key: cord-0845474-lejujfxf authors: Yamin, Rachel; Jones, Andrew T; Hoffmann, Hans-Heinrich; Kao, Kevin S; Francis, Rebecca L; Sheahan, Timothy P; Baric, Ralph S; Rice, Charles M; Ravetch, Jeffrey V; Bournazos, Stylianos title: Fc-engineered antibody therapeutics with improved efficacy against COVID-19 date: 2021-05-27 journal: Res Sq DOI: 10.21203/rs.3.rs-555612/v1 sha: 156a6ceabfb207d541ad171959fefc25964c9bba doc_id: 845474 cord_uid: lejujfxf Monoclonal antibodies (mAbs) with neutralizing activity against SARS-CoV-2 have demonstrated clinical benefit in cases of mild to moderate SARS-CoV-2 infection, substantially reducing the risk for hospitalization and severe disease(1–4). Treatment generally requires the administration of high doses of these mAbs with limited efficacy in preventing disease complications or mortality among hospitalized COVID-19 patients(5). Here we report the development and evaluation of Fc-optimized anti-SARS-CoV-2 mAbs with superior potency to prevent or treat COVID-19 disease. In several animal models of COVID-19 disease(6,7), we demonstrate that selective engagement of activating FcγRs results in improved efficacy in both preventing and treating disease-induced weight loss and mortality, significantly reducing the dose required to confer full protection upon SARS-CoV-2 challenge and treatment of pre-infected animals. Our results highlight the importance of FcγR pathways in driving antibody-mediated antiviral immunity, while excluding any pathogenic or disease-enhancing effects of FcγR engagement of anti-SARS-CoV-2 antibodies upon infection. These findings have important implications for the development of Fc-engineered mAbs with optimal Fc effector function and improved clinical efficacy against COVID-19 disease. 3 other mAbs that are currently awaiting regulatory approval offer a clear therapeutic benefit in patients with mild to moderate SARS-CoV-2 infection, reducing the risk for hospitalization and progression to severe COVID-19 disease by over 80% [1] [2] [3] . These results are in stark contrast to the findings from several phase III trials (e.g. ACTIV-3 protocols, NCT04501978) that assessed the therapeutic activity of these mAbs in hospitalized COVID-19 patients. In all cases, such trials failed to reach the primary study endpoints and were terminated prematurely, as none of the neutralizing anti-SARS-CoV-2 mAbs tested offered any therapeutic benefit over standard-ofcare, even when administered at exceedingly high doses or in combination with antivirals like remdesivir 5 . For several acute and chronic viral infections, including influenza virus, RSV, HIV, and Ebola virus, the antiviral activity of IgG antibodies is the outcome of Fab-mediated virus neutralization coupled with the capacity of the Fc domain to mediate effector functions through interactions with Fcγ receptors (FcγRs) expressed on the surface of effector leukocytes 8 . Indeed, FcγR engagement mediates pleiotropic antiviral immune functions, including the clearance of viral particles 9,10 , the cytotoxic elimination of virus-infected cells 11 , as well as the induction of protective T cell responses that contribute to antiviral immunity [12] [13] [14] . In the context of SARS-CoV-2 infection, a growing body of experimental evidence from various animal disease models supports that Fc-FcγR interactions are essential for the in vivo antiviral activity of anti-SARS-CoV-2 mAbs, as loss of the capacity of the Fc domain of these mAbs to engage FcγRs is associated with reduced antiviral activity in vivo [15] [16] [17] [18] . Given the importance of Fc-FcγR interactions in the mAb-mediated protection, maximizing the capacity of clinical neutralizing anti-SARS-CoV-2 mAbs to engage and activate the appropriate FcγR pathways is expected to lower the mAb dose required for the treatment of mild to moderate SARS-CoV-2 infection, as well as improve their therapeutic activity in hospitalized patients with severe COVID-19 disease. Currently, most mAbs in clinical use or development are expressed as human IgG1, which despite its affinity for activating