Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2019 Nov 1.
Published in final edited form as: Nat Immunol. 2018 Oct 17;19(11):1169–1178. doi: 10.1038/s41590-018-0233-9

Antibody-mediated protection against Ebola virus

Erica Ollmann Saphire 1,2,*, Sharon L Schendel 1, Bronwyn M Gunn 3, Jacob C Milligan 1, Galit Alter 3,*
PMCID: PMC6814399  NIHMSID: NIHMS1055273  PMID: 30333617

Abstract

Recent Ebola virus disease epidemics have highlighted the need for effective vaccines and therapeutics to prevent future outbreaks. Antibodies are clearly critical for control of this deadly disease; however, the specific mechanisms of action of protective antibodies have yet to be defined. In this Perspective we discuss the antibody features that correlate with in vivo protection during infection with Ebola virus, based on the results of a systematic and comprehensive study of antibodies directed against this virus. Although neutralization activity mediated by the Fab domains of the antibody is strongly correlated with protection, recruitment of immune effector functions by the Fc domain has also emerged as a complementary, and sometimes alternative, route to protection. For a subset of antibodies, Fc-mediated clearance and killing of infected cells seems to be the main driver of protection after exposure and mirrors observations in vaccination studies. Continued analysis of antibodies that achieve protection partially or wholly through Fc-mediated functions, the precise functions required, the intersection with specificity and the importance of these functions in different animal models is needed to identify and begin to capitalize on Fc-mediated protection in vaccines and therapeutics alike.

Introduction

Monoclonal antibody (mAb) therapeutics represent one of the fastest growing classes of clinically licensed drugs due to their exquisite specificity and tunable precision. Although immunotherapies for oncology and autoimmune disorders are well-established1, there are fewer clinical applications of immunotherapy for infectious diseases2,3. However, due to their rapid discovery process, as well as their remarkable specificity and low toxicity, immunotherapeutics might be particularly valuable for emerging infectious diseases for which no other therapy is yet available47.

An antibody molecule links two hypervariable, antigen-binding domains (Fab fragments) via a flexible hinge to a constant region (Fc). After anchoring to a foreign antigen, Fab fragments can mechanically neutralize pathogens by physically inactivating the viral entry machinery through blocking receptor binding, preventing necessary cleavage events, or obstructing conformational changes that drive fusion of the pathogen and host membrane. Following Fab binding, the Fc domain links the target to the innate immune system to signal the presence of a threat and to mediate destruction of the foreign body or unhealthy cell. In the design of an immunotherapeutic agent, both ends of the antibody can be tailored to enhance antigen recognition by the Fab and to drive Fc-mediated immune responses in different directions, from dampened immunopathology (‘silent Fc’) to enhanced target clearance (‘active Fc’).

Characterization of Fab-mediated protection can be straightforward through the use measurements of binding stoichiometry and neutralization of authentic viruses or pseudoviruses. Structural analysis of the binding of Fab domains to an antigen can define specific contacts that contribute to antigen recognition and antibody function.

In contrast, measurement and visualization of Fc-mediated mechanisms is complex. Different functions can be orchestrated by Fc receptors (FcRs) expressed on different cells of the immune system, with each cell type able to deploy a unique set of anti-pathogen functions such as cellular cytotoxicity, phagocytosis, direct pathogen killing, and modulation/stimulation of innate and adaptive immunity, which can be further influenced by the tissues or compartments in which they are activated. Indeed, FcRs marshal an incredible variety of anti-pathogen responses810.

Further, these immune-stimulating, Fc–FcR interactions have lower affinity than that of the often nanomolar Fab-antigen binding events, or are more transient than the latter and instead rely on avidity to reach a threshold at which the identified target is marked for destruction. These avid interactions require that antibodies of different subclasses and isotypes compete for immune complex occupancy to collectively regulate binding and activation. Thus polyclonal responses, rather than monoclonal responses, act in synergy to drive interactions involving larger complexes of proteins and cells that result in tiered signaling events that are difficult to directly observe using structural and biophysical techniques, although evolving methods such as correlation light electron microscopy could provide additional visual insights in the future11,12. Despite the challenges that accompany study of immune effector function, several examples from oncology and infectious disease have highlighted the importance of Fc activity by showing that the ability of an antibody to affect disease in animal models is dependent on Fc-mediated functions1320.

