum alanine aminotransferase and aspartate aminotransferase levels in cultured human primary hepatocytes (HPH) and hospitalized COVID-19 patients, respectively. Yet, the precise mechanism behind this protective drug-drug interaction remains largely unknown. Here, we show that through the activation of p38, c-Jun N-terminal kinase (JNK), and extracellular signal-regulated kinases 1 and 2 (ERK1/2) signaling, RDV induces apoptosis (cleavage of caspases 8, 9, and 3), autophagy (increased autophagosome and LC3-II), and mitochondrial damages (decreased membrane potential, respiration, ATP levels, and increased expression of Bax and the released cytosolic cytochrome C) in HPH. Importantly, cotreatment with DEX partially reversed RDV-induced apoptosis, autophagy, and cell death. Mechanistically, DEX deactivates/dephosphorylates p38, JNK, and ERK1/2 signaling by enhancing the expression of dual specificity protein phosphatase 1 (DUSP1), a mitogen-activated protein kinase (MAPK) phosphatase, in a glucocorticoid receptor (GR)-dependent manner. Knockdown of GR in HPH attenuates DEX-mediated DUSP1 induction, MAPK dephosphorylation, as well as protection against RDV-induced hepatotoxicity. Collectively, our findings suggest a molecular mechanism by which DEX modulates the GR-DUSP1-MAPK regulatory axis to alleviate the adverse actions of RDV in the liver. SIGNIFICANCE STATEMENT: The research uncovers the molecular mechanisms by which dexamethasone safeguards against remdesivir-associated liver damage in the context of COVID-19 treatment.
Collapse
Affiliation(s)
- Kaiyan Liu
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Zhihui Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Linhao Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Scott Heyward
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Shelley R Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Ling He
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| | - Hongbing Wang
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (K.L., Z.L., L.L., S.R.W., H.W.); BioIVT, Halethorpe, Maryland (S.H.); and Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland (L.H.)
| |
Collapse
|
12
|
Focosi D, Franchini M, Maggi F, Shoham S. COVID-19 therapeutics. Clin Microbiol Rev 2024; 37:e0011923. [PMID: 38771027 PMCID: PMC11237566 DOI: 10.1128/cmr.00119-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
SUMMARYSince the emergence of COVID-19 in 2020, an unprecedented range of therapeutic options has been studied and deployed. Healthcare providers have multiple treatment approaches to choose from, but efficacy of those approaches often remains controversial or compromised by viral evolution. Uncertainties still persist regarding the best therapies for high-risk patients, and the drug pipeline is suffering fatigue and shortage of funding. In this article, we review the antiviral activity, mechanism of action, pharmacokinetics, and safety of COVID-19 antiviral therapies. Additionally, we summarize the evidence from randomized controlled trials on efficacy and safety of the various COVID-19 antivirals and discuss unmet needs which should be addressed.
Collapse
Affiliation(s)
- Daniele Focosi
- North-Western Tuscany Blood Bank, Pisa University Hospital, Pisa, Italy
| | - Massimo Franchini
- Division of Hematology and Transfusion Medicine, Carlo Poma Hospital, Mantua, Italy
| | - Fabrizio Maggi
- National Institute for Infectious Diseases "Lazzaro Spallanzani" IRCCS, Rome, Italy
| | - Shmuel Shoham
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Hurwitz SJ, De R, LeCher JC, Downs-Bowen JA, Goh SL, Zandi K, McBrayer T, Amblard F, Patel D, Kohler JJ, Bhasin M, Dobosh BS, Sukhatme V, Tirouvanziam RM, Schinazi RF. Why Certain Repurposed Drugs Are Unlikely to Be Effective Antivirals to Treat SARS-CoV-2 Infections. Viruses 2024; 16:651. [PMID: 38675992 PMCID: PMC11053489 DOI: 10.3390/v16040651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Revised: 04/10/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Most repurposed drugs have proved ineffective for treating COVID-19. We evaluated median effective and toxic concentrations (EC50, CC50) of 49 drugs, mostly from previous clinical trials, in Vero cells. Ratios of reported unbound peak plasma concentrations, (Cmax)/EC50, were used to predict the potential in vivo efficacy. The 20 drugs with the highest ratios were retested in human Calu-3 and Caco-2 cells, and their CC50 was determined in an expanded panel of cell lines. Many of the 20 drugs with the highest ratios were inactive in human Calu-3 and Caco-2 cells. Antivirals effective in controlled clinical trials had unbound Cmax/EC50 ≥ 6.8 in Calu-3 or Caco-2 cells. EC50 of nucleoside analogs were cell dependent. This approach and earlier availability of more relevant cultures could have reduced the number of unwarranted clinical trials.
Collapse
Affiliation(s)
- Selwyn J. Hurwitz
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Ramyani De
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Julia C. LeCher
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Jessica A. Downs-Bowen
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Shu Ling Goh
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Keivan Zandi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Tamara McBrayer
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Franck Amblard
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Dharmeshkumar Patel
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - James J. Kohler
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| | - Manoj Bhasin
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Brian S. Dobosh
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Vikas Sukhatme
- Morningside Center for Innovative and Affordable Medicine, Departments of Medicine and Hematology and Oncology, Emory University School of Medicine, Atlanta, GA 30322, USA;
| | - Rabindra M. Tirouvanziam
- Center for Cystic Fibrosis & Airways Disease Research, Division of Pulmonary, Allergy & Immunology, Cystic Fibrosis and Sleep, Emory University and Children’s Healthcare of Atlanta, 2015 Uppergate Drive, Atlanta, GA 30322, USA; (M.B.); (B.S.D.); (R.M.T.)
| | - Raymond F. Schinazi
- Center for ViroScience and Cure, Laboratory of Biochemical Pharmacology, Department of Pediatrics, Emory University School of Medicine and Children’s Healthcare of Atlanta, 1760 Haygood Drive, Atlanta, GA 30322, USA; (S.J.H.); (R.D.); (J.C.L.); (J.A.D.-B.); (S.L.G.); (K.Z.); (T.M.); (F.A.); (D.P.); (J.J.K.)
| |
Collapse
|
14
|
Park S, Kim AR, Lee J, Kang SW, Sung H, Kim MN, Chang E, Bae S, Jung J, Kim MJ, Kim SH, Lee SO, Choi SH, Kim YS, Song EH, Chong YP. Clinical safety of remdesivir therapy in COVID-19 patients with renal insufficiency. J Infect Chemother 2024; 30:366-370. [PMID: 37935348 DOI: 10.1016/j.jiac.2023.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Revised: 10/17/2023] [Accepted: 10/30/2023] [Indexed: 11/09/2023]
Abstract
Though remdesivir benefits COVID-19 patients, its use in those with renal dysfunction is currently limited due to concerns about possible toxic effects of accumulated sulfobutylether-β-cyclodextrin (SBECD) on liver and kidney. We examined renal and hepatic function for a month in renally-impaired COVID-19 patients who were treated or not treated with remdesivir to assess the safety of the drug. A retrospective study was performed in adult COVID-19 patients with glomerular filtration rates of <30 ml/min/1.73 m2 at admission to a tertiary care hospital between November 2020 and March 2022. Data on serum creatinine and liver chemistry were collected serially. A total of 101 patients with impaired renal function were analyzed, comprising 64 remdesivir-treated patients and 37 who did not receive any antiviral agent. Although remdesivir-treated patients were more likely to be infected with the Omicron variant (79.7% vs. 48.6%), baseline characteristics did not differ significantly between the two groups. Among patients who initially did not require dialysis, 18.4% (7/38) of remdesivir-treated patients developed acute kidney injury (AKI) at days 4-6, compared with 51.7% (15/29) of non-remdesivir-treated patients. Liver injury severity worsened in 3.1% (2/64) of remdesivir-treated patients and 5.4% (2/37) of non-remdesivir-treated patients at days 4-6. In addition, there was no significant increase in AKI and liver injury over time in remdesivir-treated patients, and there were no cases of discontinuation of remdesivir due to adverse reactions. Concerns regarding the safety of SBECD should not lead to hasty withholding of remdesivir treatment in renally-impaired COVID-19 patients.
Collapse
Affiliation(s)
- Somi Park
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - A Reum Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiyoung Lee
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Woon Kang
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Heungsup Sung
- Department of Laboratory Meidicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Mi-Na Kim
- Department of Laboratory Meidicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Euijin Chang
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Seongman Bae
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jiwon Jung
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Min Jae Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung-Han Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Oh Lee
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sang-Ho Choi
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Yang Soo Kim
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Eun Hee Song
- Department of Internal Medicine, Gangneung Asan Hospital, University of Ulsan College of Medicine, Gangneung, Gangwon-do, Republic of Korea.
| | - Yong Pil Chong
- Department of Infectious Diseases, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
15
|
Papic I, Bistrovic P, Krecak I, Ortner Hadziabdic M, Lucijanic M. Specific adverse outcomes associated with selective serotonin reuptake inhibitors use in COVID-19 patients might be potentiated by remdesivir use. J Psychopharmacol 2024; 38:395-403. [PMID: 38481078 DOI: 10.1177/02698811241237868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/12/2024]
Abstract
BACKGROUND Due to non-consistent reports in the literature, there are uncertainties about the potential benefits and harms of selective serotonin reuptake inhibitors (SSRIs) in patients with Coronavirus disease 2019 (COVID-19). AIM To investigate associations of SSRIs with clinical characteristics and unwanted outcomes among real-life severe and critical COVID-19 patients and their relationship with remdesivir (RDV) use. METHODS This retrospective cohort study evaluated a total of 1558 COVID-19 patients of the white race treated in a tertiary center institution, among them 779 patients treated with RDV and 779 1:1 case-matched patients. RESULTS A total of 78 (5%) patients were exposed to SSRIs during hospitalization, similarly distributed among patients treated with RDV and matched patients (5.1 and 4.9%). No significant associations of SSRI use with age, sex, comorbidity burden, and COVID-19 severity were present in either of the two cohorts (p > 0.05 for all analyses). In multivariate analyses adjusted for clinically meaningful variables, SSRI use was significantly associated with higher mortality among RDV (adjusted odds ratio (aOR) 2.0, p = 0.049) and matched patients (aOR 2.22, p = 0.044) and with higher risk for mechanical-ventilation (aOR 2.57, p = 0.006), venous-thromboembolism (aOR 3.69, p = 0.007), and bacteremia (aOR 2.22, p = 0.049) among RDV treated patients. CONCLUSIONS Adverse outcomes associated with SSRI use in COVID-19 patients might be potentiated by RDV use, and clinically significant interactions between these two drug classes might exist. Although our findings raise important considerations for clinical practice, they are limited by retrospective nature of the study, lack of ethnic diversity, and the potential for unmeasured confounding factors. Future studies exploring underlying biological mechanisms are needed.
Collapse
Affiliation(s)
- Ivan Papic
- Department of Pharmacy, University hospital Dubrava, Zagreb, Croatia
| | - Petra Bistrovic
- Department of Cardiology, University hospital Dubrava, Zagreb, Croatia
| | - Ivan Krecak
- Department of Internal Medicine, General hospital of Sibenik-Knin county, Sibenik, Croatia
- Faculty of Medicine, University of Rijeka, Rijeka, Croatia
- University of Applied Sciences, Sibenik, Croatia
| | - Maja Ortner Hadziabdic
- Centre for Applied Pharmacy, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Marko Lucijanic
- Department of Hematology, University hospital Dubrava, Zagreb, Croatia
- School of Medicine, University of Zagreb, Zagreb, Croatia
| |
Collapse
|
16
|
Wang H, Luo S, Xie M, Chen Z, Zhang Y, Xie Z, Zhang Y, Zhang Y, Yang L, Wu F, Chen X, Du G, Zhao J, Sun X. ACE2 Receptor-Targeted Inhaled Nanoemulsions Inhibit SARS-CoV-2 and Attenuate Inflammatory Responses. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2311537. [PMID: 38174591 DOI: 10.1002/adma.202311537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/12/2023] [Indexed: 01/05/2024]
Abstract
Three kinds of coronaviruses are highly pathogenic to humans, and two of them mainly infect humans through Angiotensin-converting enzyme 2 (ACE2)receptors. Therefore, specifically blocking ACE2 binding at the interface with the receptor-binding domain is promising to achieve both preventive and therapeutic effects of coronaviruses. Alternatively, drug-targeted delivery based on ACE2 receptors can further improve the efficacy and safety of inhalation drugs. Here, these two approaches are innovatively combined by designing a nanoemulsion (NE) drug delivery system (termed NE-AYQ) for inhalation that targets binding to ACE2 receptors. This inhalation-delivered remdesivir nanoemulsion (termed RDSV-NE-AYQ) effectively inhibits the infection of target cells by both wild-type and mutant viruses. The RDSV-NE-AYQ strongly inhibits Severe acute respiratory syndrome coronavirus 2 at two dimensions: they not only block the binding of the virus to host cells at the cell surface but also restrict virus replication intracellularly. Furthermore, in the mouse model of acute lung injury, the inhaled drug delivery system loaded with anti-inflammatory drugs (TPCA-1-NE-AYQ) can significantly alleviate the lung tissue injury of mice. This smart combination provides a new choice for dealing with possible emergencies in the future and for the rapid development of inhaled drugs for the treatment of respiratory diseases.
