Randomised controlled trial of intravenous nafamostat mesylate in COVID pneumonitis: Phase 1b/2a experimental study to investigate safety, Pharmacokinetics and Pharmacodynamics

Tom M Quinn, Erin E Gaughan, Annya Bruce, Jean Antonelli, Richard O'Connor, Feng Li, Sarah McNamara, Oliver Koch, Claire MacKintosh, David Dockrell, Timothy Walsh, Kevin G Blyth, Colin Church, Jürgen Schwarze, Cecilia Boz, Asta Valanciute, Matthew Burgess, Philip Emanuel, Bethany Mills, Giulia Rinaldi, Gareth Hardisty, Ross Mills, Emily Gwyer Findlay, Sunny Jabbal, Andrew Duncan, Sinéad Plant, Adam D L Marshall, Irene Young, Kay Russell, Emma Scholefield, Alastair F Nimmo, Islom B Nazarov, Grant C Churchill, James S O McCullagh, Kourosh H Ebrahimi, Colin Ferrett, Kate Templeton, Steve Rannard, Andrew Owen, Anne Moore, Keith Finlayson, Manu Shankar-Hari, John Norrie, Richard A Parker, Ahsan R Akram, Daniel C Anthony, James W Dear, Nik Hirani, Kevin Dhaliwal, Tom M Quinn, Erin E Gaughan, Annya Bruce, Jean Antonelli, Richard O'Connor, Feng Li, Sarah McNamara, Oliver Koch, Claire MacKintosh, David Dockrell, Timothy Walsh, Kevin G Blyth, Colin Church, Jürgen Schwarze, Cecilia Boz, Asta Valanciute, Matthew Burgess, Philip Emanuel, Bethany Mills, Giulia Rinaldi, Gareth Hardisty, Ross Mills, Emily Gwyer Findlay, Sunny Jabbal, Andrew Duncan, Sinéad Plant, Adam D L Marshall, Irene Young, Kay Russell, Emma Scholefield, Alastair F Nimmo, Islom B Nazarov, Grant C Churchill, James S O McCullagh, Kourosh H Ebrahimi, Colin Ferrett, Kate Templeton, Steve Rannard, Andrew Owen, Anne Moore, Keith Finlayson, Manu Shankar-Hari, John Norrie, Richard A Parker, Ahsan R Akram, Daniel C Anthony, James W Dear, Nik Hirani, Kevin Dhaliwal

Abstract

Background: Many repurposed drugs have progressed rapidly to Phase 2 and 3 trials in COVID19 without characterisation of Pharmacokinetics /Pharmacodynamics including safety data. One such drug is nafamostat mesylate.

Methods: We present the findings of a phase Ib/IIa open label, platform randomised controlled trial of intravenous nafamostat in hospitalised patients with confirmed COVID-19 pneumonitis. Patients were assigned randomly to standard of care (SoC), nafamostat or an alternative therapy. Nafamostat was administered as an intravenous infusion at a dose of 0.2 mg/kg/h for a maximum of seven days. The analysis population included those who received any dose of the trial drug and all patients randomised to SoC. The primary outcomes of our trial were the safety and tolerability of intravenous nafamostat as an add on therapy for patients hospitalised with COVID-19 pneumonitis.

Findings: Data is reported from 42 patients, 21 of which were randomly assigned to receive intravenous nafamostat. 86% of nafamostat-treated patients experienced at least one AE compared to 57% of the SoC group. The nafamostat group were significantly more likely to experience at least one AE (posterior mean odds ratio 5.17, 95% credible interval (CI) 1.10 - 26.05) and developed significantly higher plasma creatinine levels (posterior mean difference 10.57 micromol/L, 95% CI 2.43-18.92). An average longer hospital stay was observed in nafamostat patients, alongside a lower rate of oxygen free days (rate ratio 0.55-95% CI 0.31-0.99, respectively). There were no other statistically significant differences in endpoints between nafamostat and SoC. PK data demonstrated that intravenous nafamostat was rapidly broken down to inactive metabolites. We observed no significant anticoagulant effects in thromboelastometry.

Interpretation: In hospitalised patients with COVID-19, we did not observe evidence of anti-inflammatory, anticoagulant or antiviral activity with intravenous nafamostat, and there were additional adverse events.

Funding: DEFINE was funded by LifeArc (an independent medical research charity) under the STOPCOVID award to the University of Edinburgh. We also thank the Oxford University COVID-19 Research Response Fund (BRD00230).

Keywords: Infectious diseases; Nafamostat mesylate; Respiratory medicine; SARS-CoV-2/COVID-19; Total manuscript word count.

Conflict of interest statement

Declaration of interests The authors report no conflict of interests.

Copyright © 2022 The Authors. Published by Elsevier B.V. All rights reserved.

Figures

Figure 1
Figure 1
Consort flow diagram.
Figure 2
Figure 2
a: Clinical course for each patient from hospital admission to discharge or death. Study identifier code, age, gender and ISARIC score (score 0–21 points) given on y-axis with clinical course, time on trial as well as key clinical events reported. b: Kaplan-Meier plot reporting duration of hospital stay following randomisation. Numbers at risk in each trial arm are shown at the bottom of the figure (just above the x-axis).
Figure 2
Figure 2
a: Clinical course for each patient from hospital admission to discharge or death. Study identifier code, age, gender and ISARIC score (score 0–21 points) given on y-axis with clinical course, time on trial as well as key clinical events reported. b: Kaplan-Meier plot reporting duration of hospital stay following randomisation. Numbers at risk in each trial arm are shown at the bottom of the figure (just above the x-axis).
Figure 3
Figure 3
Pharmacokinetics of IV nafamostat. a: Ratio of Post-NM to Pre-nafamostat in each participant in the nafamostat arm, measured by mass spectrometry with a detection m/z [M+H]+=348.1455. b: Assessment of nafamostat metabolite 4-GBA in samples by mass spectrometry at detection of m/z [M+H]+= 180.0767. c: 4-GBA concentration in patients samples taken pre-infusion and at early (2 h) and later time points (>6 h) (plotted for 12 patients in whom matched samples available). **** p value <0.0001, * p value = 0.02. Error bars: SD. d: To assess whether the conversion of nafamostat to 4-GBA could have happened ex-vivo prior to spinning, whole blood was ‘spiked’ with an aliquot of nafamostat (50 ng/ml), and samples were incubated for different periods (t0-t80min). Plasma was generated at the time points and the samples were snap-frozen before analysis performed using RP-MS. A standard curve was prepared to quantify 4-GBA levels in each sample. The area under the total ion chromatogram for each compound in each sample was integrated to obtain the intensity. Error bars: SEM.
Figure 4
Figure 4
a and b – copies per mL of E and S gene respectively in nasopharyngeal and oropharyngeal swabs. c and d – copies per mL of E and S gene respectively in saliva. In both groups, the numbers of participants declined with time representing participant discharge or withdrawal (nafamostat D1 n = 10, D3 n = 9, D5 n = 8. SOC D1 n = 11, D3 n = 10, D 5 n = 3).
Figure 5
Figure 5
Thromboelastometry (ClotPro®). Blood tests performed daily on all patients. Presented here as pooled groups depending on treatment given at time of blood draw. Results pooled to three groups- no anticoagulant treatment (No Tx), on IV NM (nafamostat), on other therapeutic anticoagulation e.g. apixaban/ dalteparin (Anti coag). Significance by one-way ANOVA and Student's t-test. a: Panel evaluating anticoagulant activity (clotting time). **** p <0.001, * p = 0.02. 1: CT EX test, 2: CT FIB test, 3: CT IN test, 4: CT RVV test, 5: CT NA test. b: Panel evaluating antifibrinolytic activity (lysis time and maximum lysis percentage). **** p <0.0001. 6: ML EX test, 7: LT TPA test. c: Panel evaluating clot strength (maximum clot firmness). * p = 0.04. 8: MCF FIB test, 9: MCF EX test. Six ClotPro tests were performed on whole blood samples daily: EX-test: assessment of coagulation with extrinsic activation using recombinant tissue factor. FIB-test: assessment of coagulation with extrinsic activation but in the presence of inhibitors of platelet aggregation. Measures the fibrin contribution to clot strength. IN–test: assessment of coagulation with intrinsic activation using ellagic acid. RVV-test: screening test for direct FXa antagonists, also sensitive to thrombin antagonists. NA-test: non-activated test - assessment of coagulation without an activator of coagulation. TPA-test: assessment of coagulation with fibrinolysis activation (using recombinant tissue plasminogen activator). Measures the effect of anti-fibrinolytic drugs.
Figure 6
Figure 6
Biomarkers over time. SOC (blue)- standard of care. NAF (pink)- Nafamostat Mesylate. a-d: Changes of COVID-19 cytokine storm related cytokines IL-6, IL-8, CXCL-10, IL-1RA. e-l: Clinical biomarkers: creatinine, CRP, D-dimer, neutrophil: lymphocyte ratio, AST, GGT, creatinine kinase and potassium. m-p: Biomarkers of coagulation such as protein C, antithrombin, APTT & fibrinogen. Data presented as mean with best-fit line and 95% confidence intervals by linear regression. All NM patients included in analysis including when off infusion.
Figure 7
Figure 7
Peripheral blood immunophenotyping. Flow cytometric analysis of circulating leukocytes was performed on recruitment prior to treatment and on D4 and D7 on patients in hospital. Histograms illustrate the proportion of: a: Activated CD8+ T cells. b: activated CD4+ T cells. c: CD38+CD27+ antibody secreting cells. d: CXCR3+ CD8+ T cells. e: Classical monocytes (CD14+CD16-), f: Transitional monocytes (CD14+CD16+). g: Non-classical monocytes (CD14lowCD16high). h: CX3CR1 monocytes. i: immature neutrophils. Filled histograms show patients on SoC (D1 n = 13, D4 n = 13 D7 n = 7). Open histograms show NM treated patients (D1 n = 12, D4 n = 12, D7 n = 7). Error Bars show standard deviation.

References

    1. Organization WH . 2020. Novel Coronavirus – China.
    1. Shankar-Hari M, Vale CL, Godolphin PJ, et al. Association between administration of IL-6 antagonists and mortality among patients hospitalized for COVID-19: a meta-analysis. Jama. 2021;326(6):499–518.
    1. Group RC. Dexamethasone in hospitalized patients with Covid-19. New England Journal of Medicine. 2020;384(8):693–704.
    1. Abani O, Abbas A, Abbas F, et al. Tocilizumab in patients admitted to hospital with COVID-19 (RECOVERY): a randomised, controlled, open-label, platform trial. The Lancet. 2021;397(10285):1637–1645.
    1. Yamamoto M, Matsuyama S, Li X, et al. Identification of nafamostat as a potent inhibitor of middle east respiratory syndrome coronavirus s protein-mediated membrane fusion using the split-protein-based cell-cell fusion assay. Antimicrob Agents Chemother. 2016;60(11):6532–6539.
    1. Yamamoto M, Kiso M, Sakai-Tagawa Y, et al. The anticoagulant nafamostat potently inhibits sars-cov-2 s protein-mediated fusion in a cell fusion assay system and viral infection in vitro in a cell-type-dependent manner. Viruses. 2020;12(6):629.
    1. Hoffmann M, Schroeder S, Kleine-Weber H, Müller MA, Drosten C, Pöhlmann S. Nafamostat mesylate blocks activation of SARS-CoV-2: new treatment option for COVID-19. Antimicrobial Agents and Chemotherapy. 2020;64(6):e00754. -20.
    1. Li K, Meyerholz DK, Bartlett JA, McCray PB. The TMPRSS2 inhibitor nafamostat reduces SARS-CoV-2 pulmonary infection in mouse models of COVID-19. mBio. 2021
    1. Choi JY, Kang YJ, Jang HM, et al. Nafamostat mesilate as an anticoagulant during continuous renal replacement therapy in patients with high bleeding risk: a randomized clinical trial. Medicine (Baltimore) 2015;94(52):e2392.
    1. Okajima K, Uchiba M, Mesilate Murakami K.Nafamostat. Cardiovascular Drug Reviews. 1995;13(1):51–65.
    1. Aoyama T, Ino Y, Ozeki M, et al. Pharmacological studies of FUT-175, nafamstat mesilate. I. Inhibition of protease activity in in vitro and in vivo experiments. Jpn J Pharmacol. 1984;35(3):203–227.
    1. Fujii S, Hitomi Y. New synthetic inhibitors of C1r̄, C1 esterase, thrombin, plasmin, kallikrein and trypsin. Biochimica et Biophysica Acta (BBA) - Enzymology. 1981;661(2):342–345.
    1. Gaughan E, Quinn T, Bruce A, et al. DEFINE: a phase iia randomised controlled trial to evaluate repurposed treatments for COVID-19. medRxiv. 2021 2021.05.20.21257513.
    1. Whiting D, DiNardo JA. TEG and ROTEM: Technology and clinical applications. American Journal of Hematology. 2014;89(2):228–232.
    1. Groene P, Sappel SR, Saller T, et al. Functional testing of tranexamic acid effects in patients undergoing elective orthopaedic surgery. J Thromb Thrombolysis. 2021;51(4):989–996.
    1. Kammerer T, Groene P, Sappel SR, et al. Functional testing for tranexamic acid duration of action using modified viscoelastometry. Transfusion Medicine and Hemotherapy. 2021;48(2):109–117.
    1. McCullagh P N, Routledge J.A. 2nd ed. 1983. Generalized Linear Models. 2nd edition ed.
    1. Ko M, Jeon S, Ryu W-S, Kim S. Comparative analysis of antiviral efficacy of FDA-approved drugs against SARS-CoV-2 in human lung cells. Journal of Medical Virology. 2021;93(3):1403–1408.
    1. Jang S, Rhee J-Y. Three cases of treatment with nafamostat in elderly patients with COVID-19 pneumonia who need oxygen therapy. Int J Infect Dis. 2020;96:500–502.
    1. Doi K, Ikeda M, Hayase N, et al. Nafamostat mesylate treatment in combination with favipiravir for patients critically ill with Covid-19: a case series. Critical Care. 2020;24(1):392.
    1. EUnetHTA Joint Action 3 WP4: NAFAMOSTAT FOR THE TREATMENT OF COVID-19. Version 7.0, May 2021
    1. Muto S, Imai M, Asano Y. Mechanisms of hyperkalemia caused by nafamostat mesilate. Gen Pharmacol. 1995;26(8):1627–1632.
    1. Cao YG, Zhang M, Yu D, Shao JP, Chen YC, Liu XQ. A method for quantifying the unstable and highly polar drug nafamostat mesilate in human plasma with optimized solid-phase extraction and ESI-MS detection: more accurate evaluation for pharmacokinetic study. Anal Bioanal Chem. 2008;391(3):1063–1071.
    1. Yamaori S, Fujiyama N, Kushihara M, et al. Involvement of human blood arylesterases and liver microsomal carboxylesterases in nafamostat hydrolysis. Drug Metabolism and Pharmacokinetics. 2006;21(2):147–155.
    1. Di L. The impact of carboxylesterases in drug metabolism and pharmacokinetics. Curr Drug Metab. 2019;20(2):91–102.
    1. group TLdi. COVID-19 Drug interactions, 2021 [accessed 5/8/21.2021].
    1. Cui S, Chen S, Li X, Liu S, Wang F. Prevalence of venous thromboembolism in patients with severe novel coronavirus pneumonia. Journal of Thrombosis and Haemostasis. 2020;18(6):1421–1424.
    1. Malato A, Dentali F, Siragusa S, et al. The impact of deep vein thrombosis in critically ill patients: a meta-analysis of major clinical outcomes. Blood Transfus. 2015;13(4):559–568.
    1. Tang N, Li D, Wang X, Sun Z. Abnormal coagulation parameters are associated with poor prognosis in patients with novel coronavirus pneumonia. Journal of Thrombosis and Haemostasis. 2020;18(4):844–847.
    1. Levi M, Thachil J, Iba T, Levy JH. Coagulation abnormalities and thrombosis in patients with COVID-19. Lancet Haematol. 2020;7(6):e438–ee40.
    1. Yuriditsky E, Horowitz JM, Merchan C, et al. Thromboelastography profiles of critically Ill patients with coronavirus disease 2019. Crit Care Med. 2020;48(9):1319–1326.
    1. Mathew D, Giles JR, Baxter AE, et al. Deep immune profiling of COVID-19 patients reveals distinct immunotypes with therapeutic implications. Science. 2020;369(6508):eabc8511.
    1. Kuri-Cervantes L, Pampena MB, Meng W, et al. Comprehensive mapping of immune perturbations associated with severe COVID-19. Science Immunology. 2020;5(49):eabd7114.
    1. Kirkpatrick DL, Millard J. Evaluation of nafamostat mesylate safety and inhibition of SARS-CoV-2 replication using a 3-dimensional human airway epithelia model. bioRxiv. 2020 2020.09.16.300483.
    1. Zeng C, Evans JP, King T, et al. SARS-CoV-2 Spreads through cell-to-cell transmission. bioRxiv. 2021

Source: PubMed

3
購読する