Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Apr 13.
Published in final edited form as: Immunity. 2021 Apr 13;54(4):815–828.e5. doi: 10.1016/j.immuni.2021.03.009

A Fc-Engineering approach to define functional humoral correlates of immunity against Ebola virus

Bronwyn M Gunn 1,14,#, Richard Lu 1,#, Matthew D Slein 1, Philipp A Ilinykh 2,3, Kai Huang 2,3, Caroline Atyeo 1, Sharon L Schendel 4, Jiyoung Kim 1, Caitlin Cain 1, Vicky Roy 1, Todd J Suscovich 1, Ayato Takada 5, Peter J Halfmann 6, Yoshihiro Kawaoka 6, Matthias G Pauthner 7, Mambu Momo 8, Augustine Goba 8, Lansana Kanneh 8, Kristian G Andersen 7,9, John S Schieffelin 10, Donald Grant 8,11, Robert F Garry 12, Erica Ollmann Saphire 4,*, Alexander Bukreyev 2,3,13,*, Galit Alter 1,*,
PMCID: PMC8111768  NIHMSID: NIHMS1687054  PMID: 33852832

Summary

Protective Ebola virus (EBOV) antibodies have neutralizing activity and induction of Fc-mediated innate immune effector functions. Efforts to enhance Fc-effector functionality often focus on maximizing antibody-dependent cellular cytotoxicity, yet distinct combinations of functions may be critical for antibody-mediated protection. As neutralizing antibodies have been cloned from EBOV disease survivors, we sought to identify survivor Fc-effector profiles to help guide Fc-optimization strategies. Survivors developed a range of functional antibody responses, and we therefore applied a rapid, high-throughput Fc-engineering platform to define the most protective profiles. We generated a library of Fc-variants with identical Fabs from an EBOV neutralizing antibody. Fc-variants with antibody-mediated complement deposition and moderate NK cell activity demonstrated complete protective activity in a stringent in vivo mouse model. Our findings highlight the importance of specific effector functions in antibody-mediated protection and the experimental platform presents a generalizable resource for identifying correlates of immunity to guide therapeutic antibody design.

Keywords: Antibody engineering, Ebola virus, Fc effector function

eTOC Blurb.

Gunn et al. profile Ebola virus disease survivors and apply a platform for engineering antibody Fc domains to define protective profiles. Fc-variants with complement deposition, yet moderate NK cell activity completely protected infected mice from disease. This experimental platform can be used for identifying correlates of immunity to other pathogens, and to guide therapeutic antibody design.

Graphical Abstract

graphic file with name nihms-1687054-f0001.jpg

Introduction

Monoclonal antibodies (mAbs) are among the fastest growing class of drugs for the treatment of cancer, autoimmunity, and infectious diseases (Walker and Burton, 2018; Weiner et al., 2009). The critical role of the antibody constant domain (Fc), both in extending half-life as well as in driving immune killing, has inspired engineering efforts that fully exploit the biological potential of these unique therapeutics. Engineering efforts including modification of Fc-glycosylation and the introduction of point mutations that collectively shape the affinity of Fc binding to a wide array of canonical and non-canonical Fc-receptors or complement provide a means to drive targeted immune clearing activities. For example, alterations in the Fc have been developed to selectively increase antibody-dependent cellular cytotoxicity (ADCC) (Lazar et al., 2006; Shields et al., 2001), antibody-dependent cellular phagocytosis (ADCP) (Lazar et al., 2006; Richards et al., 2008), and antibody-dependent complement deposition (ADCD) (Diebolder et al., 2014; Steurer et al., 1995). Although Fc modifications have been successfully exploited in the context of infectious diseases (Qiu et al., 2014), in some instances antibody Fc-effector function has been linked to both pathological and protective functions, indicating that delicate selection of Fc modifications is needed to maintain balanced Fc-activity. For example, antibody-dependent enhancement (ADE) has been linked to susceptibility to disease caused by dengue virus (Dejnirattisai et al., 2010; Wang et al., 2017), respiratory syncytial virus (Polack et al., 2002), and coronaviruses (Wan et al., 2020). Conversely, immuno-protective functions, including ADCC, have also been noted for these pathogens (Sun et al., 2019; van Erp et al., 2019), again highlighting the need for precise manipulation of Fc-effector function. Moreover, emerging correlate analyses across infectious diseases now provide a unique opportunity to define mechanistic antibody correlates of immunity indicating that discrete profiles of innate immune effector function, rather than pan-functional activity (Ackerman et al., 2018; Gunn et al., 2018), may be needed to guide therapeutic engineering to maximize protection.

In the context of Ebola virus infection, both neutralizing activity and Fc-mediated effector functions are linked to mAb-mediated protection (Gunn et al., 2018; Saphire et al., 2018a; Zeitlin et al., 2011). However, the precise effector functions that track with protection have yet to be defined. Promising mAbs with potent neutralizing activity have been isolated from human survivors of Ebola virus disease (EVD) (Bornholdt et al., 2016; Corti et al., 2016; Flyak et al., 2016; Maruyama et al., 1999), but the characterization of unique Fc-functional profiles of these antibodies and how these functions are linked to protection is only just starting. In this study we sought to link EVD survivor antibody profiling to Fc-engineering to accelerate our mechanistic understanding of protective immune responses that can guide therapeutic design.

The polyfunctional antibody profiles of nearly all EVD survivors are marked by detectable levels of at least one and often many antibody effector functions. To selectively map the precise antibody effector profiles associated with maximal protection from disease, we used a high-throughput platform for rapid, parallel generation of a panel of Fc-engineered mAbs that represent the diversity of functional profiles observed in human survivors. Using the mAb VIC16, which is neutralizing in vitro but offered only modest protection in an animal model Ebola virus infection (Saphire et al., 2018a), a subset of Fc-modifications were selected to represent EVD survivor profiles and tested in a stringent in vivo mouse model for the ability to enhance protection against Ebola virus, allowing the precise identification of Fc-effector functions - that coupled to neutralization- are required to achieve protective immunity against Ebola virus. Our findings reveal that antibodies having complement activity and moderate, rather than robust, NK cell activation were most protective, conferring complete protection from death and eliminating symptoms of infection. These data highlight the utility of coupling unbiased Fc-engineering approaches to complement correlate analyses and accelerate the development of highly protective antibodies to Ebola virus. This platform can likely be extended to engineer antibodies against other deadly pathogens.

Results

Diversity in antibody-mediated innate immune effector functions across human survivors of EVD.

Emerging correlates analyses using libraries of therapeutic mAbs pointed to the importance of phagocytic functions and NK cell activation as key functional properties that can confer protection in mice (Gunn et al., 2018; Saphire et al., 2018a). Given that limited evidence exists for reinfection after EVD survival, we hypothesized that the persisting humoral immune response in survivors points to specific functional humoral immune responses associated with protection. Thus, we profiled the functional humoral immune response in a group of 40 confirmed human survivors of EVD and 26 household contacts collected in Sierra Leone between 2018 and 2019, or ~3–4 years after acute Ebola virus infection to represent the long-lived humoral response. Complete humoral profiling was performed for each sample to measure 66 different features (Figure S1). These measurements captured: induction of six different innate immune effector functions by Ebola glycoprotein (GP)-specific antibodies (ADCP, ADCD, ADNP, ADNKA CD107a, IFNγ, and MIP-1β), levels of IgG, IgA, and IgM binding to Ebola GP, soluble GP (sGP), nucleoprotein (NP), VP40, and VP24, and binding of Ebola GP, sGP, NP, VP40, and VP24-specific IgG to FcγRs (FcγR3A, FcγR2A) and FcαR.

Similar to the profiles previously observed across EVD survivors at 6 months following infection (Gunn et al., 2020), heterogeneous functional profiles were observed for antibodies isolated from survivors and their household contacts at 36–48 months after infection (Figure S1). Unsupervised hierarchical clustering of survivors and contacts based on effector function alone demonstrated that a majority of survivors clustered separately from contacts, with only 3 misclassified survivors and contacts, which highlights the durable presence of antibody effector functions in nearly all survivors (Figure 1A). Among these durable humoral immune responses, antibody-dependent complement activation was least common among the survivors, whereas antibody dependent phagocytosis (ADCP) and NK cell activating antibodies (ADNKA) were present in nearly all survivors. Using K-means clustering, ten major functional clusters were defined within the survivor profiles (Figure 1B; left) that represented unique combinations of functional antibody profiles (Figure 1B; right). A subset of antibodies from survivors induced high levels of all effector functions (groups 1, 6, and 10). Conversely, the majority of survivors exhibited more variable combinations that were marked by low-to-intermediate NK cell function accompanied by at least one type of function related to phagocytosis or complement. Importantly, although emerging data point to a critical role for NK cells in control of EBOV infection (Bornholdt et al., 2019; Corti et al., 2016; Gunn et al., 2018; Warfield et al., 2004), the results of this study suggest that NK cell function does not occur alone.

Figure 1. Diversity in antibody-mediated innate immune effector functions across human survivors of EVD.

Figure 1.

A. Unsupervised hierarchical clustering of cluster EVD survivors (S-value; black text); household contacts (C-value; red text) and a USA-based healthy control (blue text) based on induction of antibody-mediated innate immune effector functions. The heatmap shows the relative value of the indicated functions with purple and green representing the maximum and minimum values, respectively, across the samples as indicated in the legend. A heatmap of all measured antibody features are shown in Figure S1.

B. K-means clustering of 40 EVD survivors into 10 distinct clusters. Principal component analysis of mAbs within each cluster is shown on the left. Composite functional flower plots using the mean values for each cluster were overlaid onto the mirrored PCA on the right graph. Each segment represents a different function, and the size of the segment represents the magnitude of the function relative to the entire panel as indicated in the inset legend. Cluster 9 has no detectable functional activity.

Fc-engineering to define mechanisms of protection.

Given the importance of Fc-effector function for mAb-mediated protection against Ebola virus challenge in animal models (Saphire et al., 2018b), we next performed Fc-engineering of mAbs to gain a more granular understanding of which antibody effector functions are most critical for protection. For Fc-engineering we selected VIC16, a mAb that was part of a 171-antibody panel for which antibody features that correlated with protection were extensively characterized as part of the Viral Hemorrhagic Fever Immunotherapeutics Consortium (VIC) (Saphire et al., 2018a). VIC16 conferred moderate protection in a mouse model of infection with the Zaire strain of Ebola virus (EBOV) (Figure S2A), despite having good neutralization of EBOV in cell culture (Saphire et al., 2018a) through the ability to bind to the EBOV GP internal fusion loop, a key neutralizing epitope targeted by some of the most protective mAbs known to date (Wec et al., 2017; Zhao et al., 2017). Compared to other VIC panel mAbs that bound the internal fusion loop, VIC16 alone exhibited limited to no antibody effector function (Figure S2B). Thus, we hypothesized that generating a library of VIC16 Fc-variants, able to broadly confer distinct Fc-effector functions found in EVD survivors, would provide an opportunity to rescue, improve, and define the minimal requisite antibody effector functions required to achieve protection against lethal challenge with Ebola virus, providing critical clues related to the correlates of humoral immunity to Ebola virus.