Collapse
Affiliation(s)
- Hairui Wang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Shuang Luo
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Mingxin Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhao Chen
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, P. R. China
| | - Yunming Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Zhiqiang Xie
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yongshun Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Yu Zhang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Lan Yang
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Fuhua Wu
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Xiaoyan Chen
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Guangsheng Du
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| | - Jincun Zhao
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510182, P. R. China
| | - Xun Sun
- Key Laboratory of Drug Targeting and Drug Delivery Systems, Ministry of Education, West China School of Pharmacy, Sichuan University, Chengdu, 610041, P. R. China
| |
Collapse
|
17
|
Costa B, Vale N. Virus-Induced Epilepsy vs. Epilepsy Patients Acquiring Viral Infection: Unravelling the Complex Relationship for Precision Treatment. Int J Mol Sci 2024; 25:3730. [PMID: 38612542 PMCID: PMC11011490 DOI: 10.3390/ijms25073730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 01/04/2024] [Accepted: 03/25/2024] [Indexed: 04/14/2024] Open
Abstract
The intricate relationship between viruses and epilepsy involves a bidirectional interaction. Certain viruses can induce epilepsy by infecting the brain, leading to inflammation, damage, or abnormal electrical activity. Conversely, epilepsy patients may be more susceptible to viral infections due to factors, such as compromised immune systems, anticonvulsant drugs, or surgical interventions. Neuroinflammation, a common factor in both scenarios, exhibits onset, duration, intensity, and consequence variations. It can modulate epileptogenesis, increase seizure susceptibility, and impact anticonvulsant drug pharmacokinetics, immune system function, and brain physiology. Viral infections significantly impact the clinical management of epilepsy patients, necessitating a multidisciplinary approach encompassing diagnosis, prevention, and treatment of both conditions. We delved into the dual dynamics of viruses inducing epilepsy and epilepsy patients acquiring viruses, examining the unique features of each case. For virus-induced epilepsy, we specify virus types, elucidate mechanisms of epilepsy induction, emphasize neuroinflammation's impact, and analyze its effects on anticonvulsant drug pharmacokinetics. Conversely, in epilepsy patients acquiring viruses, we detail the acquired virus, its interaction with existing epilepsy, neuroinflammation effects, and changes in anticonvulsant drug pharmacokinetics. Understanding this interplay advances precision therapies for epilepsy during viral infections, providing mechanistic insights, identifying biomarkers and therapeutic targets, and supporting optimized dosing regimens. However, further studies are crucial to validate tools, discover new biomarkers and therapeutic targets, and evaluate targeted therapy safety and efficacy in diverse epilepsy and viral infection scenarios.
Collapse
Affiliation(s)
- Bárbara Costa
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| | - Nuno Vale
- PerMed Research Group, Center for Health Technology and Services Research (CINTESIS), Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal;
- CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Professor Hernâni Monteiro, 4200-319 Porto, Portugal
- Department of Community Medicine, Information and Health Decision Sciences (MEDCIDS), Faculty of Medicine, University of Porto, Rua Doutor Plácido da Costa, s/n, 4200-450 Porto, Portugal
| |
Collapse
|
18
|
Schulz A, Huynh N, Heger M, Bakir M. Adverse effects of remdesivir for the treatment of acute COVID-19 in the pediatric population: a retrospective observational study. Mol Cell Pediatr 2024; 11:2. [PMID: 38381231 PMCID: PMC10881938 DOI: 10.1186/s40348-024-00175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Accepted: 02/05/2024] [Indexed: 02/22/2024] Open
Abstract
BACKGROUND Although the severity of coronavirus disease 2019 (COVID-19) tends to be lower in children, it can still lead to severe illness, particularly among those with chronic medical conditions. While remdesivir (RDV) is one of the few approved antiviral treatments for COVID-19 in children in many countries, the available data on the safety of RDV in this population is limited. METHODS To address this knowledge gap, a multicenter study involving 65 patients retrospectively analyzed the clinical data from individuals aged <18 who were hospitalized due to severe COVID-19 (defined as SpO2 < 94% or requiring supplemental oxygen) and received at least one dose of RDV. Additionally, the study encompassed 22 patients with mild-moderate COVID-19 who were considered at high risk of developing severe disease. RESULTS Nineteen children (29%) experienced mild-to-moderate adverse events (AEs) attributed to RDV, including transaminitis in 20% of children, bradycardia in 8%, and hypotension in 5%. AEs did not require discontinuation of RDV, except in one patient who developed premature ventricular contractions. The rate of AEs did not differ between patients with severe COVID-19 and those with mild-moderate COVID-19 but at high risk for severe disease. All but one patient were discharged within 23 days of admission, and no fatalities were recorded. Among high-risk patients with mild-moderate disease, only 2 (9%) progressed to the point of needing supplemental oxygen. CONCLUSIONS Our data suggests that RDV is safe in children, with no reported serious AEs. However, the absence of a control group limits the extent to which conclusions can be drawn. RDV may contribute to clinical improvement, particularly in high-risk patients.
Collapse
Affiliation(s)
- Abigail Schulz
- University of Illinois College of Medicine at Peoria, 530 NE Glen Oak Avenue, North Building #6606, Peoria, IL, 61637, USA
| | - Natalie Huynh
- Department of Pediatrics, University of Illinois College of Medicine at Peoria, 530 NE Glen Oak Avenue, North Building #6606, Peoria, IL, 61637, USA
| | - Margaret Heger
- Department of Pharmacy, OSF HealthCare Children's Hospital of Illinois, Peoria, IL, USA
| | - Mustafa Bakir
- Department of Pediatrics, Division of Pediatric Infectious Diseases, University of Illinois College of Medicine at Peoria, 530 NE Glen Oak Avenue, North Building #6606, Peoria, IL, 61637, USA.
| |
Collapse
|
19
|
Reus P, Guthmann H, Uhlig N, Agbaria M, Issmail L, Eberlein V, Nordling-David MM, Jbara-Agbaria D, Ciesek S, Bojkova D, Cinatl J, Burger-Kentischer A, Rupp S, Zaliani A, Grunwald T, Gribbon P, Kannt A, Golomb G. Drug repurposing for the treatment of COVID-19: Targeting nafamostat to the lungs by a liposomal delivery system. J Control Release 2023; 364:654-671. [PMID: 37939853 DOI: 10.1016/j.jconrel.2023.10.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/10/2023]
Abstract
Despite tremendous global efforts since the beginning of the COVID-19 pandemic, still only a limited number of prophylactic and therapeutic options are available. Although vaccination is the most effective measure in preventing morbidity and mortality, there is a need for safe and effective post-infection treatment medication. In this study, we explored a pipeline of 21 potential candidates, examined in the Calu-3 cell line for their antiviral efficacy, for drug repurposing. Ralimetinib and nafamostat, clinically used drugs, have emerged as attractive candidates. Due to the inherent limitations of the selected drugs, we formulated targeted liposomes suitable for both systemic and intranasal administration. Non-targeted and targeted nafamostat liposomes (LipNaf) decorated with an Apolipoprotein B peptide (ApoB-P) as a specific lung-targeting ligand were successfully developed. The developed liposomal formulations of nafamostat were found to possess favorable physicochemical properties including nano size (119-147 nm), long-term stability of the normally rapidly degrading compound in aqueous solution, negligible leakage from the liposomes upon storage, and a neutral surface charge with low polydispersity index (PDI). Both nafamostat and ralimetinib liposomes showed good cellular uptake and lack of cytotoxicity, and non-targeted LipNaf demonstrated enhanced accumulation in the lungs following intranasal (IN) administration in non-infected mice. LipNaf retained its anti-SARS-CoV 2 activity in Calu 3 cells with only a modest decrease, exhibiting complete inhibition at concentrations >100 nM. IN, but not intraperitoneal (IP) treatment with targeted LipNaf resulted in a trend to reduced viral load in the lungs of K18-hACE2 mice compared to targeted empty Lip. Nevertheless, upon removal of outlier data, a statistically significant 1.9-fold reduction in viral load was achieved. This observation further highlights the importance of a targeted delivery into the respiratory tract. In summary, we were able to demonstrate a proof-of-concept of drug repurposing by liposomal formulations with anti-SARS-CoV-2 activity. The biodistribution and bioactivity studies with LipNaf suggest an IN or inhalation route of administration for optimal therapeutic efficacy.
Collapse
Affiliation(s)
- Philipp Reus
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany; Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany
| | - Hadar Guthmann
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - Nadja Uhlig
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Majd Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Leila Issmail
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Valentina Eberlein
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Mirjam M Nordling-David
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Doaa Jbara-Agbaria
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel
| | - Sandra Ciesek
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany; Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany
| | - Denisa Bojkova
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany
| | - Jindrich Cinatl
- Goethe University Frankfurt, University Hospital, Institute for Medical Virology, Paul-Ehrlich-Straße 40, 60596 Frankfurt am Main, Germany
| | - Anke Burger-Kentischer
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Steffen Rupp
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569 Stuttgart, Germany
| | - Andrea Zaliani
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Thomas Grunwald
- Fraunhofer Institute for Cell Therapy und Immunology IZI, Perlickstrasse 1, 04103 Leipzig, Germany
| | - Philip Gribbon
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Discovery Research ScreeningPort, Schnackenburgallee 114, 22525 Hamburg, Germany
| | - Aimo Kannt
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Fraunhofer Innovation Center TheraNova, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany; Institute for Clinical Pharmacology, Goethe University Frankfurt, Theodor-Stern-Kai 7, 60596 Frankfurt am Main, Germany.
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 9112001, Israel; The Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| |
Collapse
|
20
|
Wen A, Qin ARR, Tarnowski T, Ling KHJ, Zhang H, Humeniuk R, Regan S, Saquing J, Liu W, Venkatarangan L, Xiao D. Plasma Protein Binding Determination for Unstable Ester Prodrugs: Remdesivir and Tenofovir Alafenamide. J Pharm Sci 2023; 112:3224-3232. [PMID: 37722451 DOI: 10.1016/j.xphs.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 09/20/2023]
Abstract
Remdesivir (RDV) and tenofovir alafenamide (TAF) are prodrugs designed to be converted to their respective active metabolites. Plasma protein binding (PPB) determination of these prodrugs is important for patients with possible alteration of free fraction of the drugs due to plasma protein changes in renal impairment, hepatic impairment, or pregnancy. However, the prodrugs' instability in human plasma presents a challenge for accurate PPB determination. In this research work, two approaches were used in the method development and qualification for PPB assessment of RDV and TAF. For RDV, dichlorvos was used to inhibit esterase activity to stabilize the prodrug in plasma during equilibrium dialysis (ED). The impact of dichlorvos on protein binding was evaluated and determined to be insignificant by comparing the unbound fraction (fu) determined by the ED method with dichlorvos present and the fu determined by an ultrafiltration method without dichlorvos. In contrast to RDV, TAF degradation in plasma is ∼3-fold slower, and TAF stability cannot be improved by dichlorvos. Fit-for-purpose acceptance criteria for the TAF PPB method were chosen, and an ED method was developed based on these criteria. These two methods were then qualified and applied for PPB determinations in clinical studies.