FcγRs also exhibits considerable binding to the inhibitory FcγRIIb, thereby limiting protective Fc effector activities 9 . Additionally, several clinical mAbs (etesevimab, AZD8895, and AZD1061) lack Fc effector function, as they are expressed as Fc domain variants with no FcγR binding activity. This was due to presumptive safety concerns over the capacity of antibodies to exacerbate disease through ADE (antibodydependent enhancement) mechanisms 8 . However, numerous in vivo studies in animal disease models have failed to provide evidence for ADE [15] [16] [17] [18] [19] , and therapeutic administration of high doses of convalescent plasma or neutralizing anti-SARS-CoV-2 mAbs in COVID-19 patients has not been associated with worse disease outcomes [1] [2] [3] 5, 20 . Likewise, comparable safety profiles were evident in clinical trials of neutralizing mAbs with intact or diminished Fc effector function. To assess the role of FcγRs in the mAb-mediated protection and develop mAbs with superior therapeutic potency against COVID-19 disease, we selected well-established small animal SARS-CoV-2 infection models that recapitulate the clinical and pathological features of human COVID-19 disease 6,7,21 . One of these models involves the use of Syrian golden hamsters (Mesocricetus auratus), a species that not only sustains productive virus replication with SARS-CoV-2 clinical isolates, but also exhibits evidence of severe disease upon challenge 6 . However, a major obstacle in the in vivo study of Fc effector activity of human IgG antibodies is the substantial interspecies differences in the affinity of human IgG antibodies for FcγRs expressed by rodent species, such as hamsters 22 . We therefore cloned and expressed the four classes of hamster FcγRs and characterized their binding affinity for human, hamster, and mouse IgG 5 subclasses and Fc variants ( Fig. 1a- To assess the contribution of Fc-FcγR interactions to mAb-mediated protection, we selected neutralizing mAbs in clinical use or development, including casirivimab and imdevimab (REGN cocktail 23 ) and S309/VIR-7831 (Vir 24 ) and expressed them as human IgG1 or as Fc domain variants with defined affinity for hamster FcγRs. In agreement with recent reports 16 , we observed that when mAbs are administered prophylactically, Fc effector function has minimal contribution to the mAb antiviral activity in this model (Fig. 1c) . By contrast, in the therapy setting (d+1 treatment), wild-type, but not FcR null (GRLR) variants are able to suppress lung viremia and prevent weight loss (Fig. 1d ). Since previous studies in mouse models of influenza virus and HIV-1 infection support a key role for FcγRIV in mediating protection by antiviral mAbs 11,25,26 , we compared the in vivo therapeutic activity of two Fc domain variants -GAALIE and V11-that exhibit differential hamster FcγRIV binding activity, but comparable affinity for the other hamster FcγRs (Fig. 1b) . Consistent with a protective role for FcγRIV, the FcγRIV-enhanced variant (GAALIE) demonstrates potent antiviral activity, whereas no therapeutic activity is evident for V11, which exhibits minimal affinity for hamster FcγRIV (Fig. 1e-g) . Although these findings support the importance of Fc-FcγR interactions in the mAb-mediated protection against SARS-CoV-2 infection, their translational relevance is rather limited, given the diversity of FcγR expression on immune cells, the structural complexity of the FcγR family and the divergence of these receptors between humans and other mammalian species 22 . To address this problem, we have previously developed a mouse strain in which only human FcγRs 6 are expressed in a pattern that recapitulates as faithfully as possible the expression pattern seen in human tissues 27 . Human FcγR expression among the various effector leukocyte populations is stable throughout mouse life span and does not differ between young and old mice (Extended Data Fig. 2 ). Infection of old (>15 weeks old), but not young (7 weeks), FcγR humanized mice with the mouse adapted SARS-CoV-2 strain MA10 (characterized in 7 ) results