A high-throughput cloning approach to rapidly generate Fc-variants: REFORM

Structure-guided mutational screens have identified mutations in both the CH2 and CH3 domains of the antibody Fc domain that alter binding to Fcγ–receptors (FcγRs) (Chu et al., 2008; Lazar et al., 2006; Moldt et al., 2011; Moore et al., 2010; Richards et al., 2008; Shields et al., 2001; Smith et al., 2012; Stavenhagen et al., 2007; Steurer et al., 1995; Xu et al., 2000), complement (Diebolder et al., 2014; Idusogie et al., 2000; Idusogie et al., 2001; Moore et al., 2010; Steurer et al., 1995), and/or the neonatal Fc-receptor (FcRn) (Dall’Acqua et al., 2002; Datta-Mannan et al., 2007; Hinton et al., 2004; Yeung et al., 2009; Zalevsky et al., 2010), resulting in either enhancement or ablation of different innate immune effector functions or antibody half-life. To generate a library of distinct IgG1 Fc domains encoding different Fc mutations, we used a high-throughput cloning platform, Golden Gate (Engler et al., 2008), which takes advantage of unique overhangs generated by digestion with the Type IIS endonuclease BsaI, to ligate all antibody fragments into a pUC vector in a single reaction that avoids the need for sequential cloning of individual antibody domains (variable heavy chain, variable light chain, hinge, constant heavy chain) required in traditional cloning approaches. This method, termed Rationally Engineered and Functionally Optimized Monoclonal antibodies (REFORM), allowed us to rapidly generate antibody plasmids encoding each of the 61 different Fc variants with the same Fab binding domain. The VIC16 variable heavy and light chains were used to generate a panel of VIC16 Fc variants (Table 1). The antibody plasmids were transfected into suspensions of 293F mammalian cells and supernatants were collected 7 days post-transfection for purification of mAbs using purified using protein A/G beads. The expression of each modified mAb was confirmed by SDS-PAGE, which showed that the heavy and light chains had the expected size (Figure S3AB). Production of some mAbs was poor and these were subsequently excluded from downstream analysis. FPLC analysis of the mutated mAbs produced a single peak indicating that monomeric IgG antibody was obtained (Figure S3C).

Table 1. List of REFORM variants generated for this study.

The specific amino acid mutations are shown, and the published impact on antibody-dependent cellular cytotoxicity (ADCC), antibody-dependent cellular phagocytosis (ADCP), antibody-dependent complement deposition (ADCD) or binding to FcRn is indicated.

Fc Variant AA Mutations Description Ref.
IgG1 Human IgG1
LALA L234A/L235A no FcγR binding
LALALS L234A/L235A/M428L/N434S no FcγR binding ↑FcRn (Zalevsky et al., 2010)
LALAPG L234A/L235A/P329G no FcγR binding
N297Q N297Q Aglycosylated
N297QLS N297Q/M428L/N434S Aglycosylated; ↑FcRn (Zalevsky et al., 2010)
SAEAKA S298A/E333A/K334A ↑ ADCC (Lazar et al., 2006; Shields et al., 2001)
SAEAKALS S298A/E333A/K334A/M428L/N434S ↑ ADCC ↑FcRn (Lazar et al., 2006; Shields et al., 2001; Zalevsky et al., 2010)
LPLIL F243L/R292P/Y300L/V305I/P396L ↑ ADCC (Stavenhagen et al., 2007)
LPLILLS F243L/R292P/Y300L/V305I/P396L; M428L/N434S ↑ ADCC ↑ FcRn (Stavenhagen et al., 2007; Zalevsky et al., 2010)
E345R E345R ↑ ADCD (Diebolder et al., 2014)
E345RLS E345R/M428L/N434S ↑ ADCD ↑ FcRn (Diebolder et al., 2014; Zalevsky et al., 2010)
HFST H268F/S324T ↑ ADCD (Moore et al., 2010)
HFSTLS H268F/S324T/M428L/N434S ↑ ADCD ↑FcRN (Moore et al., 2010; Zalevsky et al., 2010)
S324T S324T ↑ ADCD (Moore et al., 2010)
S324TLS S324T/M428L/N434S ↑ ADCD ↑ FcRn (Moore et al., 2010; Zalevsky et al., 2010)
RGY E345R/E430G/S440Y ↑ ADCD (Diebolder et al., 2014)
RGYLS E345R/E430G/S440Y/M428L/N434S ↑ ADCD ↑ FcRn (Diebolder et al., 2014; Zalevsky et al., 2010)
SEHFST S267E/H268F/S324T ↑ ADCD (Moore et al., 2010)
SEHFSTLS S267E/H268F/S324T/M428L/N434S ↑ ADCD ↑ FcRn (Moore et al., 2010; Zalevsky et al., 2010)
KWES K236W/E333S ↑ ADCD (Steurer et al., 1995)
KWESLS K236W/E333S/M428L/N434S ↑ ADCD ↑ FcRn (Steurer et al., 1995; Zalevsky et al., 2010)
E333A E333A ↑ ADCD, ↑ADCC (Idusogie et al., 2000; Idusogie et al., 2001)
I332E I332E ↑ ADCC ↑ADCP (Lazar et al., 2006)
I332ELS I332E/M428L/N434S ↑ ADCC ↑ADCP ↑ FcRn (Lazar et al., 2006; Zalevsky et al., 2010)
IEGA G236A/I332E ↑ ADCC ↑ADCP (Richards et al., 2008)
IEGALS G236A/I332E/M428L/N434S ↑ ADCC, ↑ADCP, ↑ FcRn (Richards et al., 2008; Zalevsky et al., 2010)
SDIE S239D/I332E ↑ ADCC ↑ADCP (Lazar et al., 2006; Richards et al., 2008)
SDIELS S239D/I332E/M428L/N434S ↑ ADCC, ↑ADCP ↑ FcRn (Lazar et al., 2006; Richards et al., 2008; Zalevsky et al., 2010)
SDIEAL S239D/A330L/I332E ↑ ADCC, ↑ADCP (Lazar et al., 2006; Moldt et al., 2011)
SDIEALLS S239D/A330L/I332E/M428L/N434S ↑ ADCC, ↑ADCP, ↑ FcRn (Lazar et al., 2006; Moldt et al., 2011; Zalevsky et al., 2010)
SDIEALYTE S239D/A330L/I332E/M252Y/S254T/T256E ↑ ADCC, ↑ADCP (Dall’Acqua et al., 2002; Lazar et al., 2006)
SDIEALYTELS S239D/A330L/I332E/M252Y/S254T/T256E/M428L/N434S ↑ADCP, ↑ FcRn (Dall’Acqua et al., 2002; Zalevsky et al., 2010)
SDIEGA G236A/S239D/I332E ↑ ADCC, ↑ADCP (Moldt et al., 2011; Richards et al., 2008)
SDIEGALS G236A/S239D/I332E/M428L/N434S ↑ ADCC, ↑ADCP, ↑ FcRn (Moldt et al., 2011; Richards et al., 2008; Zalevsky et al., 2010)
SDIESA S239D/S298A/I332E ↑ ADCC, ↑ADCP
SDIESALS S239D/S298A/I332E/M428L/N434S ↑ ADCC, ↑ADCP, ↑ FcRn (Zalevsky et al., 2010)
SDIEALGA G236A/S239D/A330L/I332E ↑ ADCC, ↑ADCP (Smith et al., 2012)
SDIEALGALS G236A/S239D/A330L/I332E/M428L/N434S ↑ ADCC, ↑ADCP, ↑ FcRn (Smith et al., 2012; Zalevsky et al., 2010)
K326W K236W ↑ ADCD, ↑ADCC (Idusogie et al., 2000; Idusogie et al., 2001)
K326WLS K236W/M428L/N434S ↑ ADCD, ↑ADCC (Idusogie et al., 2000; Idusogie et al., 2001; Zalevsky et al., 2010)
EFTEA G236A/S267E/H268F/S324T/I332E ↑ ADCC, ↑ADCP, ↑ADCD (Moore et al., 2010)
EFTEALS G236A/S267E/H268F/S324T/I332E/M428L/N434S ↑ ADCC, ↑ADCP, ↑ADCD ↑ FcRn (Moore et al., 2010; Zalevsky et al., 2010)
AAA T307A/E380A/N434A ↑ FcRn (Shields et al., 2001)
EANA E380A/N434A ↑ FcRn (Shields et al., 2001)
N434W N434W ↑ FcRn (Yeung et al., 2009)
LS M428L/N434S ↑ FcRn (Zalevsky et al., 2010)
YTE M252Y/S254T/T256E ↓ADCC, ↑ FcRn (Dall’Acqua et al., 2002)
YTELS M252Y/S254T/T256E/M428L/N434S ↓ADCC, ↑ FcRn (Dall’Acqua et al., 2002; Zalevsky et al., 2010)
DVNH D376V/N434H ↑ FcRn (Datta-Mannan et al., 2007)
N434A N434A ↑ FcRn (Shields et al., 2001)
PINH P257I/N434H ↑ FcRn (Datta-Mannan et al., 2007)
PIQI P257I/Q311I ↑ FcRn (Datta-Mannan et al., 2007)
PIQILS P257I/Q311I/M428L/N434S ↑ FcRn (Zalevsky et al., 2010)
QL T250Q/M428L ↑ FcRn (Hinton et al., 2004)
AALS T307A/E380A/M428L/N434S ↑ FcRn (Zalevsky et al., 2010)
P257ILS P257I/M428L/N434S ↑ FcRn (Zalevsky et al., 2010)
QLS T250Q/M428L/N434S ↑ FcRn (Zalevsky et al., 2010)
ALS E333A/M428L/N434S ↑ ADCD, ↑ADCC, ↑ FcRn (Zalevsky et al., 2010)
SELF S267E/L328F ↑ FcγRIIb binding (Chu et al., 2008)
SELFLS S267E/L328F/M428L/N434S ↑ FcγRIIb binding (Chu et al., 2008; Zalevsky et al., 2010)
D376VLS D376V/M428L/N434S ↑ FcRn (Datta-Mannan et al., 2007; Zalevsky et al., 2010)
E380ALS E380A/M428L/N434S ↑ FcRn (Shields et al., 2001; Zalevsky et al., 2010)

Functional characterization of the REFORM antibody panel.

To assess whether the Fc mutations affected the Fab domain, we first examined binding of each of the VIC16 REFORM antibodies (REFORMabs) to EBOV GP by ELISA (Table S1). Although the original VIC16 mAb was a murine IgG1 antibody, chimerization of VIC16 with human IgG Fc domains was previously shown not to affect its ability to bind to EBOV GP antigen (Zeitlin et al., 2011). Similarly, nearly all of the REFORMabs exhibited EBOV GP binding that was comparable to wild type. However, among the panel of 61 mAbs, 8 had significantly decreased (<50%) GP binding relative to the other mAbs in the panel, despite no apparent issues during production of these mAbs. Thus, 53 Fc-variants had binding capacities equivalent to the original IgG1 construct and could be used to examine the role of Fc-effector activity in in vivo protection.