Collapse
Affiliation(s)
- Anita Wen
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Ann Ran-Ran Qin
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | - Thomas Tarnowski
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | | | - Haeyoung Zhang
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | - Rita Humeniuk
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | - Sean Regan
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA
| | | | | | | | - Deqing Xiao
- Department of Clinical Pharmacology, Gilead Sciences, Foster City, CA, USA.
| |
Collapse
|
21
|
van Eijk N, Schmacke LC, Steinmetzer T, Pilgram O, Poór M, Pászti-Gere E. In vitro testing of host-targeting small molecule antiviral matriptase/TMPRSS2 inhibitors in 2D and 3D cell-based assays. Biomed Pharmacother 2023; 168:115761. [PMID: 37865989 DOI: 10.1016/j.biopha.2023.115761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/15/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023] Open
Abstract
The outbreak of coronavirus disease 2019 (COVID-19) pandemic strongly stimulated the development of small molecule antivirals selectively targeting type II transmembrane serine proteases (TTSP), required for the host-cell entry of numerous viruses. A set of 3-amidinophenylalanine derivatives (MI-21, MI-472, MI-477, MI-485, MI-1903 and MI-1904), which inhibit the cleavage of certain viral glycoproteins was characterized in 2D and 3D primary human hepatocyte models on collagen- and Matrigel-coating using a CCK-8 assay to evaluate their cytotoxicity, a resorufin-based method to detect redox imbalances, fluorescence and ultrafiltration experiments to evaluate their interactions with human serum albumin (HSA) and α-acidic glycoprotein (AGP), and luminescence measurement to assess CYP3A4 modulation. For elucidation of selectivity of the applied compounds towards matriptase, transmembrane serine protease 2 (TMPRRS2), thrombin and factor Xa (FXa) Ki values were determined. It was proven that cell viability was only deteriorated by inhibitor MI-1903, and redox status was not influenced by administration of the selected inhibitors at 50 µM for 24 h. MI-472 and MI-477 formed relatively stable complexes with AGP. CYP3A4 inhibition was found to be strong in PHHs exposed to all inhibitors with the exception of MI-21, which seems to be a promising drug candidate also due to its better selectivity towards matriptase and TMPRSS2 over the blood clotting proteases thrombin and FXa. Our in vitro pharmacokinetic screening with these inhibitors helps to select the compounds with the best selectivity and safety profile suitable for a further preclinical characterization without animal sacrifice.
Collapse
Affiliation(s)
- Nicholas van Eijk
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary
| | - Luna C Schmacke
- Institute of Pharmaceutical Chemistry, Department of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Torsten Steinmetzer
- Institute of Pharmaceutical Chemistry, Department of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Oliver Pilgram
- Institute of Pharmaceutical Chemistry, Department of Pharmacy, Philipps University Marburg, Marbacher Weg 6-10, Marburg 35037, Germany
| | - Miklós Poór
- Department of Pharmacology, Faculty of Pharmacy, University of Pécs, Rókus u. 2, Pécs H-7624, Hungary; Lab-on-a-Chip Research Group, János Szentágothai Research Centre, University of Pécs, Ifjúság útja 20, Pécs H-7624, Hungary
| | - Erzsébet Pászti-Gere
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, István utca 2, Budapest H-1078, Hungary.
| |
Collapse
|
22
|
Pitsillou E, Beh RC, Liang JJ, Tang TS, Zhou X, Siow YY, Ma Y, Hu Z, Wu Z, Hung A, Karagiannis TC. EpiMed Coronabank Chemical Collection: Compound selection, ADMET analysis, and utilisation in the context of potential SARS-CoV-2 antivirals. J Mol Graph Model 2023; 125:108602. [PMID: 37597309 DOI: 10.1016/j.jmgm.2023.108602] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 08/08/2023] [Accepted: 08/13/2023] [Indexed: 08/21/2023]
Abstract
Antiviral drugs are important for the coronavirus disease 2019 (COVID-19) response, as vaccines and antibodies may have reduced efficacy against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) variants. Antiviral drugs that have been made available for use, albeit with questionable efficacy, include remdesivir (Veklury®), nirmatrelvir-ritonavir (Paxlovid™), and molnupiravir (Lagevrio®). To expand the options available for COVID-19 and prepare for future pandemics, there is a need to investigate new uses for existing drugs and design novel compounds. To support these efforts, we have created a comprehensive library of 750 molecules that have been sourced from in vitro, in vivo, and in silico studies. It is publicly available at our dedicated website (https://epimedlab.org/crl/). The EpiMed Coronabank Chemical Collection consists of compounds that have been divided into 10 main classes based on antiviral properties, as well as the potential to be used for the management, prevention, or treatment of COVID-19 related complications. A detailed description of each compound is provided, along with the molecular formula, canonical SMILES, and U.S. Food and Drug Administration approval status. The chemical structures have been obtained and are available for download. Moreover, the pharmacokinetic properties of the ligands have been characterised. To demonstrate an application of the EpiMed Coronabank Chemical Collection, molecular docking was used to evaluate the binding characteristics of ligands against SARS-CoV-2 nonstructural and accessory proteins. Overall, our database can be used to aid the drug repositioning process, and for gaining further insight into the molecular mechanisms of action of potential compounds of interest.
Collapse
Affiliation(s)
- Eleni Pitsillou
- Epigenomic Medicine Laboratory at prospED, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Raymond C Beh
- Epigenomic Medicine Laboratory at prospED, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Julia J Liang
- Epigenomic Medicine Laboratory at prospED, Carlton, VIC, 3053, Australia; School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Thinh Sieu Tang
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Xun Zhou
- Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Ya Yun Siow
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Yinghao Ma
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Zifang Hu
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Zifei Wu
- Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia
| | - Andrew Hung
- School of Science, STEM College, RMIT University, VIC, 3001, Australia
| | - Tom C Karagiannis
- Epigenomic Medicine Laboratory at prospED, Carlton, VIC, 3053, Australia; Department of Microbiology and Immunology, The University of Melbourne, Parkville, VIC, 3010, Australia; Department of Clinical Pathology, The University of Melbourne, Parkville, VIC, 3010, Australia.
| |
Collapse
|
23
|
Khalil A, Mohamed A, Hassan M, Magboul S, Ali H, Elmasoudi AS, Ellithy K, Qusad M, Alhothi A, Al Maslamani E, Al Amri M, Soliman A. Efficacy and Safety of Remdesivir in Hospitalized Pediatric COVID-19: A Retrospective Case-Controlled Study. Ther Clin Risk Manag 2023; 19:949-958. [PMID: 38023628 PMCID: PMC10680468 DOI: 10.2147/tcrm.s432565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction While most children experience mild coronavirus disease 2019 (COVID-19) infections, a minority of cases progress to severe or critical illness. This study aimed to assess the efficacy and safety of Remdesivir (RDV) therapy in children with moderate to severe COVID-19, enhancing clinical decision-making and expanding our understanding of antiviral treatments for pediatric patients. Methods The study included 60 patients, 38 receiving RDV treatment and 22 serving as the control group. Data was collected retrospectively from January 2021 to January 2022 through electronic hospital records. Results Regarding the main clinical symptoms reported, most patients experienced Upper Respiratory Tract Infections (93.3%), indicating respiratory involvement. Additional symptoms included Central Nervous System (11.7%) and Gastrointestinal (10.0%). Among the 38 cases in the RDV group included in the study, the adverse effects associated with using RDV: Hypoalbuminemia in 19 cases (50.0%) and anemia in 18 cases (47.4%), making them the most common adverse effects. Only one case in the RDV group experienced non-RDV-related death with a different clinical diagnosis. The results showed that RDV treatment was well-tolerated in pediatric patients, with no significant differences in hospital stay and oxygen treatment compared to the control group with P values (0.2, 0.18), respectively. Conclusion The outcomes indicate that Remdesivir may represent a safe and therapeutic choice for children with coronavirus disease 2019 (COVID-19).
Collapse
Affiliation(s)
- Ahmed Khalil
- Section of Pediatric Clinical Pharmacy, Pharmacy Department, Hamad General Hospital, Doha, Qatar
| | - Asmaa Mohamed
- Section of Pediatric Clinical Pharmacy, Pharmacy Department, Hamad General Hospital, Doha, Qatar
| | - Manasik Hassan
- Section of Academic General Pediatrics, Department of Pediatrics, Hamad General Hospital, Doha, Qatar
| | - Samar Magboul
- Section of Academic General Pediatrics, Department of Pediatrics, Hamad General Hospital, Doha, Qatar
| | - Hossamaldein Ali
- Section of Pediatric Clinical Pharmacy, Pharmacy Department, Hamad General Hospital, Doha, Qatar
| | - Ahmed Salah Elmasoudi
- Section of Pediatric Clinical Pharmacy, Pharmacy Department, Hamad General Hospital, Doha, Qatar
| | - Khaled Ellithy
- Section of Pediatric Intensive Care Unit, Department of Pediatrics, Hamad General Hospital, Doha, Qatar
| | - Mohammad Qusad
- Section of Academic General Pediatrics, Department of Pediatrics, Hamad General Hospital, Doha, Qatar
| | - Abdulla Alhothi
- Section of Academic General Pediatrics, Department of Pediatrics, Hamad General Hospital, Doha, Qatar
| | - Eman Al Maslamani
- Section of Infectious Diseases, Department of Pediatrics, Sidra Medicine, Doha, Qatar
| | | | - Ashraf Soliman
- Department of Pediatrics, Hamad General Hospital, Doha, Qatar
| |
Collapse
|
24
|
Humeniuk R, Juneja K, Chen S, Ellis S, Anoshchenko O, Xiao D, Share A, Johnston M, Davies S, DeZure A, Llewellyn J, Osinusi A, Winter H, Girish S, Palaparthy R, Dresser M. Pharmacokinetics, safety, and tolerability of inhaled remdesivir in healthy participants. Clin Transl Sci 2023; 16:2276-2288. [PMID: 37688349 PMCID: PMC10651641 DOI: 10.1111/cts.13627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/10/2023] Open
Abstract
Intravenous remdesivir (RDV) is US Food and Drug Administration-approved for hospitalized and nonhospitalized individuals with coronavirus disease 2019. RDV undergoes intracellular metabolic activation to form the active triphosphate, GS-443902, and other metabolites. Alternative administration routes, including localized pulmonary delivery, can lower systemic exposure and maximize exposure at the site of action. This study evaluated the pharmacokinetics (PK) and safety of inhaled RDV in healthy adults. This phase Ia, randomized, placebo-controlled study evaluated inhaled RDV in healthy participants randomized 4:1 to receive RDV or placebo as single doses (4 cohorts) or multiple once-daily doses (3 cohorts). Doses in cohorts 1-6 were administered as an aerosolized solution for inhalation through a sealed facemask; doses in cohort 7 were administered as an aerosolized solution for inhalation through a mouthpiece. Safety was assessed throughout the study. Seventy-two participants were enrolled (inhaled RDV, n = 58 and placebo, n = 14). Following single RDV doses, RDV, GS-704277, and GS-441524 plasma PK parameters indicated dose-proportional increases in area under the concentration-time curve (AUC) extrapolated to infinite time, AUC from time zero to last quantifiable concentration, and maximum observed concentration. Analyte plasma concentrations after multiple RDV doses were consistent with those for single-dose RDV. Analyte plasma exposures were lower when RDV was administered with a mouthpiece versus a sealed facemask. The most common adverse events included nausea, dizziness, and cough. Single- and multiple-dose inhaled RDV exhibited linear and dose-proportional plasma PK. Administration of RDV via inhalation was generally safe and well-tolerated.