For many infectious disease targets, however, Fc-mediated clearance is or has been overshadowed by Fab-mediated neutralization, which is often the primary means for selecting mAb candidates for immunotherapy. Thus, fewer non-neutralizing, cell-targeting mAbs have been described or evaluated in animal models. Conflicting results in studies concerning filoviruses, which include ebolaviruses and marburgviruses, led us to critically examine the array of other antibody features and variables that together with neutralization correlate with in vivo protection. Our recently completed several-year, multidisciplinary analysis of antibodies against Ebola virus glycoprotein (GP) generated results that highlight the importance of Fc-mediated effector functions in post-exposure protection from filovirus infection in animal models. Here, we first describe the historical context in which the study began and how the findings relate to other recent work.

A combination of protection-driving variables

One of the first well-characterized mAbs against Ebola virus was KZ52, a human survivor antibody directed against the Ebola virus glycoprotein (GP)21. KZ52, initially described in 1999, appeared to possess the required features to be a successful immunotherapeutic against Ebola virus, including neutralization of Ebola virus in cell culture and high-affinity recognition of the receptor-binding core of the Ebola virus GP22 (Fig. 1a), as well as protection of mice and guinea pigs following challenge with lethal doses of Ebola virus23. Nevertheless, passive delivery of KZ52 alone did not protect against Ebola virus infection of nonhuman primates (NHPs), and the animals succumbed to disease. This lack of protection was not due to mutagenic escape: viruses isolated at time of death or euthanasia were sensitive to KZ52 neutralization in vitro24. Further, treated animals retained 200–400 μg/ml KZ52 in sera at day 4 postinfection, and concentrations were estimated to be more than 100-fold higher than the IC90 at time of challenge and for the first few days after exposure.

Figure 1.

Figure 1.

Evaluation of early antibody treatments evaluated for their protection of NHPs. In Figures 1–2, all mAbs that bind the GP core remaining after cathepsin cleavage (termed GPcl) are colored orange or yellow. All head- and glycan cap-binding mAbs are colored blue or purple. (a) Crystal structure of Ebola virus GP (grey) in complex with KZ5222 (orange). KZ52 was produced in CHO cells for evaluation in rhesus macaques at 50 mg/kg one day prior and four days after viral challenge24. (b) Superimposed negative stain EM structures of antibodies contained in the ZMAb cocktail28 (2G4, orange; 4G7 yellow; 1H3 blue) in complex with Ebola virus GP (grey). ZMAb antibodies were produced in murine hybridoma culture for evaluation in cynomolgus macaques at 25 mg/kg on days 1, 4 and 7 (100% survival) or 2, 5, and 8 after challenge (50% survival)27. (c) Model of the MB-003 cocktail made from the negative stain EM structure of the 13C6-GP complex28 and the crystal structure of 13F6 (purple molecular surface) bound to with its mucin-like domain linear epitope123 (white ball-and-stick). MB-003 antibodies were produced in CHO or Nicotiniana cells and evaluated in rhesus macaques at 50 mg/kg (CHO cell-produced, 50% survival) or 16.7 mg/kg (Nicotiniana-produced, 100% survival) at 1 hour and 4 and 7 days after challenge31. Delivery of Nicotiniana-produced MB-003 at later time points (5, 7, and 10 days after challenge) resulted in 43% survival32. No high-resolution structural information yet exists for the mucin-like domain or MB-003 component 6D8. These have been modeled with grey circles and a purple Fab fragment, respectively. The relative positions of the two mucin-binding antibodies 13F6 and 6D8 are as yet unknown.