To define the unique functional profiles across the VIC16 REFORMab panel, the ability of each Fc-variant to drive phagocytosis of GP-coated beads by monocytes (ADCP) and neutrophils (ADNP), as well as complement deposition (ADCD) onto GP-coated beads was profiled, as was the ability to induce NK cell degranulation (ADNKA: CD107a) and NK cell secretion of IFNγ (ADNKA: IFNγ) and MIP-1β (ADNKA: MIP-1β) (Figure 2A). All REFORMabs were compared to the original murine IgG1 VIC16 mAb. Interestingly, wild type VIC16-human IgG1 induce only ADCP activity. In contrast, a range of functional activity was clearly observed across the REFORMab panel (Figure 2A). In particular, REFORMabs carrying Fc-variants shown to enhance ADCC activity, such as SDIE (Lazar et al., 2006), showed significantly elevated NK cell activation compared to the original VIC16 and the humanized IgG1 variant. Similarly, increased ADCP activity and ADCD activity were observed for variants with corresponding mutations that are predicted to enhance these functions. Although activation of neutrophils has not been routinely screened across Fc-variants, several mutants, including SDIE, exhibited enhanced ADNP and binding to FcγR3A (Lazar et al., 2006), highlighting additional effector functions that are present in this panel of Fc-variants.

Figure 2. Functional characterization of the REFORMab panel.

Figure 2.

A. VIC16 REFORMabs (5 μg/ml) were evaluated for induction of antibody-dependent monocyte phagocytosis (ADCP), neutrophil phagocytosis (ADNP), complement deposition (ADCD), NK cell degranulation (NK: CD107a), NK cell secretion of IFNγ (NK: IFNγ) and NK secretion of MIP-1β (NK: MIP-1β) following antibody incubation with recombinant Ebola GP. The raw values for each effector function are plotted. The comparison of protection and function between internal fusion loop binding antibodies is shown in Figure S2. The quality control analysis of produced REFORMabs is shown in Figure S3, and representative flow cytometry plots are shown in Figure S4.

B. Plot of IC50 (μg/ml)−1 calculated from an 8-point, 2-fold dilution series of VIC16 REFORMabs in a neutralization assay with VSV pseudovirions expressing EBOV GP.

C. Functional profile of each REFORMab shown as a flower plot, in which each petal represents a different function, and the size of the petal represents the magnitude of the function relative to the entire panel as indicated in the legend.

Given that Fc is known to influence Fab activity (Kunert et al., 2004), we next screened the neutralizing activity of each VIC16 REFORMab using vesicular stomatitis virus (VSV) pseudotyped to display EBOV GP. The neutralizing activity of each REFORMab was compared to the original murine IgG1 VIC16 mAb and the antibody KZ52 that shows strong neutralization activity and was used as a positive control (Parren et al., 2002).The IC50 of the original VIC16 and KZ52 was 0.34μg/ml and 0.08μg/ml, respectively, which is consistent with previously published IC50 values for both mAbs (Parren et al., 2002; Saphire et al., 2018a). The majority of REFORM antibodies showed potent neutralizing activity comparable to the wildtype VIC16 activity (IC50 <1μg/ml), yet 8 mAbs lost neutralization activity while maintaining EBOV GP binding and induction of effector function (Figure 2B). Of these, 7 of 8 (87.5%) were combinations of functional variants paired with the half-life extension mutation, LS (Zalevsky et al., 2010). An additional 20 REFORMabs had an IC50 >1μg/ml and were considered to have lost neutralization potency compared to the rest of the panel. Interestingly, many of these antibodies still induced Fc-effector functions, albeit to a reduced level in some instances, indicating that specific combinations of changes in the Fc domain impacted Fab-mediated neutralizing activity as has been previously observed for engineered HIV-specific mAbs (Kunert et al., 2004).

For an overview of the polyfunctional profile of individual antibodies, for each REFORMab we generated composite flower plots wherein each effector function was represented by a color-coded petal that vary in size according to the relative activity of each function (Figure 2C). We observed robust variation across all the Fc-variants having varying degrees of neutralization (dark blue petal) and addition of individual or combinations of Fc-effector functions. Thus, this panel of REFORMabs provides a heterogeneous source of Fc-variation on a single antibody specificity, providing an opportunity to define specific protective correlates of immunity against EBOV.

Down-selection of REFORM antibodies for in vivo evaluation.

Among the 30 REFORMabs that retained neutralizing activity of IC50 <1μg/ml, heterogeneity was observed in antibody functional profiles (Figure 3A, left). To define classes of functional variants, we used K-means clustering to identify unique functional clusters (Figure 3A, right). Across these 30 REFORMabs, 6 different groups emerged that captured distinct functional profiles including non-functional mAbs (cluster 1), predominantly monofunctional mAbs (clusters 2, 3), and polyfunctional antibodies (clusters 4–6). The monofunctional antibodies in cluster 2 induced only monocyte phagocytosis (ADCP). The sole mAb in cluster 3, KWES, exhibited high levels of complement deposition. Within the polyfunctional clusters, clusters 4 and 5 differed mainly by the magnitude of NK cell-mediated activity, with mAbs in clusters 4 and 5 exhibiting moderate and high levels of NK cell activation, respectively. Cluster 6 contained only one mAb, EFTEA, that induced all functions tested, including high levels of complement activity, yet moderate levels of NK cell activation.

Figure 3. Selection of REFORMabs for in vivo evaluation.

Figure 3.

A. Unsupervised hierarchical clustering with two-way color coding was used to cluster antibodies according to induction of innate immune effector functions (left graph), and K-means clustering was used to group IgG1 VIC16 REFORMabs having a neutralization IC50 of <1μg/ml into 6 distinct clusters (right graph). Results for principal component analysis of mAbs within each cluster are shown, and the inset bi-ray plot indicates the loadings within the plot. The K-means clustering defined-cluster for each antibody is indicated below the heatmap.

B. The five down-selected REFORMabs were produced in larger quantities and re-evaluated in triplicate in dilution curves for neutralization of VSV-EBOV GP pseudovirions, and induction of antibody-dependent complement deposition (ADCD), neutrophil phagocytosis (ADNP), monocyte phagocytosis (ADCP), NK cell degranulation (NK: CD107a), NK cell secretion of IFNγ (NK: IFNγ), and NK secretion of MIP-1β (NK: MIP-1β). Antibodies were assayed for neutralization in a 3-fold dilution curve (3 μg/ml – 0.0048 μg/ml); ADCD in a 2-fold dilution curve (10 μg/ml – 0.16 μg/ml); and ADNP, ADCP, and ADNKA in a 5-fold dilution curve (5 μg/ml – 0.00064 μg/ml).

C. Selected REFORMabs were re-evaluated for induction of effector function, and the AUC for each function across the down-selected in vivo panel is shown in a flower plot

D. Binding affinity for human FcγRs (FcγR3a, FcγR2A, FcγR2B, and FcγR3B) and mouse FcγRs (FcγRI FcγRIV, FcγR3, and FcγR2b) was measured for the selected antibodies by surface plasmon resonance (human FcγR) or biolayer inferometry (mouse FcγR) and KD (M−1) was plotted. The correlation matrix across effector functions and FcγR binding is shown in Figure S5.

E. Binding of low-affinity murine FcγRs (mFcγRIIb and mFcγRIII) to mAb:Ebola GP immune complexes was determined using fluorescently labeled recombinant murine FcγRs at the indicated antibody concentrations. Median Fluorescent Intensity (MFI) of FcγR binding is plotted.

Comparing the functional profiles of the different clusters of VIC16 REFORMabs to the profiles induced in EVD survivors (Figure 1B), we noted that EVD survivor cluster 5, which exhibited moderate levels of NK cell activation, but high complement and phagocytic activity, was similar to REFORM cluster 6 (EFTEA). Higher levels of complement activity in EVD cluster 4 were mimicked by REFORM cluster 3 (KWES), and the high levels of NK cell activation and phagocytic activity yet low complement activation in EVD cluster 2 were represented in REFORM cluster 5. The non-functional EVD cluster 9 was represented by REFORM cluster 1. Thus, the functional diversity identified in EVD survivors was mimicked by the functional diversity in the REFORM panel, and five REFORMabs were selected for in vivo analysis.

Distinct functional profiles track with in vivo antibody-mediated protection.

To define the specific Fc-effector profiles linked to protection, five representative REFORMabs representing the different functional profiles were produced. Binding affinity for Ebola GP, induction of each effector function, and neutralization and was re-measured for the large-scale batches of these five antibodies (Figure 3B). IgG1 was monofunctional, and exhibited only monocyte phagocytosis; KWES induced high levels of complement deposition and low levels of ADCP and NK cell activation; LALA represented a non-functional Fc domain; SDIEALGA captured polyfunctional Fc domains with high NK cell activation and neutrophil phagocytosis; and EFTEA captured all effector functions with high levels of complement deposition, ADCP, and ADNP but moderate levels of NK cell activation (Figure 3C).

The binding affinity of each mAb for both human and mouse FcγRs was measured by surface plasmon resonance (Figure 3D; Table 2). As expected, the SDIEALGA variant known to increase antibody affinity for the activating receptors FcγR3A and FcγR2A (Smith et al., 2012) exhibited a 9-fold and 18-fold increase in KD for FcγR3A and FcγR2A, respectively, compared to the VIC16 human IgG1 (Table 2). The KD of the EFTEA variant for FcγR2A and the inhibitory FcγR2B was 120-fold and 19-fold higher compared to IgG1, respectively, consistent with increases described in previous reports (Moore et al., 2010). Only the SDIEALGA variant showed elevated binding to FcγR3B, a neutrophil-specific FcγR (Bruhns, 2012). The LALA variant showed significantly abrogated binding to the human FcγRs, as expected (Hessell et al., 2007). KWES demonstrated modest enhancement in binding to FcγR3A and FcγR2A, consistent with the moderate levels of NK cell activation and phagocytic activities observed, and no binding to FcγR3B was detected, which likely explains the limited ADNP activity seen for this variant.

Table 2. FcγR binding affinities for the downselected VIC16 REFORMabs.

The KD (μM) values for the indicated antibodies are shown and the fold-change relative to IgG1 is shown in brackets.