Collapse
Affiliation(s)
| | | | | | - Scott Ellis
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | | | - Deqing Xiao
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | - Aaron Share
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | | | | | - Adam DeZure
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | | | - Anu Osinusi
- Gilead Sciences, Inc.Foster CityCaliforniaUSA
| | | | | | | | | |
Collapse
|
25
|
Fukao K, Nobori H, Kuroda T, Baba K, Matsumoto K, Tanaka Y, Tachibana Y, Kato T, Shishido T. Pharmacokinetic and Pharmacodynamic Analysis of the 3CL Protease Inhibitor Ensitrelvir in a SARS-CoV-2 Infection Mouse Model. Viruses 2023; 15:2052. [PMID: 37896829 PMCID: PMC10612060 DOI: 10.3390/v15102052] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/28/2023] [Accepted: 09/29/2023] [Indexed: 10/29/2023] Open
Abstract
The small-molecule antiviral drug ensitrelvir targets the 3C-like protease of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This study evaluated its inhibitory effect on viral replication in a delayed-treatment mouse model and investigated the relationship between pharmacokinetic (PK) parameters and pharmacodynamic (PD) effects. SARS-CoV-2 gamma-strain-infected BALB/c mice were orally treated with various doses of ensitrelvir starting 24 h post-infection. Effectiveness was determined 48 h after first administration based on lung viral titers. Ensitrelvir PK parameters were estimated from previously reported plasma concentration data and PK/PD analyses were performed. Ensitrelvir doses ≥ 16 mg/kg once daily, ≥8 mg/kg twice daily, or ≥8 mg/kg thrice daily for two days significantly reduced lung viral titers compared to that of the vehicle. PK/PD analyses revealed that mean AUC0-48h post-first administration, plasma concentration 48 h post-first administration (C48h), and total time above the target plasma concentration (TimeHigh) were PK parameters predictive of viral titer reduction. In conclusion, ensitrelvir dose-dependently reduced lung SARS-CoV-2 titers in mice, suggesting it inhibited viral replication. PK parameters C48h and TimeHigh were associated with sustained ensitrelvir plasma concentrations and correlated with the reduced viral titers. The findings suggest that maintaining ensitrelvir plasma concentration is effective for exerting antiviral activity against SARS-CoV-2.
Collapse
Affiliation(s)
- Keita Fukao
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Haruaki Nobori
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Takayuki Kuroda
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Kaoru Baba
- Research Area for Drug Candidate Generation II, Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Kazumi Matsumoto
- Research Area for Drug Candidate Generation II, Shionogi TechnoAdvance Research Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Yukari Tanaka
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Yuki Tachibana
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Teruhisa Kato
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| | - Takao Shishido
- Pharmaceutical Research Division, Shionogi & Co., Ltd., 1-1, Futaba-cho 3-chome, Toyonaka 561-0825, Osaka, Japan
| |
Collapse
|
26
|
Hau RK, Wright SH, Cherrington NJ. Addressing the Clinical Importance of Equilibrative Nucleoside Transporters in Drug Discovery and Development. Clin Pharmacol Ther 2023; 114:780-794. [PMID: 37404197 PMCID: PMC11347013 DOI: 10.1002/cpt.2984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 05/30/2023] [Indexed: 07/06/2023]
Abstract
The US Food and Drug Administration (FDA), European Medicines Agency (EMA), and Pharmaceuticals and Medical Devices Agency (PMDA) guidances on small-molecule drug-drug interactions (DDIs), with input from the International Transporter Consortium (ITC), recommend the evaluation of nine drug transporters. Although other clinically relevant drug uptake and efflux transporters have been discussed in ITC white papers, they have been excluded from further recommendation by the ITC and are not included in current regulatory guidances. These include the ubiquitously expressed equilibrative nucleoside transporters (ENT) 1 and ENT2, which have been recognized by the ITC for their potential role in clinically relevant nucleoside analog drug interactions for patients with cancer. Although there is comparatively limited clinical evidence supporting their role in DDI risk or other adverse drug reactions (ADRs) compared with the nine highlighted transporters, several in vitro and in vivo studies have identified ENT interactions with non-nucleoside/non-nucleotide drugs, in addition to nucleoside/nucleotide analogs. Some noteworthy examples of compounds that interact with ENTs include cannabidiol and selected protein kinase inhibitors, as well as the nucleoside analogs remdesivir, EIDD-1931, gemcitabine, and fialuridine. Consequently, DDIs involving the ENTs may be responsible for therapeutic inefficacy or off-target toxicity. Evidence suggests that ENT1 and ENT2 should be considered as transporters potentially involved in clinically relevant DDIs and ADRs, thereby warranting further investigation and regulatory consideration.
Collapse
Affiliation(s)
- Raymond K Hau
- Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, USA
| | - Stephen H Wright
- Department of Physiology, College of Medicine, The University of Arizona, Tucson, Arizona, USA
| | - Nathan J Cherrington
- Department of Pharmacology & Toxicology, College of Pharmacy, The University of Arizona, Tucson, Arizona, USA
| |
Collapse
|
27
|
DeFoor N, Paul S, Li S, Basso EKG, Stevenson V, Browning JL, Prater AK, Brindley S, Tao G, Pickrell AM. Remdesivir increases mtDNA copy number causing mild alterations to oxidative phosphorylation. Sci Rep 2023; 13:15339. [PMID: 37714940 PMCID: PMC10504289 DOI: 10.1038/s41598-023-42704-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/13/2023] [Indexed: 09/17/2023] Open
Abstract
SARS-CoV-2 causes the severe respiratory disease COVID-19. Remdesivir (RDV) was the first fast-tracked FDA approved treatment drug for COVID-19. RDV acts as an antiviral ribonucleoside (adenosine) analogue that becomes active once it accumulates intracellularly. It then diffuses into the host cell and terminates viral RNA transcription. Previous studies have shown that certain nucleoside analogues unintentionally inhibit mitochondrial RNA or DNA polymerases or cause mutational changes to mitochondrial DNA (mtDNA). These past findings on the mitochondrial toxicity of ribonucleoside analogues motivated us to investigate what effects RDV may have on mitochondrial function. Using in vitro and in vivo rodent models treated with RDV, we observed increases in mtDNA copy number in Mv1Lu cells (35.26% increase ± 11.33%) and liver (100.27% increase ± 32.73%) upon treatment. However, these increases only resulted in mild changes to mitochondrial function. Surprisingly, skeletal muscle and heart were extremely resistant to RDV treatment, tissues that have preferentially been affected by other nucleoside analogues. Although our data suggest that RDV does not greatly impact mitochondrial function, these data are insightful for the treatment of RDV for individuals with mitochondrial disease.
Collapse
Affiliation(s)
- Nicole DeFoor
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Swagatika Paul
- Graduate Program in Biomedical and Veterinary Sciences, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Shuang Li
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Erwin K Gudenschwager Basso
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Valentina Stevenson
- Virginia Tech Animal Laboratory Services, Virginia-Maryland College of Veterinary Medicine, Blacksburg, VA, 24061, USA
| | - Jack L Browning
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Anna K Prater
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Samantha Brindley
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC, 29425, USA
| | - Alicia M Pickrell
- School of Neuroscience, Virginia Tech, Life Science I Room 217, 970 Washington Street SW, Blacksburg, VA, 24061, USA.
| |
Collapse
|
28
|
Mackman RL, Kalla RV, Babusis D, Pitts J, Barrett KT, Chun K, Du Pont V, Rodriguez L, Moshiri J, Xu Y, Lee M, Lee G, Bleier B, Nguyen AQ, O'Keefe BM, Ambrosi A, Cook M, Yu J, Dempah KE, Bunyan E, Riola NC, Lu X, Liu R, Davie A, Hsiang TY, Dearing J, Vermillion M, Gale M, Niedziela-Majka A, Feng JY, Hedskog C, Bilello JP, Subramanian R, Cihlar T. Discovery of GS-5245 (Obeldesivir), an Oral Prodrug of Nucleoside GS-441524 That Exhibits Antiviral Efficacy in SARS-CoV-2-Infected African Green Monkeys. J Med Chem 2023; 66:11701-11717. [PMID: 37596939 PMCID: PMC11556372 DOI: 10.1021/acs.jmedchem.3c00750] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2023]
Abstract
Remdesivir 1 is an phosphoramidate prodrug that releases the monophosphate of nucleoside GS-441524 (2) into lung cells, thereby forming the bioactive triphosphate 2-NTP. 2-NTP, an analog of ATP, inhibits the SARS-CoV-2 RNA-dependent RNA polymerase replication and transcription of viral RNA. Strong clinical results for 1 have prompted interest in oral approaches to generate 2-NTP. Here, we describe the discovery of a 5'-isobutyryl ester prodrug of 2 (GS-5245, Obeldesivir, 3) that has low cellular cytotoxicity and 3-7-fold improved oral delivery of 2 in monkeys. Prodrug 3 is cleaved presystemically to provide high systemic exposures of 2 that overcome its less efficient metabolism to 2-NTP, leading to strong SARS-CoV-2 antiviral efficacy in an African green monkey infection model. Exposure-based SARS-CoV-2 efficacy relationships resulted in an estimated clinical dose of 350-400 mg twice daily. Importantly, all SARS-CoV-2 variants remain susceptible to 2, which supports development of 3 as a promising COVID-19 treatment.
Collapse
Affiliation(s)
- Richard L Mackman
- Medicinal Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Rao V Kalla
- Medicinal Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Darius Babusis
- Drug Metabolism, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Jared Pitts
- Discovery Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Kimberly T Barrett
- Formulation and Process Development, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Kwon Chun
- Medicinal Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Venice Du Pont
- Discovery Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Lauren Rodriguez
- Clinical Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Jasmine Moshiri
- Clinical Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Yili Xu
- Biochemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Michael Lee
- Biology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Gary Lee
- Biology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Blake Bleier
- Formulation and Process Development, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Anh-Quan Nguyen
- Formulation and Process Development, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - B Michael O'Keefe
- Process Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Andrea Ambrosi
- Process Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Meredith Cook
- Process Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Joy Yu
- Process Chemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Kassibla Elodie Dempah
- Process Development, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Elaine Bunyan
- Process Development, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Nicholas C Riola
- Discovery Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Xianghan Lu
- Discovery Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Renmeng Liu
- Drug Metabolism, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Ashley Davie
- Drug Metabolism, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Tien-Ying Hsiang
- Center for Innate Immunity and Immune Disease, Department of Immunology, School of Medicine, University of Washington, Seattle, Washington 98109 United States
| | - Justin Dearing
- Lovelace Biomedical Research Institute, 2425 Ridgecrest Drive Southeast, Albuquerque, New Mexico 87108 United States
| | - Meghan Vermillion
- Lovelace Biomedical Research Institute, 2425 Ridgecrest Drive Southeast, Albuquerque, New Mexico 87108 United States
| | - Michael Gale
- Center for Innate Immunity and Immune Disease, Department of Immunology, School of Medicine, University of Washington, Seattle, Washington 98109 United States
| | - Anita Niedziela-Majka
- Biology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Joy Y Feng
- Biochemistry, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Charlotte Hedskog
- Clinical Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - John P Bilello
- Discovery Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Raju Subramanian
- Drug Metabolism, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| | - Tomas Cihlar
- Discovery Virology, Gilead Sciences Incorporated, 333 Lakeside Drive, Foster City, California 94404 United States
| |
Collapse
|
29
|
Blair HA. Remdesivir: A Review in COVID-19. Drugs 2023; 83:1215-1237. [PMID: 37589788 PMCID: PMC10474216 DOI: 10.1007/s40265-023-01926-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/25/2023] [Indexed: 08/18/2023]
Abstract
Remdesivir (Veklury®), a nucleotide analogue prodrug with broad-spectrum antiviral activity, is approved for the treatment of coronavirus disease 2019 (COVID-19), the illness caused by severe acute respiratory syndrome coronavirus 2 infection. Unlike some antivirals, remdesivir has a low potential for drug-drug interactions. In the pivotal ACTT-1 trial in hospitalized patients with COVID-19, daily intravenous infusions of remdesivir significantly reduced time to recovery relative to placebo. Subsequent trials provided additional support for the efficacy of remdesivir in hospitalized patients with moderate or severe COVID-19, with a greater benefit seen in patients with minimal oxygen requirements at baseline. Clinical trials also demonstrated the efficacy of remdesivir in other patient populations, including outpatients at high risk for progression to severe COVID-19, as well as hospitalized paediatric patients. In terms of mortality, results were equivocal. However, remdesivir appeared to have a small mortality benefit in hospitalized patients who were not already being ventilated at baseline. Remdesivir was generally well tolerated in clinical trials, but pharmacovigilance data found an increased risk of hepatic, renal and cardiovascular adverse drug reactions in the real-world setting. In conclusion, remdesivir represents a useful treatment option for patients with COVID-19, particularly those who require supplemental oxygen.
