Efficacy of COVID-HIGIV in animal models of SARS-CoV-2 infection

Aruni Jha, Douglas Barker, Jocelyne Lew, Vinoth Manoharan, Jill van Kessel, Robert Haupt, Derek Toth, Matthew Frieman, Darryl Falzarano, Shantha Kodihalli, Aruni Jha, Douglas Barker, Jocelyne Lew, Vinoth Manoharan, Jill van Kessel, Robert Haupt, Derek Toth, Matthew Frieman, Darryl Falzarano, Shantha Kodihalli

Abstract

In late 2019 the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) virus emerged in China and quickly spread into a worldwide pandemic. It has caused millions of hospitalizations and deaths, despite the use of COVID-19 vaccines. Convalescent plasma and monoclonal antibodies emerged as major therapeutic options for treatment of COVID-19. We have developed an anti-SARS-CoV-2 immunoglobulin intravenous (Human) (COVID-HIGIV), a potential improvement from using convalescent plasma. In this report the efficacy of COVID-HIGIV was evaluated in hamster and mouse models of SARS-CoV-2 infection. COVID-HIGIV treatment in both mice and hamsters significantly reduced the viral load in the lungs. Among COVID-HIGIV treated animals, infection-related body weight loss was reduced and the animals regained their baseline body weight faster than the PBS controls. In hamsters, COVID-HIGIV treatment reduced infection-associated lung pathology including lung inflammation, and pneumocyte hypertrophy in the lungs. These results support ongoing trials for outpatient treatment with COVID-HIGIV for safety and efficacy evaluation (NCT04910269, NCT04546581).

Conflict of interest statement

AJ, DB, DT and SK are employees of Emergent BioSolutions. JL, VM, JK and DF (employees of Vaccine and Infectious Disease Organization (VIDO)), and RH and MF (employees of University of Maryland School of Medicine) received funding from Emergent BioSolutions to conduct the work described.

© 2022. The Author(s).

Figures

Figure 1
Figure 1
COVID-HIGIV treatment improves the clinical signs of SARS-CoV-2 infection in mice. Five groups of mice transduced with adenovirus carrying human ACE2 (N = 18) were challenged i.n. with SARS-CoV-2 (WA-1 strain), four groups were treated with different dosages of COVID-HIGIV (6.26, 25, 100 and 400 mg/kg dose) 6 h post infection, and the fifth group was PBS treatment control. An age matched sham infected group (N = 6) was also included as controls. Body weights for all surviving animals were collected each day until the end of the study. Figures represent (A) mean percent body weight, (B) mean percent peak body weight loss relative to the baseline weight on study day 0 before infection.
Figure 2
Figure 2
COVID-HIGIV treatment dose dependently reduced the live virus load in the mouse lungs. Five groups of mice transduced with adenovirus carrying human ACE2 (N = 18) were challenged i.n. with SARS-CoV-2 (WA-1 strain), four groups were treated with different dosages of COVID-HIGIV (6.26, 25, 100 and 400 mg/kg dose) 6 h post infection, and the fifth group was PBS treatment control. Six animals from each group were sacrificed on 2 and 4 dpi to harvest lung for ‘plaque forming unit assay’ for live virus load. Figures represent live virus load (A) 2 dpi, (B) 4 dpi. Statistical comparison was made using One-way ANOVA followed by Dunnett’s pairwise comparison with PBS treated group as control group. P values are reported as * < 0.05, ** < 0.01.
Figure 3
Figure 3
COVID-HIGIV treatment at dose ≥ 400 mg/kg significantly reduced clinical signs of SARS-CoV-2 infection in hamsters. Four groups of Hamsters (N = 18) were challenged i.n. with SARS-CoV-2 (1 × 105 TCID50 in 100 mL; strain—SARS-CoV-2/Canada/ON/VIDO-01/2020), three groups were treated with different dosages of COVID-HIGIV (100, 400 and 800 mg/kg dose) 6 h before infection, and the fourth group was PBS treatment control. Body weights for all surviving animals were collected each day until the end of the study. Figures represent (A) mean percent body weight, (B) mean percent peak body weight loss relative to the baseline weight on study day 0 before infection. Statistical comparison was made using Nested One-way ANOVA followed by Dunnett’s pairwise comparison with PBS treated group as control group P values are reported as * < 0.05, ** < 0.01 and *** < 0.001.