in rapid and often lethal weight loss, which is dependent upon the virus challenge dose ( Fig. 2a-b) . Histological In a model of mAb-mediated therapy of SARS-CoV-2 infection, we observed that the REGN mAb cocktail (expressed as wild-type human IgG1) confers full protection of FcγR humanized mice when administered at 5 mg/kg one day after lethal challenge with SARS-CoV-2 (MA10, 10 4 pfu i.n.) ( Fig. 2c-f ). By contrast, no therapeutic activity is evident in mice lacking FcγRs (FcγRnull) or when mAbs are expressed as variants (GRLR) with minimal affinity for human FcγRs, highlighting the importance of Fc effector function in the therapeutic activity of neutralizing mAbs (Fig. 2c-f ). To determine the mechanisms by which human FcγRs contribute to the mAb-mediated protection, REGN cocktail mAbs were expressed as human IgG1 Fc variants that have been characterized extensively in previous studies 12,28 and exhibit differential affinities for the various classes of human FcγRs (Fig. 3a) . Following the experimental strategy outlined in Figure 3b , we assessed the therapeutic activity of Fc variants of the REGN mAb cocktail at a dose (1 mg/kg) which wild-type human IgG1 confers minimal protection (Fig. 3c ). Consistent with a protective role for activating FcγRs, Fc variants enhanced for either FcγRIIa (GA) or FcγRIII (ALIE) show a trend for improved therapeutic potency over wild-type IgG1, whereas maximal therapeutic activity was evident only for the GAALIE variant, which is enhanced for both FcγRIIa and FcγRIII and has reduced affinity for the inhibitory FcγRIIb ( Fig. 3d -e). These findings suggest that synergy between the two activating FcγRs, FcγRIIa and FcγRIII likely accounts for the therapeutic activity of the GAALIE variant, which achieves the same degree of protection as wild-type IgG1, but at a 5-times lower dose. Importantly, treatment of SARS-CoV-2-infected mice with the GAALIE variant was not associated with enhanced disease not only when administered at a low dose (1 mg/kg), but also at a much higher dose (40 mg/kg) (Extended Data Fig. 4 ). At this dose, which is typically used in the clinical setting, FcR null variants (GRLR) also exhibit full protective activity comparable to the GAALIE variants, suggesting that Fc-independent protection could be achieved once neutralizing mAbs are administered at sufficiently high doses, as has been documented previously for other viral pathogens 25, 26 . Additionally, the observed differences in the in vivo therapeutic activity among Fc variants could not be attributed to differences in Fab-mediated functions, as none of these Fc modifications impact the in vitro neutralization activity, antigen binding specificity, or in vivo half-life (Extended Data Fig. 5-6 ). Similar results were obtained when we assessed the in vivo therapeutic activity of another mAb cocktail (C135+C144; BMS/RU 29, 30 ) that is currently in clinical development and targets the SARS-CoV-2 Spike and mediates potent neutralizing activity 29 . In in vivo titration experiments, we observed that therapeutic administration of the BMS/RU mAb cocktail expressed as wild-type human IgG1 protects FcγR humanized from lethal SARS-CoV-2 challenge in a dosedependent manner (Fig. 3f) . When BMS-RU mAbs are administered to mice at 1 mg/kg, only GAALIE variants, but not wild-type human IgG1 confer protective activity and prevent diseaseinduced weight loss, confirming our findings on the REGN mAb cocktail, which demonstrated improved therapeutic activity when engineered for selective engagement of activating FcγRs (Fig. 3g ). Our findings in hamsters suggest that when neutralizing mAbs are administered prophylactically, Fc-FcγR interactions are not critical for their antiviral activity (Fig. 1c) . However, given the substantial interspecies differences in FcγR biology between hamsters and humans, we assessed the contribution of activating FcγR engagement to the mAb-mediated prophylaxis of SARS-CoV-2-challenged FcγR humanized mice (Fig. 4a) . When administered at a dose where wildtype human IgG1 exhibits no protective activity (0.5 mg/kg) (Fig. 4b) demonstrated that enhanced FcγR engagement is associated with increased pathology 34 . By A P1 stock of the SARS-CoV-2 MA10 strain 7 was amplified in VeroE6 cells obtained from the ATCC that were engineered to stably express TMPRSS2 (VeroE6TMPRSS2). To generate a P2 μg/ml streptomycin (ThermoFisher). All cell lines were maintained at 37 o C at 5% CO2. Expi293F cells (ThermoFisher) were maintained at 37 o C, 8% CO2 in Expi293 expression medium (ThermoFisher) supplemented with 10 U/ml penicillin and 10 μg/ml streptomycin. All cell lines have been tested negative for mycoplasma contamination. In vivo experiments were approved by the Rockefeller University Institutional Animal Care and Use Committee in compliance with federal laws and institutional guidelines. Hamsters and mice were maintained at the Comparative Bioscience Center at the Rockefeller University at a controlled ambient temperature environment with 12-h dark/light cycle. Golden Syrian hamsters were purchased from Charles River laboratories (strain code 049) and maintained in compliance with USDA regulations. FcγR knockout (mFcγRα −/− ; Fcgr1 −/− ) and FcγR humanized mice (mFcγRα −/− , Fcgr1 −/− , hFcγRI + , hFcγRIIaR131 + , hFcγRIIb + , hFcγRIIIaF158 + , and hFcγRIIIb + ) were generated in the C57BL/6 background and characterized in previous studies 12, 27 . Human IgG1 Fc domain variants were generated by site-directed mutagenesis using specific primers as previously described 10 and recombinant IgG antibodies were expressed and purified using previously described protocols 12 . Purity was assessed by SDS-PAGE followed by SafeStain blue staining (ThermoFisher). All antibody preparations were more than 90% pure and endotoxin levels were less than 0.05 EU/mg, as measured by the limulus amebocyte lysate assay. The two plasmid-based HIV/NanoLuc-SARS-CoV-2 pseudovirus system was kindly provided by Dr. Paul Bieniasz (described in 36 ). The S gene was modified by side-directed mutagenesis to introduce the amino acid changes present in the MA10 strain 7 . SARS-CoV-2MA10 pseudovirus particles were generated by transfection of the two plasmid-based system to 293T cells using X-tremeGENE HP DNA transfection reagent (Sigma). All experiments were performed with a Biacore Neutralization activity of IgG1 Fc domain variants was measured as previously described 36 . Briefly, HT1080ACE2 cells were seeded in 96 U-well plates 24 h prior to infection with SARS-CoV-2MA10 pseudoviruses. Pseudovirus particles were pre-incubated with mAbs (four-fold serially diluted starting at 10 μg/ml) for 1 h at 37 o C and then added to a monolayer of Recombinant SARS-CoV-2 RBD was immobilized (1 μg/ml) into high-binding 96-well microtitre plates (Nunc) and after overnight incubation at 4 °C, plates were blocked with PBS plus 2% (w/v) BSA for 2 h. After blocking, plates were incubated for 1 h with serially diluted IgG antibodies or serum samples (1:3 consecutive dilutions in PBS starting at 100 ng/ml), followed by HRP-conjugated goat anti-human IgG (1 h; 1:5,000; Jackson Immunoresearch). Plates were developed using the TMB two-component peroxidase substrate kit (KPL) and reactions were stopped with the addition of 1 M phosphoric acid. Absorbance at 450 nm was immediately recorded using a SpectraMax Plus spectrophotometer (Molecular Devices) and background absorbance from negative control samples was subtracted. Data were collected and analysed using SoftMax Pro v.7.0.2 software (Molecular Devices). Blood was collected into microvette serum gel tubes (Sarstedt) and serum was fractionated by centrifugation (10,000g, 5 min). IgG levels were determined by ELISA following previously described protocols 12 . All animal infection experiments were performed at the Comparative Bioscience Center of the Rockefeller University in animal biosafety level 3 (ABSL-3) containment in compliance with institutional and federal guidelines. Hamsters (males; 6-8 weeks old) were anaesthetized with isoflurane (3%) in a VetFlo high-flow vaporizer followed by an intraperitoneal injection of a ketamine (150 mg/kg) and xylazine (10 mg/kg) mixture. Hamsters were challenged intranasally with 10 5 pfu SARS-CoV-2 (NYC isolate, 100 μl viral inoculum). Mice (males or females; 6-7 or 16-22 weeks old) were anaesthetized with a ketamine (75 mg/kg) and xylazine (15 mg/kg) mixture (administered intraperitoneally) prior to challenge with SARS-CoV-2 (MA10 strain, 10 4 pfu in 30 μl, intranasally). After infection, animals were monitored daily and humanely euthanized by CO2 asphyxiation at endpoints authorized by the Rockefeller University Institutional Animal Care and Use Committee, including any of the following: marked lethargy or inactivity, severe respiratory distress or labored breathing, inability to ambulate, and weight loss of greater than 20% of baseline. Animals were randomized based on age, gender, and weight. Before treatment, we ensured that the mean weight, gender, and age were comparable among the various treatment groups. For antibody-mediated prophylaxis, antibodies were administered intravenously one day before virus challenge, whereas for antibody-mediated therapy, antibodies were administered one day after infection. Antibody dose was calculated as mg/kg. Lungs from euthanized mice were instilled with 10% neutral buffered formalin and fixed overnight by submersion in 10% formalin. Fixed tissues were embedded in paraffin, sectioned at 4 μm thickness, and stained with hematoxylin and eosin. Sections of lung were microscopically evaluated by a board-certified veterinary anatomic pathologist and representative images were captured with an Olympus BX45 light microscope using an SC30 camera with the cellSens Dimension software. Hamsters were euthanized at the indicated timepoints following infection and lung weights were recorded. Lungs were lysed in Trizol (ThermoFisher) and dissociated in gentle MACS M tubes using the gentleMACS Octo Dissociator (Miltenyi Biotec). Samples were transferred into Phasemaker tubes (ThermoFisher) and chloroform was added (200 μl chloroform/1 ml TRIzol). After vigorous shake, tubes were rested for 5 min and then centrifuged for 15 min at 12,000g at Samples that were stained with isotype control antibodies were also blocked with unlabeled anti- anti-human FcγRIIb (clone 2B6), and anti-human FcγRIIIa/b (clone 3G8) (used at 10 μg/ml and incubated for 5 min prior to staining with fluorescently labelled antibodies). Samples were analyzed on an Attune NxT flow cytometer (ThermoFisher) using Attune NxT software v3.1.2 and data were analyzed using FlowJo (v10.7) software. Results from multiple experiments are presented as mean ± s.e.m. One-or two-way ANOVA was used to test for differences in the mean values of quantitative variables, and where statistically significant effects were found, post hoc analysis using Bonferroni (adjusted for multiple comparisons) test was performed. Statistical differences between survival rates were analyzed by comparing Kaplan-Meier curves using the log-rank (Mantel-Cox) test. Data were analyzed with GraphPad Prism v.9.1 software (GraphPad) and p < 0.05 were considered to be statistically significant. SARS-CoV-2 Neutralizing Antibody LY-CoV555 in Outpatients with Covid-19 REGN-COV2, a Neutralizing Antibody Cocktail, in Outpatients with Covid-19 Effect of Bamlanivimab as Monotherapy or in Combination With Etesevimab on Viral Load in Patients With Mild to Moderate COVID-19: A Randomized Clinical Trial Neutralizing monoclonal antibodies for treatment of COVID-19 A Neutralizing Monoclonal Antibody for Hospitalized Patients with Covid-19 Pathogenesis and transmission of SARS-CoV-2 in golden hamsters A Mouse-Adapted SARS-CoV-2 Induces Acute Lung Injury and Mortality in Standard Laboratory Mice The role of IgG Fc receptors in antibodydependent enhancement Signaling by Antibodies: Recent Progress Broadly Neutralizing Anti-HIV-1 Antibodies Require Fc Effector Functions for Enhanced clearance of HIV-1-infected cells by broadly neutralizing antibodies against HIV-1 in vivo Fc-optimized antibodies elicit CD8 immunity to viral respiratory infection Combination anti-HIV-1 antibody therapy is associated with increased virus-specific T cell immunity Early antibody therapy can induce long-lasting immunity to SHIV Neutralizing and protective human monoclonal antibodies recognizing the N-terminal domain of the SARS-CoV-2 spike protein Human neutralizing antibodies against SARS-CoV-2 require intact Fc effector functions for optimal therapeutic protection Antibody potency, effector function, and combinations in protection and therapy for SARS-CoV-2 infection in vivo Live imaging of SARS-CoV-2 infection in mice reveals neutralizing antibodies require Fc function for optimal efficacy. bioRxiv The functions of SARS-CoV-2 neutralizing and infection-enhancing antibodies in vitro and in mice and nonhuman primates. bioRxiv Early Safety Indicators of COVID-19 Convalescent Plasma Animal models of mechanisms of SARS-CoV-2 infection and COVID-19 pathology IgG Fc Receptors: Evolutionary Considerations REGN-COV2 antibodies prevent and treat SARS-CoV-2 infection in rhesus macaques and hamsters Cross-neutralization of SARS-CoV-2 by a human monoclonal SARS-CoV antibody Broadly neutralizing antiinfluenza antibodies require Fc receptor engagement for in vivo protection Broadly neutralizing hemagglutinin stalk-specific antibodies require FcgammaR interactions for protection against influenza virus in vivo Mouse model recapitulating human Fcgamma receptor structural and functional diversity Antibodies targeting sialyl Lewis A mediate tumor clearance through distinct effector pathways mRNA vaccine-elicited antibodies to SARS-CoV-2 and circulating variants Convergent antibody responses to SARS-CoV-2 in convalescent individuals Symptomatic SARS-CoV-2 infections display specific IgG Fc structures. medRxiv, 2020 Afucosylated IgG characterizes enveloped viral responses and correlates with COVID-19 severity Delayed production of neutralizing antibodies correlates with fatal COVID-19 Dissecting strategies to tune the therapeutic potential of SARS-CoV-2-specific monoclonal antibody CR3022 Highly permissive cell lines for subgenomic and genomic hepatitis C virus RNA replication Measuring SARS-CoV-2 neutralizing antibody activity using pseudotyped and chimeric viruses De novo assembly, annotation, and characterization of the whole brain transcriptome of male and female Syrian hamsters Rockefeller University) for technical assistance and for maintaining and genotyping the mouse colonies used in this study and all the members of the Laboratory of the Molecular Genetics and Immunology (Rockefeller University) for discussions. We also thank Sara Borghi (Rockefeller University) for providing the recombinant SARS-CoV-2 RBD, Paul Bieniasz (Rockefeller University) for providing the HT1080ACE2 cell line and plasmids for generating pseudoviruses, Agnès Viale (Memorial Sloan Kettering Cancer Center) for providing the SARS-CoV-2 NYC clinical isolate, Alison Ashbrook and Gaitree McNab (Rockefeller University) for advice on handling BSL-3 agents and processing of tissues from infected animals, David Martinez and Alexandra Schäfer (University of North Carolina at Chapel Hill) for technical assistance, and Sebastian Carrasco (Memorial Sloan Kettering Cancer Center/Weill Cornell Medicine) and staff from the Laboratory Comparative Pathology for histopathology support (with funding from NIH Core Grant P30CA008748) We acknowledge support from the Rockefeller University and Vir Biotechnology Inc. The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. Author contributions: R.Y. designed and performed experiments cloned 25 and characterized hamster FcγRs, performed binding studies, and analyzed data generated and characterized virus stocks generated reagents, performed in vitro experiments, and analyzed the data designed and performed in vivo experiments and generated reagents designed and directed the study Competing interests statement: S.B. and J.V.R. are inventors on a patent (WO2019125846A1) describing the GAALIE variant and its use in therapeutic mAbs. C.M.R. and J.V.R. are SAB members of Vir Biotechnology with equity interests. The datasets generated during and/or analyzed during the current study are available from the corresponding authors on reasonable request. Correspondence and requests for materials should be addressed to SARS-CoV-2-infected hamsters (10 5 pfu, NYC isolate) were treated on day 1 post-infection with Fc variants of the REGN mAb cocktail (5 mg/kg, i.v.) exhibit differential hamster FcγR binding affinity and A/I ratio (calculated based on FcγRIV/FcγRIIb affinity). Weight loss (e, plotted over time (mean ± s.e.m.) or f, as max change) and lung viral titers (g, assessed on day 6 post-infection) were compared by one-way ANOVA Human IgG1 Fc variants with differential affinity for specific classes of human FcγRs were generated for anti-SARS-CoV-2 mAbs. Numbers indicate the fold-change in affinity compared to wild-type human IgG1. b-g, Following the experimental strategy in panel b, SARS-CoV-2-infected FcγR humanized mice were treated (i.v.) at the indicated dose with REGN (c-e) or BMS/RU (f-g) mAb cocktail expressed as wild-type human IgG1 or as Fc variants with differential affinity for human FcγRs. Weight loss (mean ± s.e.m.) (d and g, left 31 panels; e, curves from individual mice) and survival curves (c, f and d, g, right panels) of antibody-treated mice were compared with the corresponding PBS-treated group by two-way ANOVA (Bonferroni post hoc analysis adjusted for multiple comparisons) and log-rank (Mantel-Cox) test, respectively. P values are indicated. NS, not significant. c, n=6 mice per group in two independent experiments Extended Data Fig. 1: Cloning and characterization of the IgG binding activity of hamster Syrian hamster FcγRs were cloned, and their sequences were determined. The FcγR ectodomains are underlined. b-e, The affinity of human IgG1 and Fc variants (b, e, SPR sensorgrams), as well as of mouse (c) and hamster (d) IgG subclass variants for the various classes of hamster FcγRs was determined by surface plasmon resonance (SPR), using soluble hamster FcγR ectodomains FcγR humanized mice (16-22 weeks old) were harvested on day 4 post-infection and evaluated histologically to assess the pathological changes associated with SARS-CoV-2 infection. a, Uninfected mice were characterized by clear alveolar spaces and absence of inflammatory cell infiltrates (low magnification (left panel, 200x); high magnification (center and right panels, 400x). b, In contrast, SARS-CoV-2 infection was associated with perivascular and peribronchial mononuclear leukocyte infiltration (400x, left and center panels), as well as the presence of macrophages and neutrophils in alveoli and necrotic cellular debris in alveolar spaces (600x, right panel). c, In addition, SARS-CoV-2-infected mice exhibited perivenular mixed neutrophilic, histiocytic, and lymphocytic inflammation, reactive endothelium and extravasation of leukocytes inflammation with hemorrhage and single cell necrosis (center and right panels, 400x) High-dose treatment of SARS-CoV-2-infected FcγR humanized mice with anti-SARS-CoV-2 mAbs Fc variants enhanced for activating FcγR binding is not associated with enhanced disease. Following the experimental strategy in Fig. 1b, SARS-CoV-2-infected (MA10, 10 4 pfu, i.n.) FcγR humanized mice (n=3 for PBS and n=5 for mAb-treated groups in one experiment) were treated (i.v.) with 40 mg/kg REGN mAb cocktail expressed as Fc variants with Weight loss (mean ± s.e.m.) and b, survival was compared between GRLR and GAALIE-treated groups by two-way ANOVA (Bonferroni post hoc analysis adjusted for multiple comparisons) and log-rank (Mantel-Cox) test To confirm that changes in the Fc domain have no effect on the neutralization activity and Fab-mediated functions of anti-SARS-CoV-2 mAb, Fc domain variants were characterized in (a-e) in vitro neutralization assays using SARS-CoV-2 (MA10) pseudotyped reporter viruses and (f, g) in ELISA assays using SARS-CoV-2 RBD. n= 1 experiment performed in duplicates. a, b, In vitro neutralization curves and IC50 values of REGN (a) and BMS/RU (b) mAbs against SARS-CoV-2 MA10. (c, f) REGN and (d, g) BMS/RU mAb cocktails were expressed as Fc variants and their in vitro neutralization activity