Collapse
Affiliation(s)
- Hannah A Blair
- Springer Nature, Private Bag 65901, Mairangi Bay, Auckland, 0754, New Zealand.
| |
Collapse
|
30
|
Lucijanic M, Bistrovic P, Jordan A, Mihaljevic I, Bukvic S, Kovacevic S, Ranilovic D, Sakota S, Vlasac Glasnovic J, Delic-Brkljacic D. Remdesivir use in severe and critical COVID-19 patients might be associated with lower incidence of arterial thrombotic events. Am J Emerg Med 2023; 70:41-45. [PMID: 37201450 PMCID: PMC10170894 DOI: 10.1016/j.ajem.2023.05.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/04/2023] [Accepted: 05/07/2023] [Indexed: 05/20/2023] Open
Abstract
INTRODUCTION Venous thromboembolism (VTE) and arterial thrombotic (AT) events are a striking feature of severe COVID-19, however, relationship of remdesivir use and the risk of thrombotic events is unknown and has not been investigated before. METHODS We retrospectively analyzed a cohort of 876 consecutive hospitalized severe and critical COVID-19 patients who were treated with remdesivir and compared them to 876 case-matched control patients. All patients were treated in our tertiary-level institution in period from 10/2020 to 6/2021. VTE and AT were diagnosed by objective imaging and laboratory methods. RESULTS After exclusion of 71 VTE and 37 AT events present at the time of hospital admission, there were a total of 70 VTE (35 in the remdesivir and 35 in the control group) and 38 AT events occurring during hospitalization (13 in the remdesivir and 25 in the control group). There was a similar cumulative post-admission VTE incidence among both remdesivir and matched control patients (P = 0.287). Significantly lower cumulative post-admission AT incidence was observed among patients treated with remdesivir than among matched control patients (1.7% vs 3.3%, HR = 0.51, P = 0.035). Tendency for lower AT rates was evident in subgroups of patients stratified according to the type of AT, as well as according to the intensity of required oxygen supplementation at the time of remdesivir use. CONCLUSION Remdesivir use in severe and critical COVID-19 patients might be associated with lower occurrence of AT during hospitalization, whereas similar rates of VTE events were observed among both patients treated with remdesivir and control patients.
Collapse
Affiliation(s)
- Marko Lucijanic
- Hematology department, University hospital Dubrava, Zagreb, Croatia; School of Medicine University of Zagreb, Zagreb, Croatia.
| | - Petra Bistrovic
- Cardiology department, University hospital Dubrava, Zagreb, Croatia
| | - Ana Jordan
- Cardiology department, University hospital Dubrava, Zagreb, Croatia
| | - Iva Mihaljevic
- Neurology department, University hospital Dubrava, Zagreb, Croatia
| | - Stela Bukvic
- Neurology department, University hospital Dubrava, Zagreb, Croatia
| | - Stjepan Kovacevic
- Internal medicine department, General hospital dr. Josip Bencevic, Slavonski Brod, Croatia
| | - Darjan Ranilovic
- Pulmonology department, University hospital Dubrava, Zagreb, Croatia
| | - Sara Sakota
- Intensive medicine department, University hospital Dubrava, Zagreb, Croatia
| | | | - Diana Delic-Brkljacic
- School of Medicine University of Zagreb, Zagreb, Croatia; Cardiology department, University hospital center Sisters of mercy, Zagreb, Croatia
| |
Collapse
|
31
|
Ye L, Fan S, Zhao P, Wu C, Liu M, Hu S, Wang P, Wang H, Bi H. Potential herb‒drug interactions between anti-COVID-19 drugs and traditional Chinese medicine. Acta Pharm Sin B 2023; 13:S2211-3835(23)00203-4. [PMID: 37360014 PMCID: PMC10239737 DOI: 10.1016/j.apsb.2023.06.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 03/21/2023] [Accepted: 04/20/2023] [Indexed: 06/28/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has spread worldwide. Effective treatments against COVID-19 remain urgently in need although vaccination significantly reduces the incidence, hospitalization, and mortality. At present, antiviral drugs including Nirmatrelvir/Ritonavir (PaxlovidTM), Remdesivir, and Molnupiravir have been authorized to treat COVID-19 and become more globally available. On the other hand, traditional Chinese medicine (TCM) has been used for the treatment of epidemic diseases for a long history. Currently, various TCM formulae against COVID-19 such as Qingfei Paidu decoction, Xuanfei Baidu granule, Huashi Baidu granule, Jinhua Qinggan granule, Lianhua Qingwen capsule, and Xuebijing injection have been widely used in clinical practice in China, which may cause potential herb-drug interactions (HDIs) in patients under treatment with antiviral drugs and affect the efficacy and safety of medicines. However, information on potential HDIs between the above anti-COVID-19 drugs and TCM formulae is lacking, and thus this work seeks to summarize and highlight potential HDIs between antiviral drugs and TCM formulae against COVID-19, and especially pharmacokinetic HDIs mediated by metabolizing enzymes and/or transporters. These well-characterized HDIs could provide useful information on clinical concomitant medicine use to maximize clinical outcomes and minimize adverse and toxic effects.
Collapse
Affiliation(s)
- Ling Ye
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shicheng Fan
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Pengfei Zhao
- Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Chenghua Wu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Menghua Liu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Shuang Hu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Peng Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Hongyu Wang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Huichang Bi
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
32
|
Phan A, Avila H, MacKay JA. Biomimetic SARS-CoV-2 Spike Protein Nanoparticles. Biomacromolecules 2023; 24:2030-2041. [PMID: 37001147 PMCID: PMC10084924 DOI: 10.1021/acs.biomac.2c01465] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 03/21/2023] [Indexed: 04/03/2023]
Abstract
COVID-19 is an infectious respiratory disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). This virus contains a crucial coat protein that engages with target cells via a receptor binding domain (RBD) on its spike protein. To better study the RBD and its therapeutic opportunities, we genetically engineered a simple fusion with a thermo-responsive elastin-like polypeptide (ELP). These fusions express in Escherichia coli at a high yield in the soluble fraction and were easily purified using ELP-mediated phase separation (79 mg/L culture). Interestingly, they assembled peptide-based nanoparticles (Rh = 71.4 nm), which was attributed to oligomerization of RBDs (25.3 kDa) counterbalanced by steric stabilization by a soluble ELP (73.4 kDa). To investigate their biophysical properties, we explored the size, shape, and binding affinity for the human angiotensin-converting enzyme 2 (hACE2) and cellular uptake. Biomimetic nanoparticles such as these may enable future strategies to target the same cells, tissues, and cell-surface receptors as those harnessed by SARS-CoV-2.
Collapse
Affiliation(s)
- Alvin Phan
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
| | - Hugo Avila
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
| | - J. Andrew MacKay
- Department of Pharmacology and Pharmaceutical Sciences, USC Alfred E. Mann School of Pharmacy and Pharmaceutical Sciences, University of Southern California, 1985 Zonal Avenue, Los Angeles, CA 90089, United States
- Department of Ophthalmology, Keck School of Medicine of USC, University of Southern California, 1450 San Pablo Street, Los Angeles, CA 90033, United States
- Alfred E. Mann Department of Biomedical Engineering, USC Viterbi School of Engineering, University of Southern California, 1042 Downey Way, Los Angeles, CA 90089, United States
| |
Collapse
|
33
|
Ayyashi M, Darbashi H, Hakami A, Sharahili F. Evaluation of Remdesivir Utilization Pattern in Critically Ill Patients With COVID-19 in Jazan Province. Cureus 2023; 15:e36247. [PMID: 37069861 PMCID: PMC10105617 DOI: 10.7759/cureus.36247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/16/2023] [Indexed: 03/17/2023] Open
Abstract
Background Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the virus that causes coronavirus disease 2019 (COVID-19), has spread around the world, spurring the biomedical community to find and create antiviral therapies. The agent remdesivir, which has undergone a protracted and tortuous developmental path, is one potential therapeutic strategy now being assessed in several clinical trials. A broad-spectrum antiviral drug called remdesivir has already shown antiviral effects against filoviruses. Remdesivir was suggested as an exploratory medicine early in the pandemic because in vitro tests showed it to have antiviral effectiveness against SARS-CoV-2. Methods We conducted a retrospective cohort study that examined patient data captured through an electronic medical system at the Abu Arish General Hospital between 2021 and 2022. Data analysis was performed with SPSS version 25.0 (Armonk, NY: IBM Corp.). Results A total of 88 patients were included in this study. With the usage of remdesivir, our risk model is able to forecast adverse events and the case fatality rate. In contrast to D-dimer and c-reactive proteins, we showed that alanine transaminase (ALT), aspartate aminotransferase (AST), serum creatinine, and hemoglobin are relevant variables. Conclusion Our risk model can predict the adverse reactions and case fatality rate with the use of remdesivir. We demonstrated ALT, AST, serum creatinine, and hemoglobin as important variables rather than D-dimer and c-reactive proteins.
Collapse
|
34
|
Kanagala SG, Dholiya H, Jhajj P, Patel MA, Gupta V, Gupta S, Wu SI, Jain R. Remdesivir-Induced Bradycardia. South Med J 2023; 116:317-320. [PMID: 36863055 PMCID: PMC9973350 DOI: 10.14423/smj.0000000000001519] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2023]
Abstract
Remdesivir, a viral RNA-dependent RNA polymerase inhibitor, found extensive use in coronavirus disease 2019-infected patients because it curbs the viral load expansion. Among patients hospitalized as a result of lower respiratory tract infection, remdesivir proved to improve recovery time; however, remdesivir also can induce significant cytotoxic effects on cardiac myocytes. In this narrative review, we discuss the pathophysiological mechanism of remdesivir-induced bradycardia and diagnostic and management strategies for these patients. We conclude that further research is necessary to understand better the mechanism of bradycardia in coronavirus disease 2019 patients with or without cardiovascular disorder treated with remdesivir.
Collapse
Affiliation(s)
| | | | - Poonam Jhajj
- Avalon University School of Medicine, Willemstad, Curacao
| | | | - Vasu Gupta
- Dayanand Medical College and Hospital, Ludhiana, India
| | - Sachin Gupta
- Department of Internal Medicine, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Shiau-ing Wu
- Department of Internal Medicine, Penn State University Milton S. Hershey Medical Center, Hershey, Pennsylvania
| | - Rohit Jain
- Avalon University School of Medicine, Willemstad, Curacao,Associate Professor, Avalon University School of Medicine, Willemstad, Curacao
| |
Collapse
|
35
|
Tamura R, Irie K, Nakagawa A, Muroi H, Eto M, Ikesue H, Muroi N, Fukushima S, Tomii K, Hashida T. Population pharmacokinetics and exposure-clinical outcome relationship of remdesivir major metabolite GS-441524 in patients with moderate and severe COVID-19. CPT Pharmacometrics Syst Pharmacol 2023; 12:513-521. [PMID: 36798006 PMCID: PMC10088080 DOI: 10.1002/psp4.12936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 01/30/2023] [Accepted: 01/31/2023] [Indexed: 02/18/2023] Open
Abstract
Although remdesivir, a prodrug of nucleoside analog (GS-441524), has demonstrated clinical benefits in coronavirus disease 2019 (COVID-19) treatment, its pharmacokinetics (PKs) in patients with COVID-19 remain poorly understood. Therefore, in this study, the PKs of remdesivir and its major metabolite, GS-441524, were evaluated using a population PK (PopPK) approach to understand the PK aspect and exposure-clinical outcome relationship. The serum concentrations of remdesivir and GS-441524 (102 points in 39 patients) were measured using liquid chromatography-tandem mass spectrometry. All patients received 200 mg remdesivir on the first day, followed by 100 mg on 2-5 days, except for one patient who discontinued remdesivir on day 4. The median (range) age, body surface area, and estimated glomerular filtration rate (eGFR) were 70 (42-85), 1.74 m2 (1.36-2.03), and 68 mL/min/1.73 m2 (33-113), respectively. A compartment model with first-order elimination combined with remdesivir and GS-441524 was used for nonlinear mixed-effects model analysis. Remdesivir was rapidly eliminated after infusion, whereas GS-441524 was eliminated relatively slowly (half-time = 17.1 h). The estimated apparent clearance (CL) and distribution volume of GS-441524 were 11.0 L/h (intersubject variability [ISV]% = 43.0%) and 271 L (ISV% = 58.1%), respectively. The CL of GS-441524 was significantly related to the eGFR (CL × [eGFR/68]0.745 ). The post hoc area under the curve of GS-441524 was unrelated to the recovery rate or aspartate aminotransferase/alanine aminotransferase elevation. Overall, PopPK analysis showed the rapid elimination of remdesivir in the blood, and GS-441524 accumulation depended on eGFR in patients with COVID-19. However, no relevance of exposure-clinical outcome was not suggestive of the dose adjustment of remdesivir.