Optimism for the prospect of Ebola virus immunotherapy increased when a study showed that polyclonal IgG concentrated from sera of vaccinated and challenged non-human primates (NHPs) could protect naive NHPs from infection with Ebola virus or the related Marburg virus25. Subsequently, two separate groups demonstrated that combinations, or “cocktails”, of mAbs could similarly achieve primate protection. One protective cocktail, termed ZMAb26,27, contains three mAbs27: 1H3, which is poorly neutralizing and targets the glycan cap domain of the viral GP that is physically removed from the virion during cell entry, plus mAbs 2G4 and 4G7, which are both neutralizing. Interestingly, the epitopes of 2G4 and 4G7 overlap with that of KZ5228 on the GP molecule (Fig. 1b), and all three of these antibodies are susceptible to the same escape mutation- a change from Gln to Arg at residue 50829,30. The other cocktail, MB-00331,32 also contains three mAbs: 13C6 against the glycan cap28,33, and 13F6 and 6D8 against the mucin-like domain22,34,35 (Fig. 1c). Protection of NHPs observed with MB-003 was thus somewhat surprising, as none of the component antibodies were strong neutralizers and all three antibodies targeted epitopes located on the upper and outer heavily glycosylated regions of GP that are cleaved from the virion during entry36,37. The physical removal of these epitopes from virions prior to receptor binding likely explains their poor to no neutralization in cell culture. Their success in vivo, however, suggested that their protective capacity could arise instead through Fc-mediated effector functions that tag GP-expressing virions and infected cells for destruction. Notably, whereas MB-003 comprising mAbs expressed in Chinese hamster ovary (CHO) cells offered poor protection to NHPs, MB-003 comprising mAbs produced in a plant-based system involving transgenic Nicotiana benthamiana resulted in greater survival of NHPs following Ebola virus infection. MB-003 expressed in these plant cells could also be used at a three-fold lower dose31,38. Transgenic N. benthamiana attach a truncated mammalian glycan of afucosylated and agalactosylated structure39, which enhances cell targeting functions3842,4346. Afucosylated antibodies, in particular, have 50-fold higher affinity for both FcγRIIIa and FcγRIIIb and increased antibody-dependent cell-mediated cytotoxicity (ADCC) activity43,44,4749. Together, these early findings suggested that neutralization is not the sole predictor of in vivo success, and that multiple mechanisms, including cell targeting by Fc, contribute to Ebola virus protection.

In 2014, the six mAbs from the ZMAb and MB-003 cocktails were re-mixed to produce ZMapp, a cocktail that reverted advanced disease and conferred survival to NHPs50. ZMapp combined 4G7 and 2G4 from ZMab50 with 13C6 from MB-003 (Fig. 2a) and differed from the predecessor cocktails in two important ways. All three antibodies were chimerized to attach a human IgG1 Fc to the murine Fab rather than the original murine IgG1 or IgG2a Fcs. Secondly, the component antibodies were produced in Nicotiniana with afucosylated/agalatosylated glycans. The change in Fc structure and glycosylation may be linked to the cocktail’s success relative to earlier antibodies from which it was derived.

Figure 2.

Figure 2.

Antibody treatments approved for evaluation in outbreaks of Ebola virus in 2018. (a) Superimposed CryoEM structures of the ZMapp antibodies 2G4 (green), 4G7 (yellow) and 13C633 (blue). Only the variable domains (Fv) could be built into cryoEM maps. ZMapp antibodies were produced in Nicotiniana for evaluation in rhesus macaques at 50 mg/kg on days 5, 8 and 11 after challenge50. ZMapp was also evaluated in humans in 20147. (c) Crystal structure of mAb 114124 (blue) in complex with Ebola virus GP, and its dosage information in rhesus macaques at 50 mg/kg on days 51. (D) REGN-EB3 containing antibodies 3470, 3471, and 3479 visualized by negative stain EM and dosage information in rhesus macaques52. REGN-3B3 was evaluated in three-dose and single-dose regimens.