Variant huFcγR3A huFcγR2A huFcγR2B huFcγR3B mFcγRI mFcγRIV mFcγR3 mFcγR2b
IgG1 6.1 [1] 30 [1] 23 [1] 25 [1] 0.08 [1] 0.24 [1] 2.83 [1] 14.9 [1]
LALA 1700 [0.003] 892 [0.03] ND ND ND ND 820.3 [0.04] ND
SDIEALGA 0.675 [9] 1.6 [19] 21 [1.1] 3.2 [7.8] 0.1 [0.8] 0.001 [235] 0.01 [0.16] 0.13 [114]
EFTEA 6.1 [1] 0.252 [119] 1.2 [19] 475 [0.05] 0.06 [1.3] 0.12 [2] 1.1 [9] 0.49 [30]
KWES 1.6 [3.8] 8.6 [3.4] 224 [0.12] ND 0.07 [1.1] 0.16 [1.5] 0.02 [0.94] 46 [0.3]

ND = not detected

Mounting evidence points to significant parallels in human IgG1:humanFcγR (hFcγR) and human IgG1:mouse FcγRs (mFcγR) binding profiles (Dekkers et al., 2017; Overdijk et al., 2012), with the exception of mFcγRI. However, whether the REFORMab hFcγR binding profiles translated across species was uncertain, although this feature is key to in vivo efficacy testing in mice. Similar to differences in hFcγR binding profiles, the SDIEALGA variant (14-fold increase) exhibited enhanced binding to mFcγRIV (Figure 3D; Table 2), which in mice is expressed on monocytes/macrophages and neutrophils and is associated with ADCC (Nimmerjahn et al., 2005). EFTEA showed increased affinity for mFcγRIII (9-fold increase), present on all murine myeloid cells and the sole mFcγR expressed on resting murine NK cells (Bruhns, 2012), whereas SDIEALGA exhibited a 6-fold decrease in binding to mFcγRIII compared to IgG1, thus highlighting differential capacities of the variants to engage resting murine NK cells. The SDIEALGA and EFTEA variants showed a modest (4-fold and 2-fold, respectively) increase in binding to mFcγRIIB, the inhibitory receptor present on murine myeloid cells and B cells. Although the KWES variant exhibited an enhanced ability to activate complement, a pathway that is highly conserved across species (Nonaka and Kimura, 2006), its binding to other mFcγRs did not increase. The LALA variant, which showed the expected abrogation of binding to human FcγRs, exhibited abolished binding to mFcγRI, mFcγRIV and mFcγRIIb, and weak binding to mFcγRIII. All other antibodies (IgG1, EFTEA, SDIEALGA, and KWES) demonstrated comparable binding to the high affinity mFcγRI, present on murine dendritic cells. As immune complexes increase avidity of antibodies to low-affinity FcγRs, we also performed analysis of murine FcγRIIb and FcγRIII binding to Ebola GP:mAb immune complexes. The low affinity murine FcγR bound to all of the immune complexes with the exception of the LALA variant immune complexes (Figure 3E). The mFcγRIIb bound highest to the SDIEALGA variant immune complex, consistent with the high binding affinity observed by SPR, whereas KWES showed elevated binding to mFcγRIII compared with the other mAbs (Figure 3E).

To begin to start developing comparative rules sets to translate binding between human and mouse FcRs, we determined the FcγR binding affinities across a subset of the VIC16 REFORM panel to both human and mouse FcγRs (Figure S5A). Strong concordance in binding affinity was observed between mFcγRIV and human huFcγR3A and huFcγR3B, indicating similarity between these receptors and highlighting that Fc mutations aimed at enhancing binding to huFcγR3A will likely also enhance binding to mFcγRIV. Accordingly, affinity to mFcγRIV was strongly associated with human NK cell and neutrophil effector functions (Figure S5B). Moderate concordance was observed between mFcγRIV and huFcγR2A high-affinity variant H131, highlighting parallels in across the non-FcγRI high affinity activating FcγRs between species. Together, these data highlight the unique binding potential of the REFORMabs to both human and mouse FcRs and provide the foundation to build a comparative rule set to translate FcR-meditated activities across species that will help provide critical insights for potential in vivo differential therapeutic benefit.

Dissecting Fc-correlates of immunity in vivo

To define Fc-correlates that contribute to antiviral immunity in vivo, we tested the performance of REFORMabs in a stringent murine model of Ebola virus infection (Bornholdt et al., 2016; Bray et al., 1998; Saphire et al., 2018a; Wec et al., 2017; Zeitlin et al., 2011). For each REFORMab, groups of 5 BALB/c mice were infected with 1,000 plaque-forming units (pfu) of mouse-adapted Ebola virus, 1 day before administration of 100 μg of antibody per mouse. Mice were monitored for survival, clinical disease score, and weight loss for 28 days (Figure 4AB). As expected, PBS-treated mice succumbed to infection by day 5 post-infection, developing clinical disease and weight loss by day 2 post-infection (Figure 4A), highlighting the very aggressive and acute nature of the infection. The LALA variant, which showed loss of hFcγR binding but retained weak binding to mFcγRIII, protected 80% (4/5) of the mice, suggesting that neutralization activity with some mFcγR recruitment was sufficient to protect a majority of animals. However, animals given LALA REFORMabs lost weight and developed disease signs through day 12 (Figure 4B). The animals ultimately survived infection, but the treatment with these REFORMAbs could not prevent disease. Similarly, wild type IgG1, which induced a monofunctional monocyte phagocytic response, also protected 80% of animals, but did not prevent development of disease signs or weight loss. In contrast, the polyfunctional variant SDIEALGA that exhibited robust NK cell activation, but lacked complement activation, protected only 60% of the mice from death, and did not prevent development of disease. The EFTEA and KWES variants conferred 100% survival in treated animals, and, remarkably, the animals showed no evidence of development of disease. Only one animal treated with the EFTEA variant showed mild clinical signs at day 5 post-infection (Figure 4A). Both EFTEA and KWES induced high levels of complement activation and exhibited polyfunctional activity, with moderate induction of NK cell-mediated activity and monocyte phagocytosis.

Figure 4. Distinct functional profiles track with in vivo antibody-mediated protection.

Figure 4.

A. Selected REFORMAbs were administered to mice (100 μg/mouse) 24 hours after infection with 1,000 pfu of mouse-adapted Ebola virus. Mice were monitored for 28 days post-infection, and survival of mice is plotted.

B. Clinical disease score (top row) and percentage of starting weight (bottom row) of mice is shown with different colors representing individual mice. The percentage survival is indicated in the top right corner panels showing clinical disease score for each mAb.

C. Selected mAbs were administered to mice at a reduced dose (30 μg/mouse) 24 hours after infection with 1,000 pfu of mouse-adapted Ebola virus. Mice were monitored for 28 days post-infection, and the survival is plotted.

D. Clinical disease score (top row) and percentage of starting weight (bottom row) of mice treated with 30 μg of the indicated antibody is shown with different colors representing individual mice.

To further confirm the protective effects of EFTEA and KWES, a second group of mice were dosed with approximately one-third (30 μg/mouse) of the original antibody dose (Figure 4CD). Animals treated with wild type, monofunctional IgG1 at this reduced dose all succumbed to Ebola virus infection. In contrast, animals treated with reduced doses of either EFTEA and KWES variants rapidly recovered from symptoms and disease and all survived (Figure 4CD). These data suggest that even at low doses of antibodies carrying these modifications, animals could recover and survive a lethal viral challenge, which points to the critical importance of complement and low-level ADCC as mechanistic correlates of protection.

Discussion

In-depth characterization of antibodies isolated from human survivors of EVD identified critical neutralizing epitopes that can be harnessed in therapeutic treatment of infection (Bornholdt et al., 2016; Corti et al., 2016; Flyak et al., 2016; Maruyama et al., 1999). Given our emerging appreciation for the role of Fc-effector function in the control/clearance of EBOV infection (Gunn et al., 2018; Misasi et al., 2016; Olinger et al., 2012; Zeitlin et al., 2011), we hypothesized that a similar analysis of human EVD survivors could help identify specific Fc-effector profiles that are most critical for control and resolution of disease. Nearly all EVD survivors possessed functional EBOV-specific humoral immunity, albeit with variable levels and combinations of antibody effector functions. Thus, to precisely define the mechanistic Fc-effector correlates of protection, we performed Fc-engineering. While Fc-modifications that potentiate NK cell mediated ADCC have largely been explored in EBOV-monoclonal mediated engineering (Bornholdt et al., 2019; Zeitlin et al., 2011), here we exploited an agnostic Fc-engineering approach to generate a minimal set of Fc-variants on a single Fab able to trigger antibody profiles similar to those observed in EVD survivors. Surprisingly, passive transfer of Fc-engineered variants highlighted the critical role of robust complement activation and moderate NK cell activity as key mechanistic correlates of protection against EBOV. These data point to a critical role not only for combinations of effector functions, but also for the levels of induction, which may be key to antibody-mediated protection, wherein the most protective mAbs, KWES and EFTEA, completely abrogated development of clinical disease at a 100μg dose, and survival even at low antibody doses. These data highlight the disease-modifying effects of Fc-engineering, yet also highlight the critical need to customize Fc-design to maximize the recruitment of the functional correlates of immunity to the pathogen of interest.

Accumulating data points to similarities in mFcγR:IgG1 and hFcγR:IgG1 responses (Overdijk et al., 2012), although translation of findings for Fc engineering of a human IgG1 requires careful dissection of binding profiles across species. Recent analysis of the binding affinity of human IgG1 to both human and murine FcRs suggests that hIgG1 binding profiles are more comparable to mouse IgG2a than previously thought (Dekkers et al., 2017). Along these lines, our analysis with human IgG1:mFcγRs support and extend these findings, suggesting that engineered hIgG1 variants can skew binding to mouse FcRs. The Fc-silencing LALA variant, for example, has been used in mouse models to investigate the requirement for FcγR-mediated effector function in antibody-mediated protection (Gilchuk et al., 2018; Ilinykh et al., 2020). As expected, the LALA variant displayed abrogated binding to both human and mouse FcγRs, clearly demonstrating comparable activity across species, and offering a simple means to investigate the role of Fc-effector function as a correlate of protection against Ebola virus. Similarly, given the highly conserved nature of complement activity across species, we observed strong concordance in antibody-mediated complement deposition between guinea pig-derived and human-derived complement (Fischinger et al., 2019), pointing to translatability across species.

However, antibodies do not bind to individual FcRs in vivo, but instead bind to collections of FcRs present on distinct innate immune cells in vivo in the setting of immune-complexed pathogens, infected cells, or debris. We chose our down-selected panel to reflect the effector profiles in human survivors of EVD yet as protective efficacy must be tested in animal models, a complete picture is required to fully capture the cross-species translation of antibodies from animal models to humans. Thus here, each REFORMab was tested in human and murine functional assays as well as in their capacity to interact with mouse and human FcγRs. The data demonstrate remarkable concordance in expected FcR-binding:functional activity, particularly between murine FcγRIV and human FcγR3A/B binding and activation of human NK cells and neutrophils (Figure S5). Future cross-species translation must take both antibody:FcR binding profiles into consideration, but also cell type specific FcR expression profile differences across species to fully translate in vivo observations across the species. Specifically, differences in expression of FcγRs on murine and human innate immune cells, their localization, and their differential innate immune effector functions can help resolve mechanisms across the species. In the context of our data presented here, the Fc engineered mAbs, except for LALA, showed similar binding to mFcγRIII, the only FcγR expressed on murine NK cells. However, the SDIEALGA variant induced the highest levels of human NK cell degranulation, representative of ADCC, and displayed the highest affinity for mFcγRIV, the FcγR that drives ADCC in mice via macrophage and neutrophils rather than NK cells. Thus, these data suggest that SDIEALGA likely drives elevated levels of ADCC in mice but through different cell types in mice and humans, suggesting that identifying cross-species functional-matches that track with protection may be more informative than simply defining Fc-receptor binding similarities. Thus, developing cross-species maps using modified monoclonal antibodies with the same specificities, matching half-lives/biodistributions, but differential binding properties to FcRs across species may provide the critical means to help define mechanistic translatability across the models.

Immunopathology is a hallmark of EVD (Baseler et al., 2017), and although ADCC has been proposed to be a critical mechanism of protection against EBOV (Corti et al., 2016), the SDIEALGA variant, which exhibited the highest binding to mFcγRIV and human NK cell activation of the panel, showed the lowest protection activity among the VIC16 variants, indicating that maximal induction ADCC may be detrimental to antibody-mediated protection. Previous studies suggest that SDIEALGA exhibits similar pharmacokinetics, suggesting that reduced protection is unlikely related to enhanced clearance of the antibody variant (DiLillo et al., 2014). Thus, the ability to induce just enough activation without overactivation of innate immune cells may be a critical parameter of antibody-mediated protection. Of note, antibody-mediated activation of human NK cells was observed in most survivors, and the most protective REFORMabs retained moderate NK cell functions, further supporting the hypothesis that a balanced, but potentially non-overactive ADCC response in the setting of complement activation may be critical to fully contain and control this lethal pathogen.