Collapse
Affiliation(s)
- Ryo Tamura
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Kei Irie
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan.,Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan
| | - Atsushi Nakagawa
- Department of Respiratory Medicine, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hirohito Muroi
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Masaaki Eto
- Department of Clinical Laboratory, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Hiroaki Ikesue
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Nobuyuki Muroi
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Shoji Fukushima
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan.,Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan
| | - Keisuke Tomii
- Department of Respiratory Medicine, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan
| | - Tohru Hashida
- Department of Pharmacy, Kobe City Hospital Organization, Kobe City Medical Center General Hospital, Kobe, Japan.,Faculty of Pharmaceutical Science, Kobe Gakuin University, Kobe, Japan
| |
Collapse
|
36
|
Elama HS, Zeid AM, Shalan SM, El-Shabrawy Y, Eid MI. Eco-friendly spectrophotometric methods for determination of remdesivir and favipiravir; the recently approved antivirals for COVID-19 treatment. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 287:122070. [PMID: 36403556 PMCID: PMC9650262 DOI: 10.1016/j.saa.2022.122070] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/25/2022] [Accepted: 10/30/2022] [Indexed: 05/17/2023]
Abstract
Remdesivir (REM) and Favipiravir (FAV) are recently approved antivirals prescribed in severely ill COVID-19 patients. Therefore, development of new, simple, rapid, sensitive, and selective methods for analysis of such drugs in their pharmaceutical formulations will be highly advantageous. Herein, we have developed different spectrophotometric methods for analysis of the studied analytes. Method I is based on direct spectrophotometric analysis of REM and FAV in ethanol at λmax 244 and 323 nm, respectively. For simultaneous quantitation of REM and FAV, methods II-V were followed. Method II is based on derivative spectrophotometry in which REM was determined in second-order derivative spectra at 248 nm (the zero-crossing wavelength for FAV), while FAV was measured in first-order derivative spectra at 337 nm (the zero-crossing point for REM). Method III is the dual-wavelength method in which spectral intensities were subtracted at 244-207 nm for REM and at 330-400 nm for FAV. Method IV is the ratio subtraction in which ratio spectra were obtained by a suitable divisor followed by subtraction of intensities at 272-340 nm and 335-222 nm for REM and FAV, respectively. Method V is the derivative ratio method in which the obtained ratio spectra in method IV were converted to first-order derivative and then REM and FAV were recorded at 280 and 340 nm, respectively. Calibration graphs were linear in the ranges of 1-10 µg/mL for REM through all methods and 1-20 µg/mL for FAV in methods I and II, and 2-20 µg/mL by the other methods. The evolved methods were applied to pharmaceutical dosage forms of REM and FAV. All the proposed methods were further applied to human plasma samples containing both drugs with acceptable mean recoveries.
Collapse
Affiliation(s)
- Heba Samir Elama
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt.
| | - Abdallah M Zeid
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Shereen Mahmoud Shalan
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Yasser El-Shabrawy
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Manal Ibrahim Eid
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
37
|
Kale A, Shelke V, Dagar N, Anders HJ, Gaikwad AB. How to use COVID-19 antiviral drugs in patients with chronic kidney disease. Front Pharmacol 2023; 14:1053814. [PMID: 36843922 PMCID: PMC9947246 DOI: 10.3389/fphar.2023.1053814] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Antiviral drugs such as Remdesivir (Veklury), Nirmatrelvir with Ritonavir (Paxlovid), Azvudine, and Molnupiravir (Lagevrio) can reduce the risk for severe and fatal Coronavirus Disease (COVID)-19. Although chronic kidney disease is a highly prevalent risk factor for severe and fatal COVID-19, most clinical trials with these drugs excluded patients with impaired kidney function. Advanced CKD is associated with a state of secondary immunodeficiency (SIDKD), which increases the susceptibility to severe COVID-19, COVID-19 complications, and the risk of hospitalization and mortality among COVID-19 patients. The risk to develop COVID-19 related acute kidney injury is higher in patients with precedent CKD. Selecting appropriate therapies for COVID-19 patients with impaired kidney function is a challenge for healthcare professionals. Here, we discuss the pharmacokinetics and pharmacodynamics of COVID-19-related antiviral drugs with a focus on their potential use and dosing in COVID-19 patients with different stages of CKD. Additionally, we describe the adverse effects and precautions to be taken into account when using these antivirals in COVID-19 patients with CKD. Lastly, we also discuss about the use of monoclonal antibodies in COVID-19 patients with kidney disease and related complications.
Collapse
Affiliation(s)
- Ajinath Kale
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Vishwadeep Shelke
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Neha Dagar
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Internal Medicine IV, Hospital of the Ludwig Maximilians University Munich, Munich, Germany
| | - Anil Bhanudas Gaikwad
- Laboratory of Molecular Pharmacology, Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Rajasthan, India
| |
Collapse
|
38
|
Saleh MA, Hirasawa M, Sun M, Gülave B, Elassaiss-Schaap J, de Lange EC. The PBPK LeiCNS-PK3.0 framework predicts Nirmatrelvir (but not Remdesivir or Molnupiravir) to achieve effective concentrations against SARS-CoV-2 in human brain cells. Eur J Pharm Sci 2023; 181:106345. [PMID: 36462547 PMCID: PMC9710098 DOI: 10.1016/j.ejps.2022.106345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Revised: 11/17/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022]
Abstract
SARS-CoV-2 was shown to infect and persist in the human brain cells for up to 230 days, highlighting the need to treat the brain viral load. The CNS disposition of the antiCOVID-19 drugs: Remdesivir, Molnupiravir, and Nirmatrelvir, remains, however, unexplored. Here, we assessed the human brain pharmacokinetic profile (PK) against the EC90 values of the antiCOVID-19 drugs to predict drugs with favorable brain PK against the delta and the omicron variants. We also evaluated the intracellular PK of GS443902 and EIDD2061, the active metabolites of Remdesivir and Molnupiravir, respectively. Towards this, we applied LeiCNS-PK3.0, the physiologically based pharmacokinetic framework with demonstrated adequate predictions of human CNS PK. Under the recommended dosing regimens, the predicted brain extracellular fluid PK of only Nirmatrelvir was above the variants' EC90. The intracellular levels of GS443902 and EIDD2061 were below the intracellular EC90. Summarizing, our model recommends Nirmatrelvir as the promising candidate for (pre)clinical studies investigating the CNS efficacy of antiCOVID-19 drugs.
Collapse
Affiliation(s)
- Mohammed A.A. Saleh
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Makoto Hirasawa
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Ming Sun
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | - Berfin Gülave
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands
| | | | - Elizabeth C.M. de Lange
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Center for Drug Research, Leiden University, Leiden, The Netherlands,Corresponding author
| |
Collapse
|
39
|
Liu XI, Dallmann A, Brooks K, Best BM, Clarke DF, Mirochnick M, van den Anker JN, Capparelli EV, Momper JD. Physiologically-based pharmacokinetic modeling of remdesivir and its metabolites in pregnant women with COVID-19. CPT Pharmacometrics Syst Pharmacol 2023; 12:148-153. [PMID: 36479969 PMCID: PMC9877749 DOI: 10.1002/psp4.12900] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/28/2022] [Accepted: 11/29/2022] [Indexed: 12/13/2022] Open
Abstract
Pregnant individuals are at high risk for severe illness from COVID-19, and there is an urgent need to identify safe and effective therapeutics for this population. Remdesivir (RDV) is a SARS-CoV-2 nucleotide analog RNA polymerase inhibitor. Limited RDV pharmacokinetic (PK) and safety data are available for pregnant women receiving RDV. The aims of this study were to translate a previously published nonpregnant adult physiologically based PK (PBPK) model for RDV to pregnancy and evaluate model performance with emerging clinical PK data in pregnant women with COVID-19. The pregnancy model was built in the Open Systems Pharmacology software suite (Version 10) including PK-Sim® and MoBi® with pregnancy-related changes of relevant enzymes applied. PK were predicted in a virtual population of 1000 pregnant subjects, and prediction results were compared with in vivo PK data from the International Maternal, Pediatric, Adolescent AIDS Clinical Trials (IMPAACT) Network 2032 study. The developed PBPK model successfully captured RDV and its metabolites' plasma concentrations during pregnancy. The ratios of prediction versus observation for RDV area under the curve from time 0 to infinity (AUC0-∞ ) and maximum concentration (Cmax ) were 1.61 and 1.17, respectively. For GS-704277, the ratios of predicted versus observed were 0.94 for AUC0-∞ and 1.20 for Cmax . For GS-441524, the ratios of predicted versus observed were 1.03 for AUC0-24 , 1.05 for Cmax , and 1.07 for concentrations at 24 h. All predictions of AUC and Cmax for RDV and its metabolites were within a twofold error range, and about 60% of predictions were within a 10% error range. These findings demonstrate the feasibility of translating PBPK models to pregnant women to potentially guide trial design, clinical decision making, and drug development.
Collapse
Affiliation(s)
- Xiaomei I. Liu
- Division of Clinical PharmacologyChildren's National HospitalWashingtonDCUSA
| | - André Dallmann
- Pharmacometrics/Modeling and Simulation, Research and Development, Pharmaceuticals, Bayer AGLeverkusenGermany
| | - Kristina Brooks
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Brookie M. Best
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
- Pediatrics Department, School of Medicine‐Rady Children's Hospital San DiegoUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Diana F. Clarke
- Section of Pediatrics Infectious Diseases, Boston Medical CenterBostonMassachusettsUSA
| | - Mark Mirochnick
- Department of PediatricsBoston University School of MedicineBostonMassachusettsUSA
| | | | - Edmund V. Capparelli
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
- Pediatrics Department, School of Medicine‐Rady Children's Hospital San DiegoUniversity of California, San DiegoLa JollaCaliforniaUSA
| | - Jeremiah D. Momper
- Skaggs School of Pharmacy and Pharmaceutical SciencesUniversity of California, San DiegoLa JollaCaliforniaUSA
| |
Collapse
|
40
|
Abouellil A, Bilal M, Taubert M, Fuhr U. A population pharmacokinetic model of remdesivir and its major metabolites based on published mean values from healthy subjects. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2023; 396:73-82. [PMID: 36123499 PMCID: PMC9485022 DOI: 10.1007/s00210-022-02292-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 09/08/2022] [Indexed: 01/29/2023]
Abstract
Remdesivir is a direct-acting anti-viral agent. It was originally evaluated against filoviruses. However, during the COVID-19 pandemic, it was investigated due to its anti-viral activities against (SARS-CoV-2) virus. Therefore remdesivir received conditional approval for treatment of patients with severe coronavirus disease. Yet, its pharmacokinetic properties are inadequately understood. This report describes the population pharmacokinetics of remdesivir and its two plasma-detectable metabolites (GS-704277 and GS-441524) in healthy volunteers. The data was extracted from published phase I single escalating and multiple i.v remdesivir dose studies conducted by the manufacturer. The model was developed by standard methods using non-linear mixed effect modeling. Also, a series of simulations were carried out to test suggested clinical doses. The model describes the distribution of remdesivir and each of its metabolites by respective two compartments with sequential metabolism between moieties, and elimination from central compartments. As individual data were not available, only inter-cohort variability could be assessed. The estimated point estimates for central (and peripheral) volumes of distribution for remdesivir, GS-704277, and GS-441524 were 4.89 L (46.5 L), 96.4 L (8.64 L), and 26.2 L (66.2 L), respectively. The estimated elimination clearances of remdesivir, GS704277, and GS-441524 reached 18.1 L/h, 36.9 L/h, and 4.74 L/h, respectively. The developed model described the data well. Simulations of clinically approved doses showed that GS-441524 concentrations in plasma exceeded the reported EC50 values during the complete duration of treatment. Nonetheless, further studies are needed to explore the pharmacokinetics of remdesivir and its relationship to clinical efficacy, and the present model may serve as a useful starting point for additional evaluations.
