Development of an aerosol intervention for COVID-19 disease: Tolerability of soluble ACE2 (APN01) administered via nebulizer

Robert H Shoemaker, Reynold A Panettieri Jr, Steven K Libutti, Howard S Hochster, Norman R Watts, Paul T Wingfield, Philipp Starkl, Lisabeth Pimenov, Riem Gawish, Anastasiya Hladik, Sylvia Knapp, Daniel Boring, Jonathan M White, Quentin Lawrence, Jeremy Boone, Jason D Marshall, Rebecca L Matthews, Brian D Cholewa, Jeffrey W Richig, Ben T Chen, David L McCormick, Romana Gugensberger, Sonja Höller, Josef M Penninger, Gerald Wirnsberger, Robert H Shoemaker, Reynold A Panettieri Jr, Steven K Libutti, Howard S Hochster, Norman R Watts, Paul T Wingfield, Philipp Starkl, Lisabeth Pimenov, Riem Gawish, Anastasiya Hladik, Sylvia Knapp, Daniel Boring, Jonathan M White, Quentin Lawrence, Jeremy Boone, Jason D Marshall, Rebecca L Matthews, Brian D Cholewa, Jeffrey W Richig, Ben T Chen, David L McCormick, Romana Gugensberger, Sonja Höller, Josef M Penninger, Gerald Wirnsberger

Abstract

As ACE2 is the critical SARS-CoV-2 receptor, we hypothesized that aerosol administration of clinical grade soluble human recombinant ACE2 (APN01) will neutralize SARS-CoV-2 in the airways, limit spread of infection in the lung, and mitigate lung damage caused by deregulated signaling in the renin-angiotensin (RAS) and Kinin pathways. Here, after demonstrating in vitro neutralization of SARS-CoV-2 by APN01, and after obtaining preliminary evidence of its tolerability and preventive efficacy in a mouse model, we pursued development of an aerosol formulation. As a prerequisite to a clinical trial, we evaluated both virus binding activity and enzymatic activity for cleavage of Ang II following aerosolization. We report successful aerosolization for APN01, retaining viral binding as well as catalytic RAS activity. Dose range-finding and IND-enabling repeat-dose aerosol toxicology testing were conducted in dogs. Twice daily aerosol administration for two weeks at the maximum feasible concentration revealed no notable toxicities. Based on these results, a Phase I clinical trial in healthy volunteers has now been initiated (NCT05065645), with subsequent Phase II testing planned for individuals with SARS-CoV-2 infection.

Conflict of interest statement

Gerald Wirnsberger and Sonja Holler and Romana Gugensberger were employed by Apeiron Biologics A.G. Apeiron supplied the APN01 for study. Josef M. Pettinger was a founder of Apeiron, is a current shareholder and inventor of APN01. David L. McCormick is a Section Editor for PLOS One. Other authors declare no competing interests.

Figures

Fig 1. APN01 neutralization of SARS-CoV-2 in…
Fig 1. APN01 neutralization of SARS-CoV-2 in the absence of cytotoxicity.
Serial dilutions of APN01 were prepared in assay medium (MEM supplemented with 2% fetal bovine serum and 50 μg/mL gentamicin) and a suspension of SARS-CoV-2 (USA-WA1/2020) was added to assess neutralization. For assessment of APN01 on viability, assay medium without virus was added. After one-hour incubation at 37°C, the dilutions were transferred to wells containing Vero E6 target cells (Multiplicity Of Infection, 0.001). Incubation was continued for four days and cell numbers were assessed with a neutral red endpoint. Cytopathic Effect (CPE) was calculated as the average optical density (OD) for replicate infected and treated wells divided by average control OD X 100 (expressed as a percentage). Viability was calculated as the average optical density for replicate uninfected and treated wells divided by average control OD X 100 (expressed as a percentage).
Fig 2. Intranasal APN01 protects from disease…
Fig 2. Intranasal APN01 protects from disease in a mouse-adapted SARS-CoV-2 respiratory infection model.
(A) Experimental outline for infection of male BALB/c mice (n = 5 for both groups) with SARS-CoV-2 (strain maVie16) and daily intranasal treatment with APN01 for five days. (B) Body weight, temperature and survival curves for infected BALB/c mice treated with vehicle control or APN01. Body weights and temperature were compared using mixed-effect analyses. Survival differences were analyzed using a Mantel-Cox test. (C) Lung tissue weight as assessed 5 days after infection of mice; data were analyzed with the Mann-Whitney test. Statistical significances are indicated by asterisks (p-value < 0.05: *; p-value < 0.001: ***).
Fig 3. APN01 binding to the SARS-CoV-2…
Fig 3. APN01 binding to the SARS-CoV-2 RBD is not altered by nebulization.
Binding of increasing doses of APN01 to plate-immobilized RBD domain was assessed by ELISA. Curves depict pre-nebulization samples of APN01 (material remaining as liquid in the nebulizer cup), APN01 collected after nebulization (post-nebulization), and non-treated control APN01. For this experiment, APN01 was aerosolized at 2.5 mg/ml.
Fig 4. APN01 enzymatic activity is not…
Fig 4. APN01 enzymatic activity is not altered by nebulization.
Three samples were tested as defined in the legend to Fig 3. Samples were diluted to APN01 concentrations of 25 ng/ml, 50 ng/ml, and 100 ng/ml and assayed for enzymatic function by cleavage of quenched fluorogenic MAPL-DNP substrate and subsequent measurement of fluorescence activity. Data are plotted as ΔRFU (change in relative fluorescence units) vs. time (min). One representative experiment out of 4 biological replicates is shown.