Figure 4
Figure 4
COVID-HIGIV treatment at dose ≥ 400 mg/kg significantly reduced SARS-CoV-2 virus sub-genomic RNA (sg-RNA) in hamster lungs. Four groups of Hamsters (N = 18) were challenged i.n. with SARS-CoV-2 (1 × 105 TCID50 in 100 mL; strain—SARS-CoV-2/Canada/ON/VIDO-01/2020), three groups were treated with different dosages of COVID-HIGIV (100, 400 and 800 mg/kg dose) 6 h before infection, and the fourth group was PBS treatment control. Nine animals from each group were sacrificed at 3 dpi to harvest lung for sg-RNA load by RT-qPCR. Viral sg-RNA load was estimated in the four lobes of right lung (cranial, caudal middle and accessory lobes) and single lobe of the left lung. Viral load for entire right lung was extrapolated by the average viral loads for all four right lung lobes. Similarly, total lung viral load was extrapolated by the average viral loads for all four lobes from right lung and the single lobe from the left lung. Statistical comparison was made using One-way ANOVA followed by Dunnett’s pairwise comparison with PBS treated group as control group. P values are reported as ** < 0.01 and *** < 0.001.
Figure 5
Figure 5
COVID-HIGIV treatment at dose ≥ 400 mg/kg significantly reduced SARS-CoV-2 live virus load in hamster lungs. Four groups of Hamsters (N = 18) were challenged i.n. with SARS-CoV-2 (1 × 105 TCID50 in 100 mL; strain—SARS-CoV-2/Canada/ON/VIDO-01/2020), three groups were treated with different dosages of COVID-HIGIV (100, 400 and 800 mg/kg dose) 6 h before infection, and the fourth group was PBS treatment control. Nine animals from each group were sacrificed at 3 dpi to harvest lung for live virus load estimation by ‘tissue culture infection dose assay’. Live virus load was estimated in the four lobes of right lung (cranial, caudal middle and accessory lobes) and single lobe of the left lung. Viral load for entire right lung was extrapolated by the average viral loads for all four right lung lobes. Similarly, total lung viral load was extrapolated by the average viral loads for all four lobes from right lung and the single lobe from the left lung. Statistical comparison was made using One-way ANOVA followed by Dunnett’s pairwise comparison with PBS treated group as control group. P values are reported as * < 0.05, ** < 0.01 and *** < 0.001.
Figure 6
Figure 6
COVID-HIGIV treatment at dose ≥ 400 mg/kg significantly reduced SARS-CoV-2 infection induced lung pathology in hamsters. Four groups of Hamsters (N = 18) were challenged i.n. with SARS-CoV-2 (1 × 105 TCID50 in 100 mL; strain—SARS-CoV-2/Canada/ON/VIDO-01/2020), three groups were treated with different dosages of COVID-HIGIV (100, 400 and 800 mg/kg dose) 6 h before infection, and the fourth group was PBS treatment control. Nine animals from each group were sacrificed at 10 dpi to harvest left lung for histopathology. Left lobe of the lung was fixed in paraformaldehyde and embedded in wax before sectioning at 5 µm thickness and staining with H&E for light microscopic examination at ×4  magnification. Pathology scoring was performed by the pathologist in a blinded manner.

References

    1. Hui DS, Azhar EI, Madani TA, Ntoumi F, Kock R, Dar O, et al. The continuing 2019-nCoV epidemic threat of novel coronaviruses to global health—The latest 2019 novel coronavirus outbreak in Wuhan China. Int. J. Infect. Dis. 2020;91:264–266. doi: 10.1016/j.ijid.2020.01.009.