The rapid emergence of SARS-CoV-2 and resulting COVID-19 pandemic has highlighted the urgent need for the rapid development of highly effective and potent therapeutics to treat infected patients. The unprecedented speed at which neutralizing antibodies against SARS-CoV-2 have been identified allows for the rapid development of antibody-based therapeutics to help slow and end the pandemic (Ju et al., 2020; Pinto et al., 2020). However, humoral immune responses have been linked to both protection (Agnihothram et al., 2013; Corti et al., 2015; Deming et al., 2006) and pathology (Jaume et al., 2011; Liu et al., 2019; Wang et al., 2014) following coronavirus infections, rendering Fc-engineering efforts quite contentious. Nonetheless, the REFORM platform can accelerate the development of highly potent therapeutics against SARS-CoV-2 through the rapid generation of libraries of Fc-variants that are tailored to have different Fc-effector functions. These variants can be tested in animal models to define the precise Fc-effector profiles associated with pathogenesis or protection. Moreover, linked to intense pharmacokinetic and pharmacodynamic profiling of each variant, therapeutically effective antibodies with ideal therapeutic activities may be defined. Ultimately, modeling binding profiles across FcγRs:Fc effector functions and biodistribution in animal models and humans may provide bridging information to guide the accelerated development of highly effective therapeutics as countermeasures for emerging diseases.

Importantly, the Fc-effector mechanistic correlates of immunity likely differ across pathogens as well as across epitopes even within the same pathogen, with the latter related to antibody angle-of-attack or stoichiometry (Bournazos et al., 2019; He et al., 2016; Saphire et al., 2018b). Notably, the antibody panel examined in this study does not include all published IgG1 Fc-point mutants, nor does it include modifications of other IgG subclasses, IgA, and IgM, which represent a much larger explorable immune space that is naturally exploited during an immune response to drive protective immunity. However, the approach detailed here offers an unbiased systems biology approach to broadly control Fc-effector function and develop large libraries of “functionally unique” reagents that can be tested for in vivo or in vitro efficacy. Thus, the REFORM platform offers a unique approach to rapidly screen Fc-engineered mAbs to define the mechanism of antibody action against any target pathogen to accelerate monoclonal therapeutic or vaccine design against EBOV and emerging pathogens like SARS-CoV-2.

Limitations of the Study

The REFORMAb platform utilizes a rapid cloning platform to generate a panel of functionally diverse human antibodies with identical Fab domains. As the ultimate goal of this work is to guide the development of new antibody-based therapies for human Ebola virus infection, we performed effector function analysis using human innate immune cells. However, we used a murine challenge model to determine whether the protective efficacy of a neutralizing Ebola-specific antibody, VIC16, is enhanced by the incorporation of specific Fc mutations, thus creating a complex scenario in interpreting the mechanism of protection of human antibodies. Some effector functions, such as complement, are readily translated across species, and thus the finding that the REFORMabs with high complement activity protected mice from disease support the hypothesis that antibody-mediated complement activation may also be protective in humans. In contrast, ADCC activity in mice and humans are mediated by different immune cells and different FcγRs – macrophage and neutrophils mediate ADCC in mice via FcγRIV whereas NK cells are primarily responsible for ADCC in humans via FcγR3A. We observed a strong association between activation of human NK cells and antibody binding to FcγR3A across the REFORMabs and between binding of the REFORMab panel to murine FcγRIV and human FcγR3A, suggesting that activation of human NK cells may serve as a surrogate for ADCC in mice, albeit mediated by a different cell type in the mouse challenge model. Concordance between antibody-dependent phagocytosis by mouse and human innate immune cells have been previously observed (Gunn et al., 2018), yet were not compared in this study. The development of bridging analyses to translate findings across species is ongoing, and the identification of the effector function that tracks with protection rather than the cell type that mediates it may enable better translation between species. Dissecting the similarities and differences between animal models and humans will enable an improved framework towards effective translation of antibody efficacy across species to ultimately leverage the immune system to fight the infection in humans inspired by functional correlates of immunity in animal models.

STAR Methods and Materials.

RESOURCE AVAILABILITY

Lead Contact.

Further information and requests for resources and reagents should be directed to and will be fulfilled by Lead Contact, Dr. Galit Alter (galter@mgh.harvard.edu)

Materials Availability.

Plasmids and antibodies generated in this study may be requested with a material transfer agreement.

Data and Code Availability.

The datasets generated in this study are available on Mendeley (doi: 10.17632/9b2h8p9gtc.1)

EXPERIMENTAL MODEL AND SUBJECT DETAILS

Patient samples.

Plasma samples were collected from individuals with documented clinical history of EVD approximately 3–4 years after infection and from household contacts of EVD survivors. At the time of acute disease in 2014, EVD patients received standard of care at Kenema Government Hospital (e.g., intravenous fluid replacement and administration of antibiotics as determined by the treating physician). Peripheral blood mononuclear cells were collected from MGH blood bank donations for use in innate immune effector assays. All subjects provided written consent. This study was approved by the Institutional Review Board of the Human Subjects Committee of MGH, Tulane University Institutional Review Board, and the Sierra Leone Ethics and Scientific Review Committee.

Primary human innate immune cells.

Innate immune effector cells were isolated from fresh peripheral blood samples collected by the Ragon Institute or the MGH Blood bank from healthy human volunteers. All subjects signed informed consent and the study was approved by the MGH Institutional Review Board.

Cell lines.

Freestyle 293F cells were purchased through Thermo Fisher and maintained in Freestyle 293F medium (Thermo Fisher) in a HT Infors shaking incubator at 125 RPM, 37C 8% CO2. The human monocyte cell line, THP-1, was purchased through ATCC. Cells were maintained in RPMI1640 supplemented with 10% fetal bovine serum, L-Glutamine, penicillin/streptomycin, and 55μM beta-mercaptoethanol in a humidified incubator at 5% CO2.

Animals.

Naïve seven-week-old BALB/c mice used for challenge studies were purchased from Charles River Laboratory) were housed in microisolator cages in the ABSL-4 facility at the Galveston National Laboratory.

METHOD DETAILS.

Plasmid design and construction.

Four donor pUC plasmids encoding the variable heavy domain, the Fc domain, a furin P2A sequence, or the variable light domain of an antibody surrounded by BsaI sites were designed. Two destination vectors were designed that contained an IL-2 secretion signal, the suicide gene ccdB flanked by BsaI sites, and either the kappa or lambda light chain sequences. A library of 63 Fc variants were synthesized and cloned into the pUC donor plasmids for the Fc domain (Table 1). The 5 plasmids were combined in a single digestion-ligation reaction to generate an expression plasmid encoding both the heavy chain and light chain of a monoclonal antibody as follows: 50 ng of each donor and destination plasmid, 20 units BsaI-HF (NEB), 200 units T4 ligase, and 0.67 mM ATP in 1X T4 ligase buffer (NEB) supplemented with BSA were combined and incubated at 37 °C for 1 h followed by incubation at 50 °C for 15 min and 80 °C for 15 min. Ligation was performed by adding 200 units T4 ligase followed by incubation at 16°C for 30 min. Inclusion of a ccdB gene that is removed during the digestion-ligation process increases the likelihood that the recombinant plasmid is selected as opposed to the parental vector. Two alternate destination vectors were created for expression of only the light chain to optimize the heavy chain to light chain ratio during antibody production. Ligation products (2 μl) were transformed into Stellar competent cells (Clontech) and plated onto agar plates with kanamycin. The resulting colonies were screened and sequenced to verify the identity of each new plasmid.

Production of antibodies in mammalian cells.

Plasmids were expanded and transfected into 293F suspension cells grown in Freestyle™ 293 Expression media (Gibco). Cells were seeded two days pre-transfection at a density of 0.4×106 cells/mL in 50 mL in a 125 mL baffled Erlenmeyer flask with a vented closure. On the day of transfection, cells were counted again and the cell density was adjusted to 1.2×106 cells/mL in 50 mL media. Total DNA (25 μg) was transfected into cells using Polyethylenimine (PEI) (Polysciences) at 1 μg/μl in a ratio of 3 μg PEI to 1 μg DNA. Cell culture supernatants were harvested 5 days post-transfection. Supernatants were incubated with Protein G magnetic beads (Millipore) overnight at 4 °C. The Protein G Beads were washed 4 times with 1x PBS before antibodies were eluted using Pierce™ IgG Elution Buffer (Thermo Fisher Scientific) and neutralized with Tris-HCl pH 8.0 at a ratio of 1:10. For 500 mL production volumes, cells were transfected with 250 μg total DNA with PEI at the same ratio as that used for smaller production. Cell culture supernatants were harvested 5 days post-transfection and incubated overnight at 4 °C with Pierce™ Protein A/G Plus Agarose. Agarose resin was collected by pouring the supernatant over a BioRad Econo-Column Chromatography Column. Resin was washed with PBS before antibody was eluted with Pierce™ IgG Elution Buffer directly into Tris-HCl pH 8.0 at a ratio of 1:10. Antibody eluates were concentrated using Amicon Ultra-15 Centrifugal Filter Units with a 50 kDa molecular weight cut off to a final volume of approximately 1mL.

Neutralizing activity.

Three-fold dilutions of antibodies (3 μg/ml – 0.0048 μg/ml) were assayed in technical duplicates and incubated with 1×104 RLU/well of Ebola GP-pseudotyped vesicular stomatitis virus expressing luciferase (IBT Bioservices) for 1 hour at room temperature before addition to VeroE6 monolayers (6×105 cells/well) and incubation at 37 °C for 14–16h. Cells were lysed using Passive Lysis Buffer (Promega) and luciferase activity was determined using a luciferase activating reagent (Promega). IC50 titers were determined using Prism 8.0.

Antigen-binding ELISA.

Recombinant Ebola GP antigen (IBT Bioservices) was coated onto MaxiSorp 384-well plates (Nunc) at 62.5 ng/well at 4 °C overnight. Wells were washed with PBS and blocked with 5% BSA prior to addition of 2-fold dilutions of antibodies (10–0.08 μg/ml) for 2 h at room temperature. Wells were washed 6 times with PBS + 0.01% Tween 20 and incubated for 1 h with an HRP-conjugated anti-human IgG Fc antibody (1:5000; Jackson ImmunoResearch). Wells were again washed 6 times with PBS + 0.01% Tween 20, incubated with 1X TMB substrate for 5 minutes and the reaction was stopped with 1N H2SO4 before absorbance at 405 nm was measured. The area under the curve was determined using Prism 8.0.

Antigen affinity measurement by biolayer inferometry.

0.3 μg/ml of each mAb was immobilized to anti-human IgG Fc (AHC) kinetic biosensors (Sartorius/ForteBio) in 1X kinetics buffer for 180s followed by association and dissociation measurements to two-fold dilutions of recombinant Ebola GP starting at 500nM on an Octet Red96 instrument (Forte Bio). Association and dissociation were measured for 120s and 600s, respectively. Following subtraction of the reference control, the KD was calculated using a 1:1 binding model in the Octet Data Analysis software version 8.1.

Antibody-dependent NK cell degranulation.

Recombinant Ebola GP antigen (IBT Bioservices) was coated onto MaxiSorp 96-well plates (Nunc) at 300 ng/well at 4 °C overnight. Wells were washed with PBS and blocked with 5% BSA prior to addition of antibodies (5 μg/ml) and incubation for 2 h at 37 °C. Unbound antibodies were removed by washing with PBS, and NK cells enriched from the peripheral blood of human donors were added at 5×104 cells/well in the presence of 4 μg/ml brefeldin A (Sigma-Aldrich), 5 μg/ml GolgiStop (Life Technologies) and anti-CD107a antibody (Clone H4A3, BD Biosciences) for 5 h. Cells were fixed and permeabilized with Fix/Perm (Life Technologies) according to the manufacturer’s instructions to stain for intracellular IFNγ (Clone B27, BD Biosciences), and MIP-1β (Clone D21–1351, BD Biosciences). Cells were analyzed on a BD LSRII flow cytometer. Gating strategy and representative flow cytometry plots are shown in Figure S4.

Antibody-dependent neutrophil phagocytosis (ADNP).

Biotinylated Ebola GP was coupled to yellow-green Neutravidin beads (Life Technologies). Antibodies were diluted in culture medium to 5 μg/ml and incubated with GP-coated beads for 2 h at 37 °C. Freshly isolated white blood cells from human donor peripheral blood (5×104 cells/well) were incubated for 1h at 37 °C. Cells were then stained for CD66b (Clone G10F5; BioLegend), CD3 (Clone UCHT1; BD Biosciences), and CD14 (Clone MP9; BD Biosciences), fixed with 4% paraformaldehyde, and analyzed by flow cytometry. Neutrophils were defined as SSC-Ahigh CD66b+, CD3, CD14. A phagocytic score was determined using the following formula: (percentage of FITC+ cells)*(geometric mean fluorescent intensity (gMFI) of the FITC+ cells)/10,000. Gating strategy and representative flow cytometry plots are shown in Figure S4.

Antibody-dependent cellular phagocytosis by human monocytes (ADCP).

GP-coated beads were generated as described for ADNP. Antibodies were diluted in culture medium to 5μg/ml and incubated with GP-coated beads for 2 h at 37 °C. Unbound antibodies were removed by centrifugation prior to the addition of THP-1 cells at 2.5×104 cells/well. Cells were fixed with 4% paraformaldehyde and analyzed by flow cytometry. A phagocytic score was determined as described above. Gating strategy and representative flow cytometry plots are shown in Figure S4.

Antibody-mediated complement deposition (ADCD).

GP-coated beads were generated as described for ADNP, but with substitution of red-fluorescent Neutravidin beads (Life Technologies). Antibodies were diluted in culture medium to 5μg/ml and incubated with GP-coated beads for 2 h at 37 °C. Unbound antibodies were removed by centrifugation prior to the addition of reconstituted guinea pig complement (Cedarlane Labs) diluted in veronal buffer supplemented with calcium and magnesium (Boston Bioproducts) for 20 min at 37 °C. Beads were washed with PBS containing 15 mM EDTA, and stained with an FITC-conjugated anti-guinea pig C3 antibody (MP Biomedicals). C3 deposition onto beads was analyzed by flow cytometry. The gMFI of FITC of all beads was measured. Gating strategy and representative flow cytometry plots are shown in Figure S4.

Measurement of FcR binding kinetics by SPR.

Each mAb (12.5 μg/ml) was printed onto a MX96 200M chip (Xantec Bioanalytics) using a continuous flow microspotter (CFM) (Wasatch Microfluidics) with sulfo-NHS and EDC coupling chemistry. Measurement of FcγR binding kinetics was performed using an image-based array reader (MX96, IBIS Technologies). The chip was quenched with 1 M ethanolamine and 3-fold dilutions of recombinant FcγR (Duke University; 10 μM to 0.005 μM) were sequentially injected. To account for non-specific signals, the signals for each antibody were double-referenced using signals from both blank injections and from uncoupled inter-spots between antibodies. Data was processed using Sprint software (IBIS Technologies), and the kinetics of binding (KD) was determined using Kinetics software (Carterra).

Measurement of FcR binding kinetics by biolayer inferometry.

100nM of each mAb was immobilized to anti-human IgG Fab-CH1 kinetic biosensors (Sartorius/ForteBio) in 1X kinetics buffer for 300s followed by association and dissociation measurements to two-fold dilutions of recombinant murine FcRs starting at 200nM (mFcγRI and mFcγRIV) or 600nM (mFcγRIIb and mFcγRIII) on an Octet Red96 instrument (Forte Bio). Association and dissociation were measured for 120s and 600s, respectively. Following subtraction of the reference control, the KD was calculated using a 2:1 analyte binding model in the Octet Data Analysis software version 8.1.

Measurement of murine FcγR binding to immune complexes.

Recombinant EBOV GP (IBT Bioservices) were coupled to MagPlex beads (Luminex) via sulfo-NHS coupling chemistry. Murine FcγR receptors (FcγRIII, FcγRIV, FcγRI) and one inhibitory receptor (FcγR2B). Serial dilutions of the indicated antibodies samples were diluted in 1X PBS + 0.1% bovine serum albumin (BSA) + 0.05% Tween20 and incubated with antigen-coupled beads for 2 hours. Beads were washed and incubated with recombinant biotinylated Fc-receptors that were tetramerized via Streptavidin-PE for 1 hour at room temperature. Beads were washed analyzed on a Sartorius iQue screener. The median fluorescent intensity of 30 beads/region was recorded. A human IgG1 isotype control was used to establish antigen-specificity and background binding.

In vivo protection.

The animal protocol for testing of mAbs in mice was approved by the Institutional Animal Care and Use Committee of the UTMB. Seven-week-old BALB/c mice (Charles River Laboratories) were placed in the ABSL-4 facility at the Galveston National Laboratory. Groups of five animals were injected intraperitoneally with 1,000 PFU of mouse-adapted EBOV, strain Mayinga (Bray et al., 1998). The animals were injected with mAbs by the intraperitoneal route using 0.1 mg or 0.03 mg per treatment 24 h after virus challenge. Animals treated with PBS served as controls. Animal observation procedures were performed as described elsewhere (Ilinykh et al., 2018). The overall observation period was 28 days.

QUANTIFICATION AND STATISTICAL ANALYSIS

Correlation analysis.

Univariate spearman correlation r values were calculated using GraphPad Prism, and the p values were corrected for multiple comparisons using Benjamini-Hochberg correction.

K-means clustering analysis.

K-means clustering analysis was performed in JMP Pro 15, and elbow joint analysis was used to determine the optimal number of clusters in R.

Hierarchical clustering analysis.

Unsupervised hierarchical clustering analysis was performed in JMP Pro 15, using the Ward method.

Supplementary Material

1

KEY RESOURCES TABLE.

REAGENT or RESOURCE SOURCE IDENTIFIER
Antibodies
KZ52 IBT Bioservices Cat #: 0260-001
VIC16 Takada et al., 2003 n/a
Mouse anti-human CD66b (clone G10F5), Pacific Blue BioLegend RRID:AB_2563294
Mouse anti-human CD3 (clone UCHT1), Alexa Fluor 700 BD Biosciences RRID:AB_396952
Mouse anti-human CD107a (clone H4A3), PE-Cy5 BD Biosciences RRID:AB_396136
Mouse anti-human IFNgamma (clone B27), FITC BD Biosciences RRID: AB_398580
Mouse anti-human MIP-1beta (clone D21-1351), PE BD Biosciences RRID: AB_393549
Mouse anti-human CD56 (clone B159), PE-Cy7 BD Biosciences RRID:AB_396853
Mouse anti-human CD14 (clone MφP9), APC-Cy7 BD Biosciences RRID:AB_396889
Mouse anti-human CD16 (clone 3G8), APC-Cy7 BD Biosciences RRID:AB_396864
Anti-guinea pig complement C3 goat IgG fraction, FITC MP Biomedicals Cat # 0855385
anti-human IgG Fc antibody Jackson ImmunoResearch Cat # 109-035-190
Mouse Anti-Human IgG1-Fc PE Southern Biotech Cat # 9054-09
Mouse Anti-Human IgG2-Fc PE Southern Biotech Cat # 9060-09
Mouse Anti-Human IgG3-Hinge PE Southern Biotech Cat # 9210-09
Mouse Anti-Human IgG4-Fc PE Southern Biotech Cat # 9200-09
Mouse Anti-Human IgA1-Fc PE Southern Biotech Cat # 9130-09
Mouse Anti-Human IgM-Fc PE Southern Biotech Cat # 9020-09
VIC16 REFORMabs This manuscript n/a
Bacterial and Virus Strains
Mouse-adapted EBOV/Mayinga (EBOV/M.mus-tc/COD/76/Yambuku-Mayinga Bray et al., 1998 n/a
Stellar competent cells Takara Cat # 636766
Biological Samples
Recombinant Vesicular Stomatitis Virus pseudotyped Ebola glycoprotein (rVSV pseudotyped EbOV gP) IBT Bioservices Cat # 1001-001
LowTox Guinea Pig Complement CedarLane Labs Cat # CL4051
Chemicals, Peptides, and Recombinant Proteins
Brefeldin A Sigma Aldrich Cat # B7651
GolgiStop BD Biosciences Cat # 554724
EBOV GPΔTM IBT Bioservices Cat # 0501-015
Polyethylenimine Polysciences Cat # 23966-1
Recombinant FcγRs (mouse and human) Duke University Human Vaccine Institute n/a
Recombinant murine FcγRI R&D Systems Cat # 2074-FC-050
Streptavidin-R-Phycoerythrin Prozyme Cat # PJ31S
Critical Commercial Assays
RosetteSep NK cell enrichment kit Stem Cell Technologies Cat # 15025
Luciferase Assay System Promega Cat # E1500
Deposited Data
The datasets generated from this study are available through Mendeley. doi: 10.17632/9b2h8p9gtc.1
Experimental Models: Cell Lines
THP-1 monocytes ATCC RRID: CVCL_0006
FreeStyle 293F cells ThermoFisher Cat # R79007
VeroE6 ATCC RRID: CVCL_0574
Experimental Models: Organisms/Strains
Mouse: Female BALB/c Charles River; The Jackson Laboratories Strain Code 028 (CR); RRID:IMSR_JAX:000651
Recombinant DNA
pUC19 vector New England Biolabs Cat # N3041S
Software and Algorithms
GraphPad Prism 8 GraphPad Software, Inc. RRID:SCR_002798
R Studio R Project for Statistical Computing RRID:SCR_000432
Octet Data Analysis software version 8.1 Forte Bio/ Sartorius https://www.sartorius.com/en/products/protein-analysis/octet-systems-software
Sprint Software IBIS Technologies https://www.ibis-spr.nl/product/ibis-triangle-software/
Flow Jo BD Bioscience RRID:SCR_008520
JMP Pro 15 JMP RRID:SCR_014242
Kinetics Data Analysis software Carterra https://carterra-bio.com/applications/kinetics-software/
Other
anti-human IgG Fc (AHC) kinetic biosensors Sartorius 18-5060
FluoSpheres® NeutrAvidin®-Labeled Microspheres, 1.0 μm, yellow-green fluorescent (505/515), 1% solids Life Technologies Cat #: F-8776
FluoSpheres® NeutrAvidin®-Labeled Microspheres, 1.0 μm, red fluorescent (580/605), 1% solids Life Technologies Cat #: F8775
PureProteome Protein G magnetic beads Millipore Cat # LSKMAGG10
MagPlex microspheres Luminex corporation Cat # MC12001-01, MCI12040-01, MCI10077-01
  • Ebola virus disease survivors develop diverse profiles of antibody responses.

  • Fc-engineering of monoclonal antibodies can replicate human functional profiles.

  • A Fc-variant panel was generated using an anti-Ebola virus neutralizing Fab domain.

  • Fc-variants with high complement yet moderate ADCC activity were protective in vivo.

Acknowledgements.

We would like to thank all members of the Alter laboratory, the Viral Hemorrhagic Fever Immunotherapeutics Consortium, and the Consortium for Viral Systems Biology for critical discussion and advice, Madeline Davis for administrative help, and Christina Karsten and Julie Boucau for help with the FPLC analysis. This work was supported by U19 AI109762, AI142790, U19 AI135995, and partly by the Japan Agency for Medical Research and Development (JP17fk0108101). The graphical abstract was created using BioRender.

Footnotes

Declaration of Interests.

G.A. is a founder of Seromyx Systems Inc. and T.J.S. is currently an employee of Seromyx Systems Inc.

Inclusion and Diversity.

We worked to ensure gender balance in the recruitment of human subjects. One or more of the authors of this paper self-identifies as an underrepresented ethnic minority in science. One or more of the authors of this paper self-identifies as a member of the LGBTQ+ community. The author list of this paper includes contributors from the location where the research was conducted who participated in the data collection, design, analysis, and/or interpretation of the work.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Ackerman ME, Das J, Pittala S, Broge T, Linde C, Suscovich TJ, Brown EP, Bradley T, Natarajan H, Lin S, et al. (2018). Route of immunization defines multiple mechanisms of vaccine-mediated protection against SlV. Nature medicine. [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Agnihothram S, Gopal R, Yount BL Jr, Donaldson EF, Menachery VD, Graham RL, Scobey TD, Gralinski LE, Denison MR, Zambon M, et al. (2013). Evaluation of Serologic and Antigenic Relationships Between Middle Eastern Respiratory Syndrome Coronavirus and Other Coronaviruses to Develop Vaccine Platforms for the Rapid Response to Emerging Coronaviruses. The Journal of infectious diseases 209, 995–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Baseler L, Chertow DS, Johnson KM, Feldmann H, and Morens DM (2017). The Pathogenesis of Ebola Virus Disease. Annual review of pathology 12, 387–418. [DOI] [PubMed] [Google Scholar]
  4. Bornholdt ZA, Herbert AS, Mire CE, He S, Cross RW, Wec AZ, Abelson DM, Geisbert JB, James RM, Rahim MN, et al. (2019). A Two-Antibody Pan-Ebolavirus Cocktail Confers Broad Therapeutic Protection in Ferrets and Nonhuman Primates. Cell host & microbe 25, 49–58 e45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Bornholdt ZA, Turner HL, Murin CD, Li W, Sok D, Souders CA, Piper AE, Goff A, Shamblin JD, Wollen SE, et al. (2016). Isolation of potent neutralizing antibodies from a survivor of the 2014 Ebola virus outbreak. Science 351, 1078–1083. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Bournazos S, DiLillo DJ, Goff AJ, Glass PJ, and Ravetch JV (2019). Differential requirements for FcgammaR engagement by protective antibodies against Ebola virus. Proceedings of the National Academy of Sciences of the United States of America 116, 20054–20062. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Bray M, Davis K, Geisbert T, Schmaljohn C, and Huggins J (1998). A mouse model for evaluation of prophylaxis and therapy of Ebola hemorrhagic fever. The Journal of infectious diseases 178, 651–661. [DOI] [PubMed] [Google Scholar]
  8. Bruhns P (2012). Properties of mouse and human IgG receptors and their contribution to disease models. Blood 119, 5640–5649. [DOI] [PubMed] [Google Scholar]
  9. Chu SY, Vostiar I, Karki S, Moore GL, Lazar GA, Pong E, Joyce PF, Szymkowski DE, and Desjarlais JR (2008). Inhibition of B cell receptor-mediated activation of primary human B cells by coengagement of CD19 and FcgammaRIIb with Fc-engineered antibodies. Molecular immunology 45, 3926–3933. [DOI] [PubMed] [Google Scholar]
  10. Corti D, Misasi J, Mulangu S, Stanley DA, Kanekiyo M, Wollen S, Ploquin A, Doria-Rose NA, Staupe RP, Bailey M, et al. (2016). Protective monotherapy against lethal Ebola virus infection by a potently neutralizing antibody. Science 351, 1339–1342. [DOI] [PubMed] [Google Scholar]
  11. Corti D, Zhao J, Pedotti M, Simonelli L, Agnihothram S, Fett C, Fernandez-Rodriguez B, Foglierini M, Agatic G, Vanzetta F, et al. (2015). Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus. Proceedings of the National Academy of Sciences of the United States of America 112, 10473–10478. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Dall’Acqua WF, Woods RM, Ward ES, Palaszynski SR, Patel NK, Brewah YA, Wu H, Kiener PA, and Langermann S (2002). Increasing the affinity of a human IgG1 for the neonatal Fc receptor: biological consequences. Journal of immunology 169, 5171–5180. [DOI] [PubMed] [Google Scholar]
  13. Datta-Mannan A, Witcher DR, Tang Y, Watkins J, Jiang W, and Wroblewski VJ (2007). Humanized IgG1 variants with differential binding properties to the neonatal Fc receptor: relationship to pharmacokinetics in mice and primates. Drug Metab Dispos 35, 86–94. [DOI] [PubMed] [Google Scholar]
  14. Dejnirattisai W, Jumnainsong A, Onsirisakul N, Fitton P, Vasanawathana S, Limpitikul W, Puttikhunt C, Edwards C, Duangchinda T, Supasa S, et al. (2010). Cross-reacting antibodies enhance dengue virus infection in humans. Science 328, 745–748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Dekkers G, Bentlage AEH, Stegmann TC, Howie HL, Lissenberg-Thunnissen S, Zimring J, Rispens T, and Vidarsson G (2017). Affinity of human IgG subclasses to mouse Fc gamma receptors. mAbs 9, 767–773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Deming D, Sheahan T, Heise M, Yount B, Davis N,Sims A, Suthar M, Harkema J, Whitmore A, Pickles R, et al. (2006). Vaccine efficacy in senescent mice challenged with recombinant SARS-CoV bearing epidemic and zoonotic spike variants. PLoS medicine 3, e525. [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Diebolder CA, Beurskens FJ,de Jong RN, Koning RI, Strumane K,Lindorfer MA, Voorhorst M, Ugurlar D, Rosati S,Heck AJ, et al. (2014). Complement is activated by IgG hexamers assembled at the cell surface. Science 343, 1260–1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. DiLillo DJ, Tan GS, Palese P, and Ravetch JV (2014). Broadly neutralizing hemagglutinin stalk-specific antibodies require FcgammaR interactions for protection against influenza virus in vivo. Nature medicine 20, 143–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Engler C, Kandzia R, and Marillonnet S (2008). A one pot, one step, precision cloning method with high throughput capability. PLoS One 3, e3647. [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Fischinger S, Fallon JK, Michell AR, Broge T, Suscovich TJ, Streeck H, and Alter G (2019). A high-throughput, bead-based, antigen-specific assay to assess the ability of antibodies to induce complement activation. Journal of immunological methods 473, 112630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Flyak AI, Shen X, Murin CD, Turner HL, David JA, Fusco ML, Lampley R, Kose N, Ilinykh PA, Kuzmina N, et al. (2016). Cross-Reactive and Potent Neutralizing Antibody Responses in Human Survivors of Natural Ebolavirus Infection. Cell. [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Gilchuk P, Kuzmina N, Ilinykh PA, Huang K, Gunn BM, Bryan A, Davidson E, Doranz BJ, Turner HL, Fusco ML, et al. (2018). Multifunctional Pan-ebolavirus Antibody Recognizes a Site of Broad Vulnerability on the Ebolavirus Glycoprotein. Immunity 49, 363–374 e310. [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Gunn BM, Roy V, Karim MM, Hartnett JN, Suscovich TJ, Goba A, Momoh M, Sandi JD, Kanneh L, Andersen KG, et al. (2020). Survivors of Ebola Virus Disease Develop Polyfunctional Antibody Responses. The Journal of infectious diseases 221, 156–161. [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Gunn BM, Yu WH, Karim MM, Brannan JM, Herbert AS, Wec AZ, Halfmann PJ, Fusco ML, Schendel SL, Gangavarapu K, et al. (2018). A Role for Fc Function in Therapeutic Monoclonal Antibody-Mediated Protection against Ebola Virus. Cell host & microbe 24, 221–233 e225. [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. He W, Tan GS, Mullarkey CE, Lee AJ, Lam MM, Krammer F, Henry C, Wilson PC, Ashkar AA, Palese P, et al. (2016). Epitope specificity plays a critical role in regulating antibody-dependent cell-mediated cytotoxicity against influenza A virus. Proceedings of the National Academy of Sciences of the United States of America 113, 11931–11936. [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Hessell AJ, Hangartner L, Hunter M, Havenith CE, Beurskens FJ, Bakker JM, Lanigan CM, Landucci G, Forthal DN, Parren PW, et al. (2007). Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449, 101–104. [DOI] [PubMed] [Google Scholar]
  27. Hinton PR, Johlfs MG, Xiong JM, Hanestad K, Ong KC, Bullock C, Keller S, Tang MT, Tso JY, Vasquez M, et al. (2004). Engineered human IgG antibodies with longer serum half-lives in primates. The Journal of biological chemistry 279, 6213–6216. [DOI] [PubMed] [Google Scholar]
  28. Idusogie EE, Presta LG, Gazzano-Santoro H, Totpal K, Wong PY, Ultsch M, Meng YG, and Mulkerrin MG (2000). Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. Journal of immunology 164, 4178–4184. [DOI] [PubMed] [Google Scholar]
  29. Idusogie EE, Wong PY, Presta LG, Gazzano-Santoro H, Totpal K, Ultsch M, and Mulkerrin MG (2001). Engineered antibodies with increased activity to recruit complement. Journal of immunology 166, 2571–2575. [DOI] [PubMed] [Google Scholar]
  30. Ilinykh PA, Graber J, Kuzmina NA, Huang K, Ksiazek TG, Crowe JE Jr., and Bukreyev A (2018). Ebolavirus Chimerization for the Development of a Mouse Model for Screening of Bundibugyo-Specific Antibodies. J Infect Dis 218, S418–S422. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Ilinykh PA, Huang K, Santos RI, Gilchuk P, Gunn BM, Karim MM, Liang J, Fouch ME, Davidson E, Parekh DV, et al. (2020). Non-neutralizing Antibodies from a Marburg Infection Survivor Mediate Protection by Fc-Effector Functions and by Enhancing Efficacy of Other Antibodies. Cell host & microbe 27, 976–991 e911. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Jaume M, Yip MS, Cheung CY, Leung HL, Li PH, Kien F, Dutry I, Callendret B, Escriou N, Altmeyer R, et al. (2011). Anti-Severe Acute Respiratory Syndrome Coronavirus Spike Antibodies Trigger Infection of Human Immune Cells via a pH- and Cysteine Protease-Independent FcγR Pathway. Journal of virology 85, 10582–10597. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Ju B, Zhang Q, Ge X, Wang R, Yu J, Shan S, Zhou B, Song S, Tang X, Yu J, et al. (2020). Potent human neutralizing antibodies elicited by SARS-CoV-2 infection. bioRxiv. [DOI] [PubMed] [Google Scholar]