References

    1. Yan RH, Zhang YY, Li YN, Xia L, Guo YY, Zhou Q. Structural basis for the recognition of SARS-CoV-2 by full-length human ACE2. Science. 2020;367(6485):1444–1448. doi: 10.1126/science.abb2762
    1. Hoffmann M, Kleine-Weber H, Schroeder S, Kruger N, Herrler T, Erichsen S, et al.. SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor. Cell. 2020;181(2):271–280. doi: 10.1016/j.cell.2020.02.052
    1. Walls AC, Park YJ, Tortorici MA, Wall A, McGuire AT, Veesler D. Structure, Function, and Antigenicity of the SARS-CoV-2 Spike Glycoprotein. Cell. 2020;181(2):281–292. doi: 10.1016/j.cell.2020.02.058
    1. Shang J, Ye G, Shi K, Wan YS, Luo CM, Aihara H, et al.. Structural basis of receptor recognition by SARS-CoV-2. Nature. 2020;581(7807):221–224. doi: 10.1038/s41586-020-2179-y
    1. Wang QH, Zhang YF, Wu LL, Niu S, Song CL, Zhang ZY, et al.. Structural and Functional Basis of SARS-CoV-2 Entry by Using Human ACE2. Cell. 2020;181(4):894–904. doi: 10.1016/j.cell.2020.03.045
    1. Wrapp D, Wang NS, Corbett KS, Goldsmith JA, Hsieh CL, Abiona O, et al.. Cryo-EM structure of the 2019-nCoV spike in the prefusion conformation. Science. 2020;367(6483):1260–1263. doi: 10.1126/science.abb2507
    1. Creech CB, Walker SC, Samuels RJ. SARS-CoV-2 Vaccines. Jama-J Am Med Assoc. 2021;325(13):1318–1320. doi: 10.1001/jama.2021.3199
    1. Linsky TW, Vergara R, Codina N, Nelson JW, Walker MJ, Su W, et al.. De novo design of potent and resilient hACE2 decoys to neutralize SARS-CoV-2. Science. 2020;370(6521):1208–1214. doi: 10.1126/science.abe0075
    1. Jiang SB, Hillyer C, Du LY. Neutralizing Antibodies against SARS-CoV-2 and Other Human Coronaviruses. Trends Immunol. 2020;41(5):355–359. doi: 10.1016/j.it.2020.03.007
    1. Wang YQ, Zhang L, Sang L, Ye F, Ruan SC, Zhong B, et al.. Kinetics of viral load and antibody response in relation to COVID-19 severity. J Clin Invest. 2020;130(10):5235–5244. doi: 10.1172/JCI138759
    1. Weinreich DM, Sivapalasingam S, Norton T, Ali S, Gao H, Bhore R, et al.. REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19. New Engl J Med. 2021;384(3):238–251. doi: 10.1056/NEJMoa2035002
    1. Monteil V, Dyczynski M, Lauschke VM, Kwon H, Wirnsberger G, Youhanna S, et al.. Human soluble ACE2 improves the effect of remdesivir in SARS-CoV-2 infection. Embo Mol Med. 2021;13(1). doi: 10.15252/emmm.202013426
    1. Wirnsberger G, Monteil V, Eaton B, Postnikova E, Murphy M, Braunsfeld B, et al.. Clinical grade ACE2 as a universal agent to block SARS-CoV-2 variants. bioRxiv. 2021:2021.09.10.459744. doi: 10.1101/2021.09.10.459744
    1. Monteil V, Stephanie D, Klingström J, Thålin C, Kellner MJ, Christ W, et al.. Clinical grade ACE2 effectively inhibits SARS-CoV-2 Omicron infections. bioRxiv. 2021:2021.12.25.474113.
    1. Kuba K, Imai Y, Rao SA, Gao H, Guo F, Guan B, et al.. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat Med. 2005;11(8):875–879. doi: 10.1038/nm1267
    1. Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, et al.. Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 2005;436(7047):112–116. doi: 10.1038/nature03712
    1. Khan A, Benthin C, Zeno B, Albertson TE, Boyd J, Christie JD, et al.. A pilot clinical trial of recombinant human angiotensin-converting enzyme 2 in acute respiratory distress syndrome. Crit Care. 2017;21.
    1. Sims JJ, Greig JA, Michalson KT, Lian SR, Martino RA, Meggersee R, et al.. Intranasal gene therapy to prevent infection by SARS-CoV-2 variants. Plos Pathog. 2021;17(7). doi: 10.1371/journal.ppat.1009544
    1. Monteil V, Kwon H, Prado P, Hagelkruys A, Wimmer RA, Stahl M, et al.. Inhibition of SARS-CoV-2 Infections in Engineered Human Tissues Using Clinical-Grade Soluble Human ACE2. Cell. 2020;181(4):905–913. doi: 10.1016/j.cell.2020.04.004
    1. Gawish R, Starkl P, Pimenov L, Hladik A, Lakovits K, Oberndorfer F, et al.. ACE2 is the critical in vivo receptor for SARS-CoV-2 in a novel COVID-19 mouse model with TNF- and IFN gamma-driven immunopathology. Elife. 2022;11.
    1. Baazim H, Antonio-Herrera L, Bergthaler A. The interplay of immunology and cachexia in infection and cancer. Nat Rev Immunol. 2021:309–321. doi: 10.1038/s41577-021-00624-w
    1. DeGeorge JJ, Ahn CH, Andrews PA, Brower ME, Choi YS, Chun MY, et al.. Considerations for toxicology studies of respiratory drug products. Regul Toxicol Pharm. 1997;25(2):189–193. doi: 10.1006/rtph.1997.1099
    1. Zoufaly A, Poglitsch M, Aberle JH. Human recombinant soluble ACE2 in severe COVID-19 (vol 8, pg 1154, 2020). Lancet Resp Med. 2020;8(11):E78-E.

Source: PubMed

3
Abonnieren