    1. Wu JT, Leung K, Leung GM. Nowcasting and forecasting the potential domestic and international spread of the 2019-nCoV outbreak originating in Wuhan, China: A modelling study. Lancet. 2020;395:689–697. doi: 10.1016/S0140-6736(20)30260-9.
    1. WHO_Updates. COVID-19 weekly epidemiological update. (2022). Accessed 28 Sept 2022.
    1. Deng SQ, Peng HJ. Characteristics of and public health responses to the coronavirus disease 2019 outbreak in China. J. Clin. Med. 2020 doi: 10.3390/jcm9020575.
    1. Han Q, Lin Q, Jin S, You L. Coronavirus 2019-nCoV: A brief perspective from the front line. J. Infect. 2020;80:373–377. doi: 10.1016/j.jinf.2020.02.010.
    1. FDA. COVID-19 vaccines. (2022). Accessed 28 Sept 2022.
    1. Rawat P, Sharma D, Srivastava A, Janakiraman V, Gromiha MM. Exploring antibody repurposing for COVID-19: Beyond presumed roles of therapeutic antibodies. Sci. Rep. 2021;11:10220. doi: 10.1038/s41598-021-89621-6.
    1. RECOVERY Collaborative Group. Horby P, Lim WS, Emberson JR, Mafham M, Bell JL, et al. Dexamethasone in hospitalized patients with Covid-19. N. Engl. J. Med. 2021;384:693–704. doi: 10.1056/NEJMoa2021436.
    1. Beigel JH, Tomashek KM, Dodd LE, Mehta AK, Zingman BS, Kalil AC, et al. Remdesivir for the treatment of COVID-19—Final report. N. Engl. J. Med. 2020;383:1813–1826. doi: 10.1056/NEJMoa2007764.
    1. Khoo SH, Fitzgerald R, Fletcher T, Ewings S, Jaki T, Lyon R, et al. Optimal dose and safety of molnupiravir in patients with early SARS-CoV-2: A Phase I, open-label, dose-escalating, randomized controlled study. J. Antimicrob. Chemother. 2021;76:3286–3295. doi: 10.1093/jac/dkab318.
    1. Merck and Ridgeback Biotherapeutics. Merck and ridgeback biotherapeutics provide update on results from MOVe-OUT study of molnupiravir, an investigational oral antiviral medicine, in at risk adults with mild-to-moderate COVID-19. (2021). Accessed 28 Sept 2022.
    1. Pfizer. Pfizer’s novel COVID-19 oral antiviral treatment candidate reduced risk of hospitalization or death by 89% in interim analysis of phase 2/3 EPIC-HR study. (2021). Accessed 28 Sept 2022.
    1. Owen DR, Allerton CMN, Anderson AS, Aschenbrenner L, Avery M, Berritt S, et al. An oral SARS-CoV-2 M(pro) inhibitor clinical candidate for the treatment of COVID-19. Science. 2021 doi: 10.1126/science.abl4784.
    1. FDA. Coronavirus (COVID-19) update: FDA authorizes monoclonal antibodies for treatment of COVID-19 (casirivimab and imdevimab). (2021). Accessed 28 Sept 2022.
    1. FDA. Coronavirus (COVID-19) Update: FDA authorizes monoclonal antibody for treatment of COVID-19 (bamlanivimab). (2020). Accessed 28 Sept 2022.
    1. FDA. Coronavirus (COVID-19) Update: FDA authorizes additional monoclonal antibody for treatment of COVID-19 (sotrovimab). (2021). Accessed 28 September 2022.
    1. FDA. Coronavirus (COVID-19) Update: FDA authorizes monoclonal antibodies for treatment of COVID-19 (bamlanivimab and etesevimab). (2021). Accessed 28 Sept 2022.
    1. Tuccori M, Ferraro S, Convertino I, Cappello E, Valdiserra G, Blandizzi C, et al. Anti-SARS-CoV-2 neutralizing monoclonal antibodies: Clinical pipeline. MAbs. 2020;12:1854149. doi: 10.1080/19420862.2020.1854149.
    1. Taylor PC, Adams AC, Hufford MM, de la Torre I, Winthrop K, Gottlieb RL. Neutralizing monoclonal antibodies for treatment of COVID-19. Nat. Rev. Immunol. 2021;21:382–393. doi: 10.1038/s41577-021-00542-x.
    1. Alexander HE, Leidy G, et al. Hemophilus influenzae meningitis treated with streptomycin. J. Am. Med. Assoc. 1946;132:434–440. doi: 10.1001/jama.1946.02870430014005.
    1. Casadevall A, Dadachova E, Pirofski LA. Passive antibody therapy for infectious diseases. Nat. Rev. Microbiol. 2004;2:695–703. doi: 10.1038/nrmicro974.
    1. Hammon WM, Coriell LL, Wehrle PF, Stokes J., Jr Evaluation of red cross gamma globulin as a prophylactic agent for poliomyelitis. IV. Final report of results based on clinical diagnoses. J. Am. Med. Assoc. 1953;151:1272–1285.
    1. Janeway CA. Use of concentrated human serum gamma-globulin in the prevention and attenuation of measles. Bull. N. Y. Acad. Med. 1945;21:202–222.
    1. Luke TC, Casadevall A, Watowich SJ, Hoffman SL, Beigel JH, Burgess TH. Hark back: Passive immunotherapy for influenza and other serious infections. Crit. Care Med. 2010;38:e66–73. doi: 10.1097/CCM.0b013e3181d44c1e.
    1. Cheng Y, Wong R, Soo YO, Wong WS, Lee CK, Ng MH, et al. Use of convalescent plasma therapy in SARS patients in Hong Kong. Eur. J. Clin. Microbiol. Infect. Dis. 2005;24:44–46. doi: 10.1007/s10096-004-1271-9.
    1. Soo YO, Cheng Y, Wong R, Hui DS, Lee CK, Tsang KK, et al. Retrospective comparison of convalescent plasma with continuing high-dose methylprednisolone treatment in SARS patients. Clin. Microbiol. Infect. 2004;10:676–678. doi: 10.1111/j.1469-0691.2004.00956.x.
    1. Yeh KM, Chiueh TS, Siu LK, Lin JC, Chan PK, Peng MY, et al. Experience of using convalescent plasma for severe acute respiratory syndrome among healthcare workers in a Taiwan hospital. J. Antimicrob. Chemother. 2005;56:919–922. doi: 10.1093/jac/dki346.
    1. Duan K, Liu B, Li C, Zhang H, Yu T, Qu J, et al. Effectiveness of convalescent plasma therapy in severe COVID-19 patients. Proc. Natl. Acad. Sci. USA. 2020;117:9490–9496. doi: 10.1073/pnas.2004168117.
    1. Liu STH, Lin HM, Baine I, Wajnberg A, Gumprecht JP, Rahman F, et al. Convalescent plasma treatment of severe COVID-19: A propensity score-matched control study. Nat. Med. 2020;26:1708–1713. doi: 10.1038/s41591-020-1088-9.
    1. Shen C, Wang Z, Zhao F, Yang Y, Li J, Yuan J, et al. Treatment of 5 critically ill patients with COVID-19 with convalescent plasma. JAMA. 2020;323:1582–1589. doi: 10.1001/jama.2020.4783.
    1. Focosi D, Anderson AO, Tang JW, Tuccori M. Convalescent plasma therapy for COVID-19: State of the art. Clin. Microbiol. Rev. 2020 doi: 10.1128/CMR.00072-20.
    1. Klassen SA, Senefeld JW, Johnson PW, Carter RE, Wiggins CC, Shoham S, et al. The effect of convalescent plasma therapy on mortality among patients with COVID-19: Systematic review and meta-analysis. Mayo Clin. Proc. 2021;96:1262–1275. doi: 10.1016/j.mayocp.2021.02.008.
    1. WHO. WHO recommends against the use of convalescent plasma to treat COVID-19. (2021). Accessed 28 Sept 2022.
    1. Jungbauer C, Weseslindtner L, Weidner L, Gansdorfer S, Farcet MR, Gschaider-Reichhart E, et al. Characterization of 100 sequential SARS-CoV-2 convalescent plasma donations. Transfusion. 2021;61:12–16. doi: 10.1111/trf.16119.
    1. Hassan AO, Case JB, Winkler ES, Thackray LB, Kafai NM, Bailey AL, et al. A SARS-CoV-2 Infection model in mice demonstrates protection by neutralizing antibodies. Cell. 2020 doi: 10.1016/j.cell.2020.06.011.
    1. Zost SJ, Gilchuk P, Case JB, Binshtein E, Chen RE, Nkolola JP, et al. Potently neutralizing and protective human antibodies against SARS-CoV-2. Nature. 2020;584:443–449. doi: 10.1038/s41586-020-2548-6.
    1. Baum A, Ajithdoss D, Copin R, Zhou A, Lanza K, Negron N, et al. REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters. Science. 2020;370:1110–1115. doi: 10.1126/science.abe2402.
    1. Haagmans BL, Noack D, Okba NMA, Li W, Wang C, Bestebroer T, et al. SARS-CoV-2 neutralizing human antibodies protect against lower respiratory tract disease in a hamster model. J. Infect. Dis. 2021 doi: 10.1093/infdis/jiab289.
    1. Imai M, Iwatsuki-Horimoto K, Hatta M, Loeber S, Halfmann PJ, Nakajima N, et al. Syrian hamsters as a small animal model for SARS-CoV-2 infection and countermeasure development. Proc. Natl. Acad. Sci. USA. 2020;117:16587–16595. doi: 10.1073/pnas.2009799117.
    1. Kreye J, Reincke SM, Kornau HC, Sanchez-Sendin E, Corman VM, Liu H, et al. A therapeutic non-self-reactive SARS-CoV-2 antibody protects from lung pathology in a COVID-19 hamster model. Cell. 2020;183:1058–1069.e19. doi: 10.1016/j.cell.2020.09.049.
    1. Alexander MR, Schoeder CT, Brown JA, Smart CD, Moth C, Wikswo JP, et al. Predicting susceptibility to SARS-CoV-2 infection based on structural differences in ACE2 across species. FASEB J. 2020;34:15946–15960. doi: 10.1096/fj.202001808R.
    1. Hancock JT, Rouse RC, Stone E, Greenhough A. Interacting proteins, polymorphisms and the susceptibility of animals to SARS-CoV-2. Animals. 2021 doi: 10.3390/ani11030797.
    1. Bao L, Deng W, Gao H, Xiao C, Liu J, Xue J, et al. Reinfection could not occur in SARS-CoV-2 infected rhesus macaques. bioRxiv. 2020 doi: 10.1101/2020.03.13.990226.
    1. McCray PB, Jr, Pewe L, Wohlford-Lenane C, Hickey M, Manzel L, Shi L, et al. Lethal infection of K18-hACE2 mice infected with severe acute respiratory syndrome coronavirus. J. Virol. 2007;81:813–821. doi: 10.1128/JVI.02012-06.
    1. Tseng CT, Huang C, Newman P, Wang N, Narayanan K, Watts DM, et al. Severe acute respiratory syndrome coronavirus infection of mice transgenic for the human Angiotensin-converting enzyme 2 virus receptor. J. Virol. 2007;81:1162–1173. doi: 10.1128/JVI.01702-06.
    1. Gu H, Chen Q, Yang G, He L, Fan H, Deng YQ, et al. Adaptation of SARS-CoV-2 in BALB/c mice for testing vaccine efficacy. Science. 2020;369:1603–1607. doi: 10.1126/science.abc4730.
    1. Winkler ES, Bailey AL, Kafai NM, Nair S, McCune BT, Yu J, et al. SARS-CoV-2 infection of human ACE2-transgenic mice causes severe lung inflammation and impaired function. Nat. Immunol. 2020;21:1327–1335. doi: 10.1038/s41590-020-0778-2.
    1. Yinda CK, Port JR, Bushmaker T, Offei Owusu I, Purushotham JN, Avanzato VA, et al. K18-hACE2 mice develop respiratory disease resembling severe COVID-19. PLoS Pathog. 2021;17:e1009195. doi: 10.1371/journal.ppat.1009195.
    1. Zost SJ, Gilchuk P, Case JB, Binshtein E, Chen RE, Reidy JX, et al. Potently neutralizing human antibodies that block SARS-CoV-2 receptor binding and protect animals. bioRxiv. 2020 doi: 10.1101/2020.05.22.111005.
    1. Sun J, Zhuang Z, Zheng J, Li K, Wong RL, Liu D, et al. Generation of a broadly useful model for COVID-19 pathogenesis, vaccination and treatment. Cell. 2020;182:734–743.e5. doi: 10.1016/j.cell.2020.06.010.
    1. Francis ME, Goncin U, Kroeker A, Swan C, Ralph R, Lu Y, et al. SARS-CoV-2 infection in the Syrian hamster model causes inflammation as well as type I interferon dysregulation in both respiratory and non-respiratory tissues including the heart and kidney. PLoS Pathog. 2021;17:e1009705. doi: 10.1371/journal.ppat.1009705.
    1. Weston S, Coleman CM, Haupt R, Logue J, Matthews K, Li Y, et al. Broad anti-coronavirus activity of food and drug administration-approved drugs against SARS-CoV-2 in vitro and SARS-CoV in vivo. J. Virol. 2020 doi: 10.1128/JVI.01218-20.
    1. Coleman CM, Frieman MB. Growth and quantification of MERS-CoV infection. Curr. Protoc. Microbiol. 2015;37:11–19. doi: 10.1002/9780471729259.mc15e02s37.
    1. Li W, Chen C, Drelich A, Martinez DR, Gralinski LE, Sun Z, et al. Rapid identification of a human antibody with high prophylactic and therapeutic efficacy in three animal models of SARS-CoV-2 infection. Proc. Natl. Acad. Sci. USA. 2020;117:29832–29838. doi: 10.1073/pnas.2010197117.
    1. Bennett RS, Postnikova EN, Liang J, Gross R, Mazur S, Dixit S, et al. Scalable, micro-neutralization assay for qualitative assessment of SARS-CoV-2 (COVID-19) virus-neutralizing antibodies in human clinical samples. bioRxiv. 2021 doi: 10.1101/2021.03.05.434152.
    1. Chan JF, Zhang AJ, Yuan S, Poon VK, Chan CC, Lee AC, et al. Simulation of the clinical and pathological manifestations of coronavirus disease 2019 (COVID-19) in golden Syrian hamster model: Implications for disease pathogenesis and transmissibility. Clin. Infect. Dis. 2020 doi: 10.1093/cid/ciaa325.
    1. Sia SF, Yan LM, Chin AWH, Fung K, Choy KT, Wong AYL, et al. Pathogenesis and transmission of SARS-CoV-2 in golden hamsters. Nature. 2020;583:834–838. doi: 10.1038/s41586-020-2342-5.
    1. Ramakrishnan MA. Determination of 50% endpoint titer using a simple formula. World J. Virol. 2016;5:85–86. doi: 10.5501/wjv.v5.i2.85.
    1. Zhou X, Moore BB. Lung section staining and microscopy. Bio- Protocol. 2017 doi: 10.21769/BioProtoc.2286.
    1. Katz LM. (A Little) Clarity on convalescent plasma for COVID-19. N. Engl. J. Med. 2021;384:666–668. doi: 10.1056/NEJMe2035678.
    1. Bégin P, Callum J, Jamula E, Cook R, Heddle NM, Tinmouth A, et al. Convalescent plasma for hospitalized patients with COVID-19: An open-label, randomized controlled trial. Nat. Med. 2021 doi: 10.1038/s41591-021-01488-2.
    1. Libster R, Pérez Marc G, Wappner D, Coviello S, Bianchi A, Braem V, et al. Early high-titer plasma therapy to prevent severe COVID-19 in older adults. N. Engl. J. Med. 2021;384:610–618. doi: 10.1056/NEJMoa2033700.
    1. Byrne, J. Inhaled mAb therapy against COVID-19 in the works: 'The product is not dependent on cold chain distribution and storage'. (2021). Accessed 28 Sept 2022.
    1. Chen RE, Winkler ES, Case JB, Aziati ID, Bricker TL, Joshi A, et al. In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains. Nature. 2021 doi: 10.1038/s41586-021-03720-y.
    1. Diamond M, Chen R, Winkler E, Case J, Aziati I, Bricker T, et al. In vivo monoclonal antibody efficacy against SARS-CoV-2 variant strains. Res. Sq. 2021 doi: 10.21203/-448370/v1.
    1. Weisblum Y, Schmidt F, Zhang F, DaSilva J, Poston D, Lorenzi JC, et al. Escape from neutralizing antibodies by SARS-CoV-2 spike protein variants. Elife. 2020 doi: 10.7554/eLife.61312.
    1. Tharmalingam T, Han X, Wozniak A, Saward L. Polyclonal hyper immunoglobulin: A proven treatment and prophylaxis platform for passive immunization to address existing and emerging diseases. Hum. Vaccin. Immunother. 2021 doi: 10.1080/21645515.2021.1886560.
    1. Graham BS, Ambrosino DM. History of passive antibody administration for prevention and treatment of infectious diseases. Curr. Opin. HIV AIDS. 2015;10:129–134. doi: 10.1097/COH.0000000000000154.
    1. Sparrow E, Friede M, Sheikh M, Torvaldsen S. Therapeutic antibodies for infectious diseases. Bull. World Health Organ. 2017;95:235–237. doi: 10.2471/BLT.16.178061.
    1. Stauft CB, Tegenge M, Khurana S, Lee Y, Selvaraj P, Golding H, et al. Pharmacokinetics and efficacy of human hyperimmune intravenous immunoglobulin treatment of SARS-CoV-2 infection in adult Syrian hamsters. Clin. Infect. Dis. 2021 doi: 10.1093/cid/ciab854.
    1. Magyarics Z, Leslie F, Bartko J, Rouha H, Luperchio S, Schorgenhofer C, et al. Randomized, double-blind, placebo-controlled, single-ascending-dose study of the penetration of a monoclonal antibody combination (ASN100) targeting Staphylococcus aureus cytotoxins in the lung epithelial lining fluid of healthy volunteers. Antimicrob. Agents Chemother. 2019 doi: 10.1128/AAC.00350-19.
    1. Shah DK, Betts AM. Antibody biodistribution coefficients: Inferring tissue concentrations of monoclonal antibodies based on the plasma concentrations in several preclinical species and human. MAbs. 2013;5:297–305. doi: 10.4161/mabs.23684.
    1. Wang M, Zhan Y, O'Neil SP, Harris S, Henson C, McEwen A, et al. Quantitative biodistribution of biotherapeutics at whole body, organ and cellular levels by autoradiography. Bioanalysis. 2018;10:1487–1500. doi: 10.4155/bio-2018-0046.
    1. Piepenbrink MS, Park JG, Oladunni FS, Deshpande A, Basu M, Sarkar S, et al. Therapeutic activity of an inhaled potent SARS-CoV-2 neutralizing human monoclonal antibody in hamsters. Cell Rep. Med. 2021;2:100218. doi: 10.1016/j.xcrm.2021.100218.
    1. Lee CY, Lowen AC. Animal models for SARS-CoV-2. Curr. Opin. Virol. 2021;48:73–81. doi: 10.1016/j.coviro.2021.03.009.
    1. Muñoz-Fontela C, Dowling WE, Funnell SGP, Gsell P-S, Riveros-Balta AX, Albrecht RA, et al. Animal models for COVID-19. Nature. 2020;586:509–515. doi: 10.1038/s41586-020-2787-6.
    1. Shou S, Liu M, Yang Y, Kang N, Song Y, Tan D, et al. Animal models for COVID-19: Hamsters, mouse, ferret, mink, tree shrew and non-human primates. Front. Microbiol. 2021 doi: 10.3389/fmicb.2021.626553.

Source: PubMed

3
Tilaa