Two novel human antibody therapies have now also been placed on the WHO list of investigational therapies for use in the 2018 Democratic Republic of the Congo (DRC) Ebola virus outbreaks (http://www.who.int/emergencies/ebola/MEURI-Ebola.pdf?ua=1). These include a mAb monotherapy from NIAID (mAb 114, a.k.a. VRC 608)51 (Fig. 2b) and a three-mAb cocktail from Regeneron (REGN 3470–3471-3479, a.k.a. REGN-EB3)52 (Fig. 2c). Both mAb 114 and the REGN-EB3 greatly promoted survival of non-human primates when delivered five days post-exposure.

mAb 114 demonstrates both neutralization and Fc-dependent cell-targeting activities in vitro51. In the VIC study, described in more detail below, all but one of the antibodies in the GP1 head-binding epitope group to which mAb 114 belongs exhibit both neutralization and cell-targeting functions29. Thus, the head epitope appears to exist at the physical intersection of those regions of GP that drive neutralization with the regions of GP that exhibit greater activation of phagocytosis and natural killer (NK) cells.

In contrast to the mAb 114 monotherapy, the three mAbs in REGN-EB3 provide complementary activities across the three antibody footprints. One component, REGN 3479, recognizes the conserved GP2 fusion loop and is neutralizing. A second component, REGN 3471, recognizes the outer glycan cap and has cell-targeting functions. The third, REGN 3470, binds the GP1 Head and offers both neutralizing and cell-targeting activities, including FcγRIIIa and other FcγR-related functions52. Thus, all three candidate immunotherapeutic treatments for Ebola virus incorporate a mix of neutralizing and cell-targeting functions, and bind to a mixture of neutralizing and cell-targeting epitopes (Fig. 2c). These newer candidate therapies were produced for NHP studies in 293Freestyle cells (mAb 114)51 and modified CHO cells (Regeneron)52.

A Comprehensive Study

In 2012, after the first protection results were described, we wondered if the neutralizing, but non-protective KZ52 and the poorly neutralizing, but more protective MB-003 examples were outliers or were typical features of many antibodies. We aimed to better determine, on a more statistically relevant scale, what kinds of antibodies or antibody features correlate with protection in animal models of Ebola virus infection. We also wondered what in vitro assays would best forecast in vivo protection, information which could streamline the research pipeline to identify or rationally design candidate therapeutics more quickly. Cognizant that each research group had their own assays and criteria for identifying, downselecting and producing antibodies, and that we needed to understand the range of antibody behavior, researchers in the filovirus field pooled their intellectual and physical resources into a single collaborative study.

Over 43 academic, industry and government laboratories across five continents united to form the Viral Hemorrhagic Fever Immunotherapeutic Consortium (VIC)53, supported by a Center of Excellence in Translational Research grant from the National Institute for Allergy and Infectious Disease (NIAID). This consortium performed a large, multidisciplinary study of filovirus antibodies isolated by multiple laboratories and produced by multiple methods. Some participating research laboratories contributed one or a few favorite antibodies (often selected based on neutralization), while other groups contributed a larger array of different samples. All antibodies were code-named to protect the investigators’ ability to publish findings from their own antibodies, many of which were not yet described in the literature.

This recently completed project29 analyzed 171 donated mAbs across a battery of in vitro and in vivo assays encompassing biochemistry, structural biology, glycan sequencing, neutralization, measurement of immune effector functions, and protection in the mouse model for each antibody in the study pool. Epitopes were determined by ELISA and structural biology at Scripps Research, and by alanine scanning at Integral Molecular. The neutralization activity of each mAb in the panel was measured by three different assays involving: (i) authentic Ebola virus at biosafety level (BSL)-4 (USAMRIID); (ii) a biologically contained Ebola virus (ΔVP30) for which an essential replication complex component, VP30, is expressed in trans at BSL2/3 (University of Wisconsin)54; and (iii) at BSL-2 recombinant vesicular stomatitis virus (rVSV) engineered to display Ebola virus GP on its surface (Albert Einstein College of Medicine). For the rVSV assay, the fraction of viral particles left un-neutralized at maximal antibody concentration was also analyzed55. In the rVSV and ΔVP30 systems, infected cells were quantified using a reporter gene inserted into the pseudoviral genome. In the authentic Ebola virus assay, infected cells were quantified by detection of KZ52 binding.

For immune effector function, we evaluated seven parameters for each of the 171 mAbs: the ability of human and murine phagocytes and neutrophils to engulf particles bearing Ebola virus GP trimers upon incubation with a given mAb (four different measurements); and the ability of each mAb to activate human natural killer cells (three measurements- cell surface expression of CD107a, secretion of the inflammatory chemokine MIP1-β (CCL4) and cytokine interferon γ (IFN-g)56,57).