  34. Kunert R, Wolbank S, Stiegler G, Weik R, and Katinger H (2004). Characterization of molecular features, antigen-binding, and in vitro properties of IgG and IgM variants of 4E10, an anti-HIV type 1 neutralizing monoclonal antibody. AIDS research and human retroviruses 20, 755–762. [DOI] [PubMed] [Google Scholar]
  35. Lazar GA, Dang W, Karki S, Vafa O, Peng JS, Hyun L, Chan C, Chung HS, Eivazi A, Yoder SC, et al. (2006). Engineered antibody Fc variants with enhanced effector function. Proceedings of the National Academy of Sciences of the United States of America 103, 4005–4010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Liu L, Wei Q, Lin Q, Fang J, Wang H, Kwok H, Tang H, Nishiura K, Peng J, Tan Z, et al. (2019). Anti-spike IgG causes severe acute lung injury by skewing macrophage responses during acute SARS-CoV infection. JCI Insight 4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Maruyama T, Rodriguez LL, Jahrling PB, Sanchez A, Khan AS, Nichol ST, Peters CJ, Parren PW, and Burton DR (1999). Ebola virus can be effectively neutralized by antibody produced in natural human infection. Journal of virology 73, 6024–6030. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Misasi J, Gilman MS, Kanekiyo M, Gui M, Cagigi A, Mulangu S, Corti D, Ledgerwood JE, Lanzavecchia A, Cunningham J, et al. (2016). Structural and molecular basis for Ebola virus neutralization by protective human antibodies. Science 351, 1343–1346. [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Moldt B, Schultz N, Dunlop DC, Alpert MD, Harvey JD, Evans DT, Poignard P, Hessell AJ, and Burton DR (2011). A panel of IgG1 b12 variants with selectively diminished or enhanced affinity for Fcgamma receptors to define the role of effector functions in protection against HIV. J Virol 85, 10572–10581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Moore GL, Chen H, Karki S, and Lazar GA (2010). Engineered Fc variant antibodies with enhanced ability to recruit complement and mediate effector functions. mAbs 2, 181–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Nimmerjahn F, Bruhns P, Horiuchi K, and Ravetch JV (2005). FcgammaRIV: a novel FcR with distinct IgG subclass specificity. Immunity 23, 41–51. [DOI] [PubMed] [Google Scholar]
  42. Nonaka M, and Kimura A (2006). Genomic view of the evolution of the complement system. Immunogenetics 58, 701–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Olinger GG Jr., Pettitt J, Kim D, Working C, Bohorov O, Bratcher B, Hiatt E, Hume SD, Johnson AK, Morton J, et al. (2012). Delayed treatment of Ebola virus infection with plant-derived monoclonal antibodies provides protection in rhesus macaques. Proceedings of the National Academy of Sciences of the United States of America 109, 18030–18035. [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Overdijk MB, Verploegen S, Ortiz Buijsse A, Vink T, Leusen JH, Bleeker WK, and Parren PW (2012). Crosstalk between human IgG isotypes and murine effector cells. Journal of immunology 189, 3430–3438. [DOI] [PubMed] [Google Scholar]
  45. Parren PW, Geisbert TW, Maruyama T, Jahrling PB, and Burton DR (2002). Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. Journal of virology 76, 6408–6412. [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Pinto D, Park Y-J, Beltramello M, Walls AC, Tortorici MA, Bianchi S, Jaconi S, Culap K, Zatta F, De Marco A, et al. (2020). Structural and functional analysis of a potent sarbecovirus neutralizing antibody. bioRxiv. [Google Scholar]
  47. Polack FP, Teng MN, Collins PL, Prince GA, Exner M, Regele H, Lirman DD, Rabold R, Hoffman SJ, Karp CL, et al. (2002). A role for immune complexes in enhanced respiratory syncytial virus disease. The Journal of experimental medicine 196, 859–865. [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Qiu X, Wong G, Audet J, Bello A, Fernando L, Alimonti JB, Fausther-Bovendo H, Wei H, Aviles J, Hiatt E, et al. (2014). Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp. Nature 514, 47–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Richards JO, Karki S, Lazar GA, Chen H, Dang W, and Desjarlais JR (2008). Optimization of antibody binding to FcgammaRIIa enhances macrophage phagocytosis of tumor cells. Mol Cancer Ther 7, 2517–2527. [DOI] [PubMed] [Google Scholar]
  50. Saphire EO, Schendel SL, Fusco ML, Gangavarapu K, Gunn BM, Wec AZ, Halfmann PJ, Brannan JM, Herbert AS, Qiu X, et al. (2018a). Systematic Analysis of Monoclonal Antibodies against Ebola Virus GP Defines Features that Contribute to Protection. Cell 174, 938–952 e913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Saphire EO, Schendel SL, Gunn BM, Milligan JC, and Alter G (2018b). Antibody-mediated protection against Ebola virus. Nature immunology 19, 1169–1178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Shields RL, Namenuk AK, Hong K, Meng YG, Rae J, Briggs J, Xie D, Lai J, Stadlen A, Li B, et al. (2001). High resolution mapping of the binding site on human IgG1 for Fc gamma RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the Fc gamma R. The Journal of biological chemistry 276, 6591–6604. [DOI] [PubMed] [Google Scholar]
  53. Smith P, DiLillo DJ, Bournazos S, Li F, and Ravetch JV (2012). Mouse model recapitulating human Fcgamma receptor structural and functional diversity. Proceedings of the National Academy of Sciences of the United States of America 109, 6181–6186. [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Stavenhagen JB, Gorlatov S, Tuaillon N, Rankin CT, Li H, Burke S, Huang L, Vijh S, Johnson S, Bonvini E, et al. (2007). Fc optimization of therapeutic antibodies enhances their ability to kill tumor cells in vitro and controls tumor expansion in vivo via low-affinity activating Fcgamma receptors. Cancer research 67, 8882–8890. [DOI] [PubMed] [Google Scholar]
  55. Steurer W, Nickerson PW, Steele AW, Steiger J, Zheng XX, and Strom TB (1995). Ex vivo coating of islet cell allografts with murine CTLA4/Fc promotes graft tolerance. Journal of immunology 155, 1165–1174. [PubMed] [Google Scholar]
  56. Sun P, Williams M, Nagabhushana N, Jani V, Defang G, and Morrison BJ (2019). NK Cells Activated through Antibody-Dependent Cell Cytotoxicity and Armed with Degranulation/IFN-gamma Production Suppress Antibody-dependent Enhancement of Dengue Viral Infection. Scientific reports 9, 1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. van Erp EA, Luytjes W, Ferwerda G, and van Kasteren PB (2019). Fc-Mediated Antibody Effector Functions During Respiratory Syncytial Virus Infection and Disease. Frontiers in immunology 10, 548. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Walker LM, and Burton DR (2018). Passive immunotherapy of viral infections: ‘super-antibodies’ enter the fray. Nature reviews Immunology 18, 297–308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Wan Y, Shang J, Sun S, Tai W, Chen J, Geng Q, He L, Chen Y, Wu J, Shi Z, et al. (2020). Molecular Mechanism for Antibody-Dependent Enhancement of Coronavirus Entry. Journal of virology 94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  60. Wang SF, Tseng SP, Yen CH, Yang JY, Tsao CH, Shen CW, Chen KH, Liu FT, Liu WT, Chen YM, et al. (2014). Antibody-dependent SARS coronavirus infection is mediated by antibodies against spike proteins. Biochemical and biophysical research communications 451, 208–214. [DOI] [PMC free article] [PubMed] [Google Scholar]
  61. Wang TT, Sewatanon J, Memoli MJ, Wrammert J, Bournazos S, Bhaumik SK, Pinsky BA, Chokephaibulkit K, Onlamoon N, Pattanapanyasat K, et al. (2017). IgG antibodies to dengue enhanced for FcgammaRIIIA binding determine disease severity. Science 355, 395–398. [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Warfield KL, Perkins JG, Swenson DL, Deal EM, Bosio CM, Aman MJ, Yokoyama WM, Young HA, and Bavari S (2004). Role of natural killer cells in innate protection against lethal ebola virus infection. The Journal of experimental medicine 200, 169–179. [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Wec AZ, Herbert AS, Murin CD, Nyakatura EK, Abelson DM, Fels JM, He S, James RM, de La Vega MA, Zhu W, et al. (2017). Antibodies from a Human Survivor Define Sites of Vulnerability for Broad Protection against Ebolaviruses. Cell 169, 878–890 e815. [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Weiner LM, Dhodapkar MV, and Ferrone S (2009). Monoclonal antibodies for cancer immunotherapy. Lancet 373, 1033–1040. [DOI] [PMC free article] [PubMed] [Google Scholar]
  65. Xu D, Alegre ML, Varga SS, Rothermel AL, Collins AM, Pulito VL, Hanna LS, Dolan KP, Parren PW, Bluestone JA, et al. (2000). In vitro characterization of five humanized OKT3 effector function variant antibodies. Cell Immunol 200, 16–26. [DOI] [PubMed] [Google Scholar]
  66. Yeung YA, Leabman MK, Marvin JS, Qiu J, Adams CW, Lien S, Starovasnik MA, and Lowman HB (2009). Engineering human IgG1 affinity to human neonatal Fc receptor: impact of affinity improvement on pharmacokinetics in primates. Journal of immunology 182, 7663–7671. [DOI] [PubMed] [Google Scholar]
  67. Zalevsky J, Chamberlain AK, Horton HM, Karki S, Leung IW, Sproule TJ, Lazar GA, Roopenian DC, and Desjarlais JR (2010). Enhanced antibody half-life improves in vivo activity. Nat Biotechnol 28, 157–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Zeitlin L, Pettitt J, Scully C, Bohorova N, Kim D, Pauly M, Hiatt A, Ngo L, Steinkellner H, Whaley KJ, et al. (2011). Enhanced potency of a fucose-free monoclonal antibody being developed as an Ebola virus immunoprotectant. Proceedings of the National Academy of Sciences of the United States of America 108, 20690–20694. [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Zhao X, Howell KA, He S, Brannan JM, Wec AZ, Davidson E, Turner HL, Chiang CI, Lei L, Fels JM, et al. (2017). Immunization-Elicited Broadly Protective Antibody Reveals Ebolavirus Fusion Loop as a Site of Vulnerability. Cell 169, 891–904 e815. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1

Data Availability Statement

The datasets generated in this study are available on Mendeley (doi: 10.17632/9b2h8p9gtc.1)

RESOURCES