Collapse
Affiliation(s)
- Ahmed Abouellil
- grid.411097.a0000 0000 8852 305XFaculty of Medicine, Center for Pharmacology, Department I of Pharmacology, University Hospital Cologne, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany ,grid.15090.3d0000 0000 8786 803XImmunosensation Cluster of Excellence, University Hospital Bonn, Bonn, Germany
| | - Muhammad Bilal
- grid.411097.a0000 0000 8852 305XFaculty of Medicine, Center for Pharmacology, Department I of Pharmacology, University Hospital Cologne, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany ,grid.10388.320000 0001 2240 3300Department of Clinical Pharmacy, Institute of Pharmacy, University of Bonn, Bonn, Germany
| | - Max Taubert
- grid.411097.a0000 0000 8852 305XFaculty of Medicine, Center for Pharmacology, Department I of Pharmacology, University Hospital Cologne, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany
| | - Uwe Fuhr
- grid.411097.a0000 0000 8852 305XFaculty of Medicine, Center for Pharmacology, Department I of Pharmacology, University Hospital Cologne, University of Cologne, Gleueler Straße 24, 50931 Cologne, Germany
| |
Collapse
|
41
|
Wang Z, Yang L, Song XQ. Oral GS-441524 derivatives: Next-generation inhibitors of SARS-CoV-2 RNA-dependent RNA polymerase. Front Immunol 2022; 13:1015355. [PMID: 36561747 PMCID: PMC9763260 DOI: 10.3389/fimmu.2022.1015355] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
GS-441524, an RNA-dependent RNA polymerase (RdRp) inhibitor, is a 1'-CN-substituted adenine C-nucleoside analog with broad-spectrum antiviral activity. However, the low oral bioavailability of GS-441524 poses a challenge to its anti-SARS-CoV-2 efficacy. Remdesivir, the intravenously administered version (version 1.0) of GS-441524, is the first FDA-approved agent for SARS-CoV-2 treatment. However, clinical trials have presented conflicting evidence on the value of remdesivir in COVID-19. Therefore, oral GS-441524 derivatives (VV116, ATV006, and GS-621763; version 2.0, targeting highly conserved viral RdRp) could be considered as game-changers in treating COVID-19 because oral administration has the potential to maximize clinical benefits, including decreased duration of COVID-19 and reduced post-acute sequelae of SARS-CoV-2 infection, as well as limited side effects such as hepatic accumulation. This review summarizes the current research related to the oral derivatives of GS-441524, and provides important insights into the potential factors underlying the controversial observations regarding the clinical efficacy of remdesivir; overall, it offers an effective launching pad for developing an oral version of GS-441524.
Collapse
Affiliation(s)
- Zhonglei Wang
- Key Laboratory of Green Natural Products and Pharmaceutical Intermediates in Colleges and Universities of Shandong Province, School of Chemistry and Chemical Engineering, Qufu Normal University, Qufu, China,School of Pharmaceutical Sciences, Tsinghua University, Beijing, China,*Correspondence: Zhonglei Wang, ; Liyan Yang, ; Xian-qing Song,
| | - Liyan Yang
- Shandong Provincial Key Laboratory of Laser Polarization and Information Technology, School of Physics and Physical Engineering, Qufu Normal University, Qufu, China,*Correspondence: Zhonglei Wang, ; Liyan Yang, ; Xian-qing Song,
| | - Xian-qing Song
- General Surgery Department, Ningbo Fourth Hospital, Xiangshan, China,*Correspondence: Zhonglei Wang, ; Liyan Yang, ; Xian-qing Song,
| |
Collapse
|
42
|
Real-life experience with remdesivir for treatment of hospitalized coronavirus disease 2019 patients: matched case-control study from a large tertiary hospital registry. Croat Med J 2022; 63:536-543. [PMID: 36597565 PMCID: PMC9837722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
AIM To evaluate the association of remdesivir use and the survival of hospitalized patients with coronavirus disease 2019 (COVID-19). METHODS We retrospectively reviewed the medical records of 5959 COVID-19 patients admitted to our tertiary-level hospital from March 2020 to June 2021. A total of 876 remdesivir-treated patients were matched with 876 control patients in terms of age, sex, Charlson comorbidity index (CCI), WHO-defined COVID-19 severity on admission, and oxygen requirement at the time of remdesivir use. RESULTS Among 1752 COVID-19 patients (median age 66 years, 61.8% men), 1405 (80.2%) had severe and 311 (17.8%) had critically severe COVID-19 on admission. Remdesivir was given at a median of one day after hospital admission and at a median of eight days from the onset of symptoms. Overall, 645 (73.6%) patients received remdesivir before high-flow oxygen therapy (HFOT) or mechanical ventilation (MV), 198 (22.6%) after HFOT institution, and 83 (9.5%) after MV institution. Remdesivir use was associated with improved survival in the entire cohort (hazard ratio 0.79, P=0.006). Survival benefit was evident among patients receiving remdesivir during low-flow oxygen requirement (hazard ratio 0.61, P<0.001) but not among patients who received it after starting HFOT (P=0.499) or MV (P=0.380). CONCLUSION Remdesivir, if given during low-flow oxygen therapy, might be associated with survival benefit in hospitalized COVID-19 patients.
Collapse
|
43
|
Lucijanić M, Bušić N, Bistrović P, Papić I, Zelenika Margeta M, Babić P, Barčan M, Pasarić A, Mustapić M, Piskač Živković N, Ortner Hadžiabdić M, Lucijanić T, Lukšić I, Baršić B. Real-life experience with remdesivir for treatment of hospitalized coronavirus disease 2019 patients: matched case-control study from a large tertiary hospital registry. Croat Med J 2022; 63. [PMID: 36597565 PMCID: PMC9837722 DOI: 10.3325/cmj.2022.63.536] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
AIM To evaluate the association of remdesivir use and the survival of hospitalized patients with coronavirus disease 2019 (COVID-19). METHODS We retrospectively reviewed the medical records of 5959 COVID-19 patients admitted to our tertiary-level hospital from March 2020 to June 2021. A total of 876 remdesivir-treated patients were matched with 876 control patients in terms of age, sex, Charlson comorbidity index (CCI), WHO-defined COVID-19 severity on admission, and oxygen requirement at the time of remdesivir use. RESULTS Among 1752 COVID-19 patients (median age 66 years, 61.8% men), 1405 (80.2%) had severe and 311 (17.8%) had critically severe COVID-19 on admission. Remdesivir was given at a median of one day after hospital admission and at a median of eight days from the onset of symptoms. Overall, 645 (73.6%) patients received remdesivir before high-flow oxygen therapy (HFOT) or mechanical ventilation (MV), 198 (22.6%) after HFOT institution, and 83 (9.5%) after MV institution. Remdesivir use was associated with improved survival in the entire cohort (hazard ratio 0.79, P=0.006). Survival benefit was evident among patients receiving remdesivir during low-flow oxygen requirement (hazard ratio 0.61, P<0.001) but not among patients who received it after starting HFOT (P=0.499) or MV (P=0.380). CONCLUSION Remdesivir, if given during low-flow oxygen therapy, might be associated with survival benefit in hospitalized COVID-19 patients.
Collapse
Affiliation(s)
- Marko Lucijanić
- Hematology Department, Dubrava University Hospital, Zagreb, Croatia,Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,University of Zagreb, School of Medicine, Zagreb, Croatia
| | - Nikolina Bušić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia
| | - Petra Bistrović
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Cardiology Department, Dubrava University Hospital, Zagreb, Croatia
| | - Ivan Papić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Pharmacy Department, Dubrava University Hospital, Zagreb, Croatia
| | - Marina Zelenika Margeta
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Pulmonology Department, Dubrava University Hospital, Zagreb, Croatia
| | - Paško Babić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Department of Emergency and Intensive Care Medicine, Dubrava University Hospital, Zagreb, Croatia
| | - Mihaela Barčan
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Department of Emergency and Intensive Care Medicine, Dubrava University Hospital, Zagreb, Croatia
| | - Antica Pasarić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Department of Clinical Immunology, Allergology, and Rheumatology, Dubrava University Hospital, Zagreb, Croatia
| | - Mirna Mustapić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Endocrinology Department, Dubrava University Hospital, Zagreb, Croatia
| | - Nevenka Piskač Živković
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Pulmonology Department, Dubrava University Hospital, Zagreb, Croatia
| | - Maja Ortner Hadžiabdić
- Centre for Applied Pharmacy, Faculty of Pharmacy and Biochemistry, University of Zagreb, Zagreb, Croatia
| | - Tomo Lucijanić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,Endocrinology Department, Dubrava University Hospital, Zagreb, Croatia
| | - Ivica Lukšić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,University of Zagreb, School of Medicine, Zagreb, Croatia,Department of Maxillofacial Surgery, Dubrava University Hospital, Zagreb, Croatia
| | - Bruno Baršić
- Primary Respiratory and Intensive Care Center, Dubrava University Hospital, Zagreb, Croatia,University of Zagreb, School of Medicine, Zagreb, Croatia
| |
Collapse
|
44
|
Vulturar DM, Neag MA, Vesa ȘC, Maierean AD, Gherman D, Buzoianu AD, Orăsan OH, Todea DA. Therapeutic Efficacy and Outcomes of Remdesivir versus Remdesivir with Tocilizumab in Severe SARS-CoV-2 Infection. Int J Mol Sci 2022; 23:ijms232214462. [PMID: 36430945 PMCID: PMC9698366 DOI: 10.3390/ijms232214462] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 11/17/2022] [Accepted: 11/18/2022] [Indexed: 11/23/2022] Open
Abstract
The infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) generated many challenges to find an effective drug combination for hospitalized patients with severe forms of coronavirus disease 2019 (COVID-19) pneumonia. We conducted a retrospective cohort study, including 182 patients with severe COVID-19 pneumonia hospitalized between March and October 2021 in a Pneumology Hospital from Cluj-Napoca, Romania. Among patients treated with standard of care, 100 patients received remdesivir (R group) and 82 patients received the combination of remdesivir plus tocilizumab (RT group). We compared the clinical outcomes, the inflammatory markers, superinfections, oxygen requirement, intensive care unit (ICU) admission and mortality rate before drug administration and 7 days after in R group and RT group. Borg score and oxygen support showed an improvement in the R group (p < 0.005). Neutrophiles, C-reactive protein (CRP) and serum ferritin levels decreased significantly in RT group but with a higher rate of superinfection in this group. ICU admission and death did not differ significantly between groups. The combination of remdesivir plus tocilizumab led to a significantly improvement in the inflammatory markers and a decrease in the oxygen requirement. Although the superinfection rate was higher in RT group than in R group, no significant difference was found in the ICU admission and mortality rate between the groups.
Collapse
Affiliation(s)
- Damiana-Maria Vulturar
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Maria Adriana Neag
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
- Correspondence:
| | - Ștefan Cristian Vesa
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Anca-Diana Maierean
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| | - Diana Gherman
- Department of Radiology, Iuliu Hatieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Anca Dana Buzoianu
- Pharmacology, Toxicology and Clinical Pharmacology Department, Iuliu Hațieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania
| | - Olga Hilda Orăsan
- 5th Department Internal Medicine, 4th Medical Clinic, Iuliu Hațieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
| | - Doina-Adina Todea
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400332 Cluj-Napoca, Romania
| |
Collapse
|
45
|
De Forni D, Poddesu B, Cugia G, Chafouleas J, Lisziewicz J, Lori F. Synergistic drug combinations designed to fully suppress SARS-CoV-2 in the lung of COVID-19 patients. PLoS One 2022; 17:e0276751. [PMID: 36355808 PMCID: PMC9648746 DOI: 10.1371/journal.pone.0276751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 10/12/2022] [Indexed: 11/12/2022] Open
Abstract
Despite new antivirals are being approved against SARS-CoV-2 they suffer from significant constraints and are not indicated for hospitalized patients, who are left with few antiviral options. Repurposed drugs have previously shown controversial clinical results and it remains difficult to understand why certain trials delivered positive results and other trials failed. Our manuscript contributes to explaining the puzzle: this might have been caused by a suboptimal drug exposure and, consequently, an incomplete virus suppression, also because the drugs have mostly been used as add-on monotherapies. As with other viruses (e.g., HIV and HCV) identifying synergistic combinations among such drugs could overcome monotherapy-related limitations. In a cell culture model for SARS-CoV-2 infection the following stringent criteria were adopted to assess drug combinations: 1) identify robust, synergistic antiviral activity with no increase in cytotoxicity, 2) identify the lowest drug concentration inhibiting the virus by 100% (LIC100) and 3) understand whether the LIC100 could be reached in the lung at clinically indicated drug doses. Among several combinations tested, remdesivir with either azithromycin or ivermectin synergistically increased the antiviral activity with no increase in cytotoxicity, improving the therapeutic index and lowering the LIC100 of every one of the drugs to levels that are expected to be achievable and maintained in the lung for a therapeutically relevant period of time. These results are consistent with recent clinical observations showing that intensive care unit admission was significantly delayed by the combination of AZI and RDV, but not by RDV alone, and could have immediate implications for the treatment of hospitalized patients with COVID-19 as the proposed "drug cocktails" should have antiviral activity against present and future SARS-CoV-2 variants without significant overlapping toxicity, while minimizing the onset of drug resistance. Our results also provide a validated methodology to help sort out which combination of drugs are most likely to be efficacious in vivo, based on their in vitro activity, potential synergy and PK profiles.
Collapse
Affiliation(s)
| | | | | | | | - Julianna Lisziewicz
- Research Institute for Genetic and Human Therapy, Colorado Springs, CO, United States of America
| | - Franco Lori
- ViroStatics s.r.l., Tramariglio, Italy
- Research Institute for Genetic and Human Therapy, Colorado Springs, CO, United States of America
| |
Collapse
|
46
|
Dhanalakshmi M, Das K, Pandya M, Shah S, Gadnayak A, Dave S, Das J. Artificial Neural Network-Based Study Predicts GS-441524 as a Potential Inhibitor of SARS-CoV-2 Activator Protein Furin: a Polypharmacology Approach. Appl Biochem Biotechnol 2022; 194:4511-4529. [PMID: 35507249 PMCID: PMC9066385 DOI: 10.1007/s12010-022-03928-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Furin, a pro-protein convertase, plays a significant role as a biological scissor in bacterial, viral, and even mammalian substrates which in turn decides the fate of many viral and bacterial infections along with the numerous ailments caused by cancer, diabetes, inflammations, and neurological disorders. In the wake of the current pandemic caused by the virus SARS-CoV-2, furin has become the center of attraction for researchers as the spike protein contains a polybasic furin cleavage site. In the present work, we have searched for novel inhibitors against this interesting human target from FDA-approved antiviral. To enhance the selection of new inhibitors, we employed Kohonen's artificial neural network-based self-organizing maps for ligand-based virtual screening. Promising results were obtained which can help in drug repurposing and network pharmacology studies can address the errors generated due to promiscuity/polypharmacology. We found 15 existing FDA antiviral drugs having the potential to inhibit furin. Among these, six compounds have targets on important human proteins (LDLR, FCGR1A, PCK1, TLR7, DNA, and PNP). The role of these 15 drugs inhibiting furin can be established by studying further on patients infected with number of viruses including SARS-CoV-2. Here we propose two promising candidate FDA drugs GS-441524 and Grazoprevir (MK-5172) for repurposing as inhibitors of furin. The best results were observed with GS-441524.
Collapse
Affiliation(s)
- M Dhanalakshmi
- Research and Development Centre, Bharathiar University, Marudhamalai Rd, Coimbatore, Tamil Nadu, India
| | - Kajari Das
- Department of Biotechnology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, Odisha, India
| | - Medha Pandya
- The KPES Science College, Maharaja krishnakumarsinhji Bhavnagar University, Bhavnagar, Gujarat, India
| | - Sejal Shah
- Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - Ayushman Gadnayak
- Centre for Genomics & Biomedical Informatics, IMS and SUM Hospital, Siksha "O" Anusandhan (Deemed to be University), Bhubaneswar, Odisha, India
| | - Sushma Dave
- Department of Applied Sciences, JIET, Jodhpur, Rajasthan, India.
| | | |
Collapse
|
47
|
Zhang Q, Melchert PW, Markowitz JS. In vitro evaluation of the impact of Covid-19 therapeutic agents on the hydrolysis of the antiviral prodrug remdesivir. Chem Biol Interact 2022; 365:110097. [PMID: 35964681 PMCID: PMC9367181 DOI: 10.1016/j.cbi.2022.110097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/01/2022] [Accepted: 08/06/2022] [Indexed: 11/27/2022]
Abstract
Remdesivir (RDV, Veklury®) is an FDA-approved prodrug for the treatment of hospitalized patients with COVID-19. Recent in vitro studies have indicated that human carboxylesterase 1 (CES1) is the major metabolic enzyme catalyzing RDV activation. COVID-19 treatment for hospitalized patients typically also involves a number of antibiotics and anti-inflammatory drugs. Further, individuals who are carriers of a CES1 variant (polymorphism in exon 4 codon 143 [G143E]) may experience impairment in their ability to metabolize therapeutic agents which are CES1 substrates. The present study assessed the potential influence of nine therapeutic agents (hydroxychloroquine, ivermectin, erythromycin, clarithromycin, roxithromycin, trimethoprim, ciprofloxacin, vancomycin, and dexamethasone) commonly used in treating COVID-19 and 5 known CES1 inhibitors on the metabolism of RDV. Additionally, we further analyzed the mechanism of inhibition of cannabidiol (CBD), as well as the impact of the G143E polymorphism on RDV metabolism. An in vitro S9 fraction incubation method and in vitro to in vivo pharmacokinetic scaling were utilized. None of the nine therapeutic agents evaluated produced significant inhibition of RDV hydrolysis; CBD was found to inhibit RDV hydrolysis by a mixed type of competitive and noncompetitive partial inhibition mechanism. In vitro to in vivo modeling suggested a possible reduction of RDV clearance and increase of AUC when coadministration with CBD. The same scaling method also suggested a potentially lower clearance and higher AUC in the presence of the G143E variant. In conclusion, a potential CES1-mediated DDI between RDV and the nine assessed medications appears unlikely. However, a potential CES1-mediated DDI between RDV and CBD may be possible with sufficient exposure to the cannabinoid. Patients carrying the CES1 G143E variant may exhibit a slower biotransformation and clearance of RDV. Further clinical studies would be required to evaluate and characterize the clinical significance of a CBD-RDV interaction.
Collapse
Affiliation(s)
- Qingchen Zhang
- Department of Pharmacotherapy and Translational Research, Gainesville, FL, USA
| | - Philip W Melchert
- Department of Pharmacotherapy and Translational Research, Gainesville, FL, USA
| | - John S Markowitz
- Department of Pharmacotherapy and Translational Research, Gainesville, FL, USA; Center for Pharmacogenomics and Precision Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
48
|
Clinically Relevant Interactions Between Ritonavir-Boosted Nirmatrelvir and Concomitant Antiseizure Medications: Implications for the Management of COVID-19 in Patients with Epilepsy. Clin Pharmacokinet 2022; 61:1219-1236. [PMID: 35895276 PMCID: PMC9325946 DOI: 10.1007/s40262-022-01152-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 01/06/2023]
Abstract
Ritonavir-boosted nirmatrelvir (RBN) has been authorized recently in several countries as an orally active anti-SARS-CoV-2 treatment for patients at high risk of progressing to severe COVID-19 disease. Nirmatrelvir is the active component against the SARS-CoV-2 virus, whereas ritonavir, a potent CYP3A inhibitor, is intended to boost the activity of nirmatrelvir by increasing its concentration in plasma to ensure persistence of antiviral concentrations during the 12-hour dosing interval. RBN is involved in many clinically important drug–drug interactions both as perpetrator and as victim, which can complicate its use in patients treated with antiseizure medications (ASMs). Interactions between RBN and ASMs are bidirectional. As perpetrator, RBN may increase the plasma concentration of a number of ASMs that are CYP3A4 substrates, possibly leading to toxicity. As victims, both nirmatrelvir and ritonavir are subject to metabolic induction by concomitant treatment with potent enzyme-inducing ASMs (carbamazepine, phenytoin, phenobarbital and primidone). According to US and European prescribing information, treatment with these ASMs is a contraindication to the use of RBN. Although remdesivir is a valuable alternative to RBN, it may not be readily accessible in some settings due to cost and/or need for intravenous administration. If remdesivir is not an appropriate option, either bebtelovimab or molnupiravir may be considered. However, evidence about the clinical efficacy of bebtelovimab is still limited, and molnupiravir, the only orally active alternative, is deemed to have appreciably lower efficacy than RBN and remdesivir.
Collapse
|
49
|
Yang M, Xu X. Important roles of transporters in the pharmacokinetics of anti-viral nucleoside/nucleotide analogs. Expert Opin Drug Metab Toxicol 2022; 18:483-505. [PMID: 35975669 PMCID: PMC9506706 DOI: 10.1080/17425255.2022.2112175] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 08/02/2022] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Nucleoside analogs are an important class of antiviral agents. Due to the high hydrophilicity and limited membrane permeability of antiviral nucleoside/nucleotide analogs (AVNAs), transporters play critical roles in AVNA pharmacokinetics. Understanding the properties of these transporters is important to accelerate translational research for AVNAs. AREAS COVERED The roles of key transporters in the pharmacokinetics of 25 approved AVNAs were reviewed. Clinically relevant information that can be explained by the modulation of transporter functions is also highlighted. EXPERT OPINION Although the roles of transporters in the intestinal absorption and renal excretion of AVNAs have been well identified, more research is warranted to understand their roles in the distribution of AVNAs, especially to immune privileged compartments where treatment of viral infection is challenging. P-gp, MRP4, BCRP, and nucleoside transporters have shown extensive impacts in the disposition of AVNAs. It is highly recommended that the role of transporters should be investigated during the development of novel AVNAs. Clinically, co-administered inhibitors and genetic polymorphism of transporters are the two most frequently reported factors altering AVNA pharmacokinetics. Physiopathology conditions also regulate transporter activities, while their effects on pharmacokinetics need further exploration. Pharmacokinetic models could be useful for elucidating these complicated factors in clinical settings.
Collapse
Affiliation(s)
- Mengbi Yang
- Drug Metabolism and Pharmacokinetics, Division of Preclinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| | - Xin Xu
- Drug Metabolism and Pharmacokinetics, Division of Preclinical Innovation (DPI), National Center for Advancing Translational Sciences (NCATS), National Institutes of Health, 9800 Medical Center Drive, Rockville, MD 20850, USA
| |
Collapse
|
50
|
Islam T, Hasan M, Rahman MS, Islam MR. Comparative evaluation of authorized drugs for treating Covid-19 patients. Health Sci Rep 2022; 5:e671. [PMID: 35734340 PMCID: PMC9194463 DOI: 10.1002/hsr2.671] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/22/2022] [Accepted: 05/08/2022] [Indexed: 12/15/2022] Open
Abstract
Background and Aims Vaccines are the first line of defense against coronavirus disease 2019 (Covid-19). However, the antiviral drugs provide a new tool to fight the Covid-19 pandemic. Here we aimed for a comparative evaluation of authorized drugs for treating Covid-19 patients. Methods We searched in PubMed and Google Scholar using keywords and terms such as Covid, SARS-CoV-2, Coronavirus disease 2019, therapeutic management, hospitalized Covid-19 patients, Covid-19 treatment. We also gathered information from reputed newspapers, web portals, and websites. We thoroughly observed, screened, and included the studies relevant to our inclusion criteria. We included only the United States Food and Drug Administration (FDA) authorized drugs for this review. Results We found that molnupiravir and paxlovid are available for oral use, and remdesivir is for only hospitalized patients. Paxlovid is a combination of nirmatrelvir and ritonavir, nirmatrelvir is a protease inhibitor (ritonavir increases the concentration of nirmatrelvir), and the other two (remdesivir and molnupiravir) are nucleoside analog prodrugs. Remdesivir and molnupiravir doses do not need to adjust in renal and hepatic impairment. However, the paxlovid dose adjustment is required for mild to moderate renal or hepatic impaired patients. Also, the drug is not allowed for Covid-19 patients with severe renal or hepatic impairment. Preliminary studies showed oral antiviral drugs significantly reduce hospitalization or death among mild to severe patients. Moreover, the US FDA has approved four monoclonal antibodies for Covid-19 treatment. Studies suggest that these drugs would reduce the risk of hospitalization or severity of symptoms. World Health Organization strongly recommended the use of corticosteroids along with other antiviral drugs for severe or critically hospitalized patients. Conclusion All authorized drugs are effective in inhibiting viral replication for most SARS-CoV-2 variants. Therefore, along with vaccines, these drugs might potentially aid in fighting the Covid-19 pandemic.
Collapse
Affiliation(s)
- Towhidul Islam
- Department of PharmacyUniversity of Asia PacificDhakaBangladesh
| | - Moynul Hasan
- Department of PharmacyJagannath UniversityDhakaBangladesh
| | | | | |
Collapse
|