Protection was evaluated in the mouse model for Ebola virus infection at both USAMRIID and the Public Health Agency of Canada (PHAC). To measure protection, groups of 10 mice were infected with mouse-adapted Ebola virus and treated 2 days after infection with a given mAb from the panel. Survival and body weight were monitored for 28 days and the percentage of mice surviving was determined. mAbs that resulted in 6 or more mice surviving (60%) were deemed protective. Machine learning approaches at Scripps, Ragon, and Los Alamos National Laboratory established correlation networks illustrating how antibody features related to each other and to in vivo protection in the mouse model29.

Several relationships were clear. First, epitopes linked to neutralization of virions in cell culture are predominantly located on the receptor-binding core of the glycoprotein that is retained during virus entry (Fig. 3a). Second, epitopes in the study linked to maximal phagocytic functions are predominantly located in the uppermost regions of GP, including the head, glycan cap and mucin-like domains (Fig. 3b). Antibodies with strong NK cell activity, however, are distributed more broadly across the glycoprotein surface.

Figure 3.

Figure 3.

Structure, epitopes of Ebola virus GP, and antibody functions. (a) Ebola virus GP is cleaved in the endosome to remove the glycan cap and mucin like domains. The remaining GP core, outlined in black, is termed GPcl and is competent for receptor binding. Antibodies against GPcl are much more likely to be neutralizing29. The base region of GP that encompasses the epitopes of KZ52, 2G4 and 4G7, is indicated by the orange circle. The glycan cap, including the 13C6 and 3471 epitopes, is deleted by enzymatic cleavage of GP and is not retained on GPcl. Light blue circles represent the mucin-like domain of GP (light blue), which is disordered in higher resolution structures. The viral membrane is indicated by a grey dotted line. (b) Antibodies against epitopes in the upper tier of GP, Tier 1, exhibit stronger effector functions on average than antibodies against the middle and lower regions of GP, as measured by immune ‘polyfunctionality’29. This greater sum of Fc-mediated immune functions in Tier 1 is driven by measures of phagocytosis. Epitopes of antibodies with strong NK activities occur across the GP29. (c) Mechanical neutralization and effector function protection mediated by antibodies. At left, IgG antibodies are shown anchoring to viral GP (blue) on Ebola virus (yellow). By anchoring to the GP, neutralizing antibodies prevent viral entry into potential target cells (purple). At right, IgG antibodies are illustrated as anchoring to viral GPs on Ebola virus (yellow) or an infected cell (purple). Through GP anchoring, the antibody Fc couples to neutrophils, monocytes, macrophages, NK cells and other immune mediators to mediate destruction and clearance of the virus and infected cells. Neutralization and effector-mediated clearance are independent functions and may occur on the same or different antibodies.

Physical clustering of neutralization-linked epitopes on the GP core and phagocytosis-linked epitopes on the upper, heavily glycosylated domains suggests that neutralization functions operate on virion-surface GP, while Fc-mediated phagocytic functions operate on cell-surface GP on infected cells (Fig. 3c). Fc-mediated NK cell functions are spread more broadly. Those mAbs that anchor to the GP core may interfere with steps involved in viral entry: receptor binding, cathepsin cleavage, and fusion5861. In contrast, antibodies bound to glycan cap and mucin-like domain epitopes on virion-surface GP would be removed by cathepsin cleavage of GP in the endosome37,6265, but remain on GP molecules displayed on the surface of infected cells. Enhanced phagocytic activity by antibodies bound to these upper regions may result from their greater accessibility to Fc receptors, and/or greater structural flexibility at these epitopes that facilitates multivalent binding, Fc-Fc interactions and immune cell recruitment.

The glycan cap and some GP1 head epitopes are also found on an abundantly secreted dimeric form of the GP, termed sGP. The role of sGP with respect to antibodies remains unclear, but it has been proposed to preferentially adsorb antibody, redirect the antibody response66 or form immune complexes33. Unexpectedly, across the VIC pool, sGP cross-reactivity was neither beneficial nor detrimental to the ability of an antibody to offer protection. Further, those mAbs that cross-react to sGP unexpectedly performed similarly in neutralization assays, whether sGP was present in wild-type amounts (authentic Ebola virus and ΔVP30 Ebola virus) or absent (rVSV). We did note in the VIC study that sGP–cross-reactive antibodies could activate FcR-bearing immune cells. Since sGP is secreted from infected cells, such sGP-containing immune complexes would not directly tag infected cells for destruction, but could form immune complexes that stimulate protection in other ways.

We opted to unblind KZ52 and the murine versions of 13C6, 2G4 and 4G7 to provide reference points. KZ52 is VIC 136, a human IgG1, and was produced for this study in CHO cells24. 4G7 and 2G4 in their original murine versions are VIC 18 and 24, murine IgG2a and IgG2b, respectively and were produced in hybridoma culture.

In the VIC study, KZ52, 2G4 and 4G7 all exhibit equivalent neutralization activity, but lower effector function scores than other antibodies analyzed, which included two of seven possible Fc-mediated functions. In contrast, cell-targeting activity for the ZMapp versions of 2G4 and 4G7 was likely improved by the glycan modifications afforded by production in Nicotiniana50.

Correlates of in vivo protection

In the overall VIC study on these and other antibodies in the field, our expectation from prior work on HIV-1 and other viruses was that neutralization would be a strong forecaster of protection6772. Of the 171 mAbs in the VIC panel, 20 (12%) robustly neutralized in every assay and also conferred strong levels of protection in the mouse model. Measures of neutralization were the strongest univariate predictors of protection in the study (ρ=0.61–0.68). Neutralization alone, however, was an incomplete predictor of in vivo success. Eight mAbs in the study (5%) neutralized potently and robustly, but failed to protect in vivo. Another nine mAbs in the study (5.4%) neutralized either poorly or not at all, yet did protect in vivo (Fig. 4). Of those nine, three offered high levels of in vivo protection in the absence of any measurable neutralization in any assay. The remaining six neutralized in some assays, albeit weakly (only 55% reduction in infected cells at 20 μg/ml compared to 99.5% reduction in infected cells at 0.5 μg/ml by more robustly neutralizing samples). For these nine antibodies, the ability to confer post-exposure protection in the mouse model was not predicted by their capacity to demonstrate in vitro neutralization.

Figure 4.

Figure 4.

Outliers in the VIC study. At left, nine antibodies protect ≥60% of mice, yet offer little to no neutralization (neutralization score of 0 (no neutralization measured at all in any assay) to 1 (weak neutralization measured in one assay, no neutralization measured in the other three) or 2 (strong neutralization in only one assay). Seven of these nine recognize Tier 1 or unknown epitopes. At right are eight antibodies that protect ≤30% of mice, despite relatively strong neutralization scores of 5–7. These antibodies typically neutralize at least moderately, and often potently, in all four measures, and recognize Tier 2 and 3 epitopes. In parentheses are the average polyfunctionality, neutralization score and protection value for the protective/poorly neutralizing group at left and the neutralizing/poorly protective group on the right. Each antibody in the panel is labelled atop a vertical column of boxes representing polyfunctionality, immune effector functions scored, protection and overall neutralization. For clarity, each measure is indicated by a colored box rather than an absolute value. Blue, yellow and red boxes indicate strong, moderate and weak scores, respectively. Polyfunctionality is a sum of yes/no answers for each of the seven effector functions measured including markers of both phagocytosis and natural killer cell function. For polyfunctionality, one point was given for strong or moderate activity on each of the seven readouts, with a maximum polyfunctionality score of 7. Antibody neutralization scores were calculated with 2, 1 or 0 points given for strong, moderate and weak/no activity on the four neutralization readouts, for a maximum score of 8. Epitopes of each antibody are indicated at the bottom with the color code at right.

We note that researcher choice in antibody selection could have biased the composition of the VIC study pool. In vitro neutralization, followed by in vivo protection in a mouse model are the two most common primary initial selection criteria in filovirus research. Those labs that contributed just a few antibodies tended to contribute neutralizing antibodies. We expect that those neutralizing antibodies already known to succeed in in vivo protection would be more likely to be contributed to the study than neutralizing antibodies already known to fail in in vivo protection. Such selection processes could amplify apparent correlations of neutralization to protection in our results. Further, if most labs first winnowed antibodies based on in vitro neutralization activity, those antibodies that protect without strong neutralization would not have been identified or contributed. As a result, the ~5% population of the VIC that appear to succeed primarily through cell-targeting Fc functions could be an underrepresentation of what is produced in an authentic polyclonal response. In addition, cell-targeting, non-neutralizing antibodies may function better in concert than alone, which is particularly relevant for polyclonal antibody elicited by vaccination or natural infection. Thus, the analysis of single monoclonal antibodies may have further undervalued the therapeutic potential of Fc-mediating antibody.

Importantly, the performance of antibodies for which neutralization did not adequately forecast protection could be linked to the presence or absence of NK cell and other Fc-mediated functions. All antibodies in the VIC study that neutralize well, but fail to protect in vivo, register zero to low scores of NK cell activity (Fig. 4). Meanwhile, all antibodies in the study that neutralize weakly or not at all, but do achieve in vivo protection, register high positive scores in natural killer and/or phagocytic functions. The VIC study suggests that evaluating and enhancing NK functions should be key goals of immunotherapeutic efforts.

Recent work in the research field further emphasizes the importance of Fc-mediated cell-targeting functions. Notably, in several studies of Ebola virus vaccines, total binding antibody, rather than neutralizing antibody, is the strongest correlate of protection7375. Total binding antibody, by definition, would include the fraction that operates via cell-targeted Fc functions. Another study of three novel mAbs against Ebola virus found that protection in the mouse model correlated with their ability to confer ADCC, not with their neutralization capacity76. These antibodies were functional only in wild-type mice bearing NK cells, but not in NK cell-deficient mice. The additional link to enhanced NK cell activity corroborates evidence from natural infection: fatal cases of Ebola virus disease are associated with lower NK cell function than survivable cases77.

In the comprehensive VIC study and in other recent work, we thus note a spectrum of antibody behavior. At one end of the spectrum are those antibodies that achieve protection by effector mechanisms alone. Nearly all of these mAbs target the upper and outer regions of the viral glycoprotein, and their physical position may facilitate antibody access to FcR-bearing cells as well as promote multivalent binding, and decoration of infected cells, circulating virions, or even secreted forms of sGP for immune activation. At the other end of the spectrum are potent neutralizing antibodies that protect in the absence of effector activity. These antibodies may achieve protection via Fab-mediated mechanical inactivation of the entry glycoprotein alone. Many of these antibodies bind to the GP core, in the middle to lower (membrane-proximal) tiers of the structure. Some of these epitopes may preclude access to effector cells and thus engineering to enhance effector function may or may not improve their performance. In the middle of the spectrum are antibodies that exhibit a variety of features, with a range of neutralization behavior and effector function strength.

Our immune effector assays were performed in a high-throughput standardized format to characterize the immune functions of all 171 VIC panel mAbs. Since the GP trimer display on beads may differ from that on cells, assays using GP-transfected or Ebola virus-infected cells are now in progress. Nonetheless, results from these high-throughput screens go beyond mere antibody affinity for GP to correlate with experimental protection in the mouse model. Moreover, epitopes that recruit an array of effector functions often have moderate GP affinity, whereas those antibodies that lack apparent effector function and protect via mechanical neutralization tend to have higher GP affinity.

A major limitation of the VIC study is that the in vivo protection results were obtained in the BALB/c mouse model. The apparent importance of effector functions of vaccines and immunotherapies tested in humans and NHPs, however, suggests that some of these general findings in the mouse model will translate to higher animals. However, the degree to which protection in the BALB/c mouse model compares to protection in larger animal models like guinea pigs, ferrets or NHPs, or even alternative mouse models such as those expressing human Fc receptors7881 awaits systematic study. Fc receptor-mediated mechanisms of protection among animal models are expected to differ since guinea pigs, NHPs and humans have evolved analogous, but often discrete Fc–FcR systems that have varying affinity for different antibody-FcR combinations. Indeed, mice lack both FcγRIIA and FcγRIIIB receptors, the latter being important for neutrophil activation in humans82. Mechanical neutralization solely due to Fab binding, which requires only a strict biophysical interaction between virus and antibody to confer protection, likely translates more faithfully from cell culture to rodents and to primates, perhaps reinforcing initial downselection via neutralization. In other words, antibodies that function primarily by physical, Fab-mediated blockade may have been easier to identify because they do not require immune functionality, which is, as yet, more difficult to recapitulate in vitro and across animal models.

The VIC study findings strongly suggest that we must also consider immune effector function in development of immunotherapeutics and capitalize on the full repertoire of immunological options at our disposal. We need to develop better bridging methods to study antibodies, for use in humans, across rodent and other non-human species used for in vivo evaluation. Given the potential differences among animal models and the effector functions themselves, a systematic comparison of different antibodies, that achieve protection by different mechanisms, across the standard animal models in use in filovirus research is needed. Such analyses would contribute to the understanding of the relative factors that contribute to protection in each animal model, as well as the role of immune effector function. Our previous expectation prompted the assumption that immunotherapies should focus on neutralizing antibodies. However, the existence of a class of antibodies that likely achieve protection via their Fc alone indicates that complementary or alternate routes to protection are under-appreciated and insufficiently understood. Identification of antibodies that mediate protection through immune effector function will facilitate dissection of the specific Fc features and geometry of Fc recognition that lead to protection, and will illuminate Fc engineering strategies that could improve protection by these and neutralizing antibodies alike.

Relevance of glycan modifications on antibodies

A common assumption regarding antibody production is that all in vitro-produced antibodies will be glycosylated similarly, and will faithfully represent the profiles that may be generated in vivo. However, more than two decades of monoclonal therapeutic research has pointed to major differences in glycosylation among recombinant antibodies produced within 293T- or across CHO-based or hybridoma-based expression systems, all of which likely do not reflect the original glycan profile that may have been present on the original antibody. Differences in culture conditions, cell quality, medium content, and even transfection conditions result in substantial changes in glycan content observed even within the same production laboratory8385. Across the VIC mAb panel, we observed significant heterogeneity in glycan content across the 19 glycan structures that we measured29,86, even among the 94 human IgG1 mAbs produced in traditional mammalian cell lines. We wondered, across the VIC pool, how glycan structure influenced induction of different effector functions.

We used an unbiased hierarchical clustering algorithm to group antibodies by glycan content alone, resulting in 17 different clusters (Fig. 5a). As a comparison, we also used an unsupervised principal component analysis of glycan content (Fig. 5b), which also separated the antibodies into these 17 clusters, or 17 distinct glycan profiles. To visualize the average glycan profile of the antibodies within each cluster, we generated a heat map that depicts the glycan content of each cluster: average total galactose content (divided into G0, G1, and G2), fucose, bisecting GlcNAc, and sialic acid (Fig. 5c). Next, to determine if the different clusters resulted in different functional antibody profiles, we graphed the average effector functional response of each cluster, as high, medium or low/no activity (Fig. 5d). Notably, several clusters were characterized by high functionality (clusters 1, 3, 11, and 12), whereas others were characterized by low activity (cluster 7) or limited functional diversity (clusters 4, 5, 9, 10, 13, 17). Of note, the highly functional clusters (1, 3, 11 and 12) represent different glycan profiles: afucosylated/agalactosylated (clusters 1 and 3) and mono-galactosylated, bisected glycan profiles (clusters 11 and 12). Both been independently associated with enhanced effector functions8789. In contrast, the low/limited functionality clusters were predominantly characterized by higher content of fucose in combination with sialic acid (clusters 7, 8, 9), suggesting that these glycans may hinder effector functionality. While afucsoylation was clearly linked to enhanced FcR functions, additional glycan changes were also linked to both unique functional and polyfunctional profiles. These known, and perhaps other, unexplored glycan profiles naturally and actively leverage Fc receