A randomized clinical trial of lipid metabolism modulation with fenofibrate for acute coronavirus disease 2019

Julio A Chirinos, Patricio Lopez-Jaramillo, Evangelos J Giamarellos-Bourboulis, Gonzalo H Dávila-Del-Carpio, Abdul Rahman Bizri, Jaime F Andrade-Villanueva, Oday Salman, Carlos Cure-Cure, Nelson R Rosado-Santander, Mario P Cornejo Giraldo, Luz A González-Hernández, Rima Moghnieh, Rapti Angeliki, María E Cruz Saldarriaga, Marcos Pariona, Carola Medina, Ioannis Dimitroulis, Charalambos Vlachopoulos, Corina Gutierrez, Juan E Rodriguez-Mori, Edgar Gomez-Laiton, Rosa Cotrina Pereyra, Jorge Luis Ravelo Hernández, Hugo Arbañil, José Accini-Mendoza, Maritza Pérez-Mayorga, Charalampos Milionis, Garyfallia Poulakou, Gregorio Sánchez, Renzo Valdivia-Vega, Mirko Villavicencio-Carranza, Ricardo J Ayala-García, Carlos A Castro-Callirgos, Rosa M Alfaro Carrasco, Willy Garrido Lecca Danos, Tiffany Sharkoski, Katherine Greene, Bianca Pourmussa, Candy Greczylo, Juan Ortega-Legaspi, Douglas Jacoby, Jesse Chittams, Paraskevi Katsaounou, Zoi Alexiou, Styliani Sympardi, Nancy K Sweitzer, Mary Putt, Jordana B Cohen, FERMIN Investigators, Ciro Barrantes Alarcón, Denisse Marylyn Mendoza Sanchez, Eduardo Francisco Bernales Salas, Claudia Jesús Chamby Díaz, Ursula Milagros Vargas Gómez, Cynthia Daniela Salinas Herrera, Naldy Lidia Barriga Triviños, Johanna Carolina Coacalla Guerra, Evelyn Marrón Veria, Preethi William, Hugo Espinoza-Rojas, Irwing Renato Benites-Flores, Pedro Antonio Segura-Saldaña, Julio A Chirinos, Patricio Lopez-Jaramillo, Evangelos J Giamarellos-Bourboulis, Gonzalo H Dávila-Del-Carpio, Abdul Rahman Bizri, Jaime F Andrade-Villanueva, Oday Salman, Carlos Cure-Cure, Nelson R Rosado-Santander, Mario P Cornejo Giraldo, Luz A González-Hernández, Rima Moghnieh, Rapti Angeliki, María E Cruz Saldarriaga, Marcos Pariona, Carola Medina, Ioannis Dimitroulis, Charalambos Vlachopoulos, Corina Gutierrez, Juan E Rodriguez-Mori, Edgar Gomez-Laiton, Rosa Cotrina Pereyra, Jorge Luis Ravelo Hernández, Hugo Arbañil, José Accini-Mendoza, Maritza Pérez-Mayorga, Charalampos Milionis, Garyfallia Poulakou, Gregorio Sánchez, Renzo Valdivia-Vega, Mirko Villavicencio-Carranza, Ricardo J Ayala-García, Carlos A Castro-Callirgos, Rosa M Alfaro Carrasco, Willy Garrido Lecca Danos, Tiffany Sharkoski, Katherine Greene, Bianca Pourmussa, Candy Greczylo, Juan Ortega-Legaspi, Douglas Jacoby, Jesse Chittams, Paraskevi Katsaounou, Zoi Alexiou, Styliani Sympardi, Nancy K Sweitzer, Mary Putt, Jordana B Cohen, FERMIN Investigators, Ciro Barrantes Alarcón, Denisse Marylyn Mendoza Sanchez, Eduardo Francisco Bernales Salas, Claudia Jesús Chamby Díaz, Ursula Milagros Vargas Gómez, Cynthia Daniela Salinas Herrera, Naldy Lidia Barriga Triviños, Johanna Carolina Coacalla Guerra, Evelyn Marrón Veria, Preethi William, Hugo Espinoza-Rojas, Irwing Renato Benites-Flores, Pedro Antonio Segura-Saldaña

Abstract

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) cytotoxicity may involve inhibition of peroxisome proliferator-activated receptor alpha. Fenofibrate activates peroxisome proliferator-activated receptor alpha and inhibits SARS-CoV-2 replication in vitro. Whether fenofibrate can be used to treat coronavirus disease 2019 (COVID-19) infection in humans remains unknown. Here, we randomly assigned inpatients and outpatients with COVID-19 within 14 d of symptom onset to 145 mg of oral fenofibrate nanocrystal formulation versus placebo for 10 d, in a double-blinded fashion. The primary endpoint was a severity score whereby participants were ranked across hierarchical tiers incorporating time to death, mechanical ventilation duration, oxygenation, hospitalization and symptom severity and duration. In total, 701 participants were randomized to fenofibrate (n = 351) or placebo (n = 350). The mean age of participants was 49 ± 16 years, 330 (47%) were female, mean body mass index was 28 ± 6 kg/m2 and 102 (15%) had diabetes. Death occurred in 41 participants. Compared with placebo, fenofibrate had no effect on the primary endpoint. The median (interquartile range) rank in the placebo arm was 347 (172, 453) versus 345 (175, 453) in the fenofibrate arm (P = 0.819). There was no difference in secondary and exploratory endpoints, including all-cause death, across arms. There were 61 (17%) adverse events in the placebo arm compared with 46 (13%) in the fenofibrate arm, with slightly higher incidence of gastrointestinal side effects in the fenofibrate group. Overall, among patients with COVID-19, fenofibrate has no significant effect on various clinically relevant outcomes ( NCT04517396 ).

Conflict of interest statement

In the last 2 years, J.B.C. has received research grants from the National Institutes of Health and American Heart Association. In the last 2 years, J.A.C. has received consulting honoraria from Sanifit, Bristol Myers Squibb, Merck, Edwards Lifesciences, Bayer, JNJ, the University of Delaware, and research grants from the National Institutes of Health, Abbott, Microsoft, Fukuda-Denshi and Bristol Myers Squibb. J.A.C. has received compensation from the American Heart Association and the American College of Cardiology for editorial roles, and visiting speaker honoraria from Washington University, University of Utah, the Japanese Association for Cardiovascular Nursing and the Korean Society of Cardiology. E.J.G. has received honoraria from Abbott CH, bioMérieux, Brahms, GSK, InflaRx, Sobi and XBiotech; independent educational grants from Abbott CH, AxisShield, bioMérieux, InflaRx, Johnson & Johnson, MSD, Sobi and XBiotech; and funding from the Horizon 2020 Marie-Curie Project European Sepsis Academy (granted to the National and Kapodistrian University of Athens), and the Horizon 2020 European Grants ImmunoSep and RISKinCOVID (granted to the Hellenic Institute for the Study of Sepsis). In the last 2 years, N.K.S. has received compensation from the American Heart Association for editorial duties. The remaining authors declare no competing interests.

© 2022. The Author(s), under exclusive licence to Springer Nature Limited.

Figures

Fig. 1
Fig. 1
Participant enrolment, randomization and follow-up in the FEnofibRate as a Metabolic INtervention for COVID-19 (FERMIN) trial.
Fig. 2. Key outcomes among participants in…
Fig. 2. Key outcomes among participants in each randomization arm.
a, Distribution of the primary endpoint (ranked severity score) between the randomization arms (placebo N = 347 participants; fenofibrate N = 347 participants). The y axis represents the range of ranked severity scores, and the x axis represents the frequency density of distributions of the ranks in each treatment arm. The white dot represents the median ranked severity score, the solid box represents the IQR, and the vertical lines represent the upper-adjacent and lower-adjacent values. The upper-adjacent value and the IQR values were identical. b, Cumulative incidence for all-cause death at 30 d.
Fig. 3. Forest plot of the differences…
Fig. 3. Forest plot of the differences in ranked severity scores across subgroups.
The plot represents the differences in median ranked severity scores between participants randomized to fenofibrate versus placebo in each subgroup. Positive values indicate better outcomes in the fenofibrate arm. Negative values indicate better outcomes in the placebo group. The central dot represents the difference in median scores and the error bars represent the 95% CIs.
Fig. 4. Primary endpoint of the trial…
Fig. 4. Primary endpoint of the trial (ranked severity score).
Participants were ranked hierarchically according to their clinical course. The primary endpoint of the trial was a global rank score that ranked patient outcomes according to five factors, shown as tiers from top to bottom (labelled as subsets 1–5). The left-sided icons (walking person and person in hospital bed) indicate the participant status at the time of enrolment (outpatient and inpatient, respectively). The top tier included inpatients and outpatients, whereas other subsets included inpatients or outpatients, but not both. Outcomes of hypothetical participants are represented within each tier by the right-pointing arrows. The icons on the right of the arrows represent the participant status at trial completion or at the time of the relevant outcome event (such as death, hospital discharge or hospital admission). Participants were ranked within each tier according to the following specific criteria: tier 1, time to death (ranked from shortest to longest, up to 30 d after randomization); tier 2, for participants enrolled as inpatients, the number of days supported by mechanical ventilation (invasive or noninvasive) or ECMO (until hospital discharge, up to 30 d after randomization, ranked from longest to shortest); tier 3, for participants enrolled as inpatients who did not require mechanical ventilation or ECMO, the FiO2/SpO2 ratio area under the curve until hospital discharge, up to 30 d after randomization, ranked from highest to lowest; tier 4, for participants enrolled as outpatients who were subsequently hospitalized, the number of days out of the hospital during the 30-d period following randomization (ranked from lowest to highest); tier 5, for participants enrolled as outpatients who did not get hospitalized during the 30 d observation period, the modified dyspnoea Borg scale (mean value of assessments at ~5, ~10 and ~15 d, ranked from highest to lowest). AUC, area under the curve.
Extended Data Fig. 1
Extended Data Fig. 1
Kaplan–Meier curve of time to hospitalization (among participants enrolled as outpatients).
Extended Data Fig. 2
Extended Data Fig. 2
Cumulative incidence curve of time to discharge (among participants enrolled as inpatients).

References

    1. Bornstein SR, Dalan R, Hopkins D, Mingrone G, Boehm BO. Endocrine and metabolic link to coronavirus infection. Nat. Rev. Endocrinol. 2020;16:297–298. doi: 10.1038/s41574-020-0353-9.
    1. Zhou F, et al. Clinical course and risk factors for mortality of adult inpatients with COVID-19 in Wuhan, China: a retrospective cohort study. Lancet. 2020;395:1054–1062. doi: 10.1016/S0140-6736(20)30566-3.
    1. Guan WJ, et al. Clinical characteristics of coronavirus disease 2019 in China. N. Engl. J. Med. 2020;382:1708–1720. doi: 10.1056/NEJMoa2002032.
    1. Wu C, et al. Risk factors associated with acute respiratory distress syndrome and death in patients with coronavirus disease 2019 pneumonia in Wuhan, China. JAMA Intern. Med. 2020;180:934–943. doi: 10.1001/jamainternmed.2020.0994.
    1. Oguz, S. H. & Okan Yildiz, B. Endocrine disorders and COVID-19. Annu. Rev. Med.10.1146/annurev-med-043021-033509 (2022).
    1. Vassilopoulou E, Bumbacea RS, Pappa AK, Papadopoulos AN, Bumbacea D. Obesity and infection: what have we learned from the COVID-19 pandemic. Front Nutr. 2022;9:931313. doi: 10.3389/fnut.2022.931313.
    1. Zhu L, et al. Association of blood glucose control and outcomes in patients with COVID-19 and preexisting type 2 diabetes. Cell Metab. 2020;31:1068–1077. doi: 10.1016/j.cmet.2020.04.021.
    1. Wu Z, et al. Palmitoylation of SARS-CoV-2 S protein is essential for viral infectivity. Signal Transduct. Target Ther. 2021;6:231. doi: 10.1038/s41392-021-00651-y.
    1. Li D, Liu Y, Lu Y, Gao S, Zhang L. Palmitoylation of SARS-CoV-2 S protein is critical for S-mediated syncytia formation and virus entry. J. Med Virol. 2022;94:342–348. doi: 10.1002/jmv.27339.
    1. Nguyen HT, et al. Spike glycoprotein and host cell determinants of SARS-CoV-2 entry and cytopathic effects. J. Virol. 2020;95:e02304–20.
    1. Ehrlich, E. et al. The SARS-CoV-2 transcriptional metabolic signature in lung epithelium. SSRN Journal10.2139/ssrn.3650499 (2020).
    1. Davies SP, et al. The hyperlipidaemic drug fenofibrate significantly reduces infection by SARS-CoV-2 in cell culture models. Front. Pharm. 2021;12:660490. doi: 10.3389/fphar.2021.660490.
    1. Schaefer MB, et al. Peroxisome proliferator-activated receptor-alpha reduces inflammation and vascular leakage in a murine model of acute lung injury. Eur. Respir. J. 2008;32:1344–1353. doi: 10.1183/09031936.00035808.
    1. Hecker M, et al. PPAR-α activation reduced LPS-induced inflammation in alveolar epithelial cells. Exp. Lung Res. 2015;41:393–403. doi: 10.3109/01902148.2015.1046200.
    1. Huang D, Zhao Q, Liu H, Guo Y, Xu H. PPAR-α agonist WY-14643 inhibits LPS-induced inflammation in synovial fibroblasts via NF-kB pathway. J. Mol. Neurosci. 2016;59:544–553. doi: 10.1007/s12031-016-0775-y.
    1. Nardacci R, et al. Evidences for lipid involvement in SARS-CoV-2 cytopathogenesis. Cell Death Dis. 2021;12:263. doi: 10.1038/s41419-021-03527-9.
    1. Sagan SM, et al. The influence of cholesterol and lipid metabolism on host cell structure and hepatitis C virus replication. Biochem. Cell Biol. 2006;84:67–79. doi: 10.1139/o05-149.
    1. Dorobantu CM, et al. Modulation of the host lipid landscape to promote RNA virus replication: the picornavirus encephalomyocarditis virus converges on the pathway used by hepatitis C virus. PLoS Pathog. 2015;11:e1005185. doi: 10.1371/journal.ppat.1005185.
    1. Yan, B. et al. Characterization of the lipidomic profile of human coronavirus-infected cells: implications for lipid metabolism remodeling upon coronavirus replication. Viruses10.3390/v11010073 (2019).
    1. Lamers MM, Haagmans BL. SARS-CoV-2 pathogenesis. Nat. Rev. Microbiol. 2022;20:270–284. doi: 10.1038/s41579-022-00713-0.
    1. Alipoor SD, et al. COVID-19: molecular and cellular response. Front. Cell Infect. Microbiol. 2021;11:563085. doi: 10.3389/fcimb.2021.563085.
    1. Wiersinga WJ, Rhodes A, Cheng AC, Peacock SJ, Prescott HC. Pathophysiology, transmission, diagnosis and treatment of coronavirus disease 2019 (COVID-19): a review. JAMA. 2020;324:782–793. doi: 10.1001/jama.2020.12839.
    1. Ling H, Luoma JT, Hilleman D. A review of currently available fenofibrate and fenofibric acid formulations. Cardiol. Res. 2013;4:47–55.
    1. Ladabaum U, Mannalithara A, Myer PA, Singh G. Obesity, abdominal obesity, physical activity, and caloric intake in US adults: 1988 to 2010. Am. J. Med. 2014;127:717–727. doi: 10.1016/j.amjmed.2014.02.026.
    1. O’Connor CM, et al. Cardiovascular outcomes with minute ventilation-targeted adaptive servo-ventilation therapy in heart failure: the CAT-HF trial. J. Am. Coll. Cardiol. 2017;69:1577–1587. doi: 10.1016/j.jacc.2017.01.041.
    1. Margulies KB, et al. Effects of liraglutide on clinical stability among patients with advanced heart failure and reduced ejection fraction: a randomized clinical trial. JAMA. 2016;316:500–508. doi: 10.1001/jama.2016.10260.
    1. Felker GM, Maisel AS. A global rank end point for clinical trials in acute heart failure. Circ. Heart Fail. 2010;3:643–646. doi: 10.1161/CIRCHEARTFAILURE.109.926030.
    1. World Health Organization. R&D Blueprint: novel coronavirus. COVID-19 therapeutic trial synopsis draft. 18 February 2020. . Accessed 28 April 2020.
    1. van Elteren PH. On the combination of independent two sample tests of Wilcoxon. Bull. Int. Stat. Inst. 1960;37:351–361.
    1. Fay MP, Malinovsky Y. Confidence intervals of the Mann–Whitney parameter that are compatible with the Wilcoxon Mann–Whitney test. Stat. Med. 2018;37:3991–4006. doi: 10.1002/sim.7890.
    1. Wang D, et al. Clinical characteristics of 138 hospitalized patients with 2019 novel coronavirus-infected pneumonia in Wuhan, China. JAMA. 2020;323:1061–1069. doi: 10.1001/jama.2020.1585.
    1. Hozo SP, Djulbegovic B, Hozo I. Estimating the mean and variance from the median, range, and the size of a sample. BMC Med. Res. Methodol. 2005;5:13. doi: 10.1186/1471-2288-5-13.
    1. Julious SA. Tutorial in biostatistics. Sample sizes for clinical trials with normal data. Stat. Med. 2004;23:1921–1986. doi: 10.1002/sim.1783.
    1. O’Brien PC, Fleming TR. A multiple testing procedure for clinical trials. Biometrics. 1979;35:549–556. doi: 10.2307/2530245.
    1. PASS 16 Power Analysis and Sample Size software. NCSS, LLC. Kaysville, Utah, USA. . (2018).

Source: PubMed

3
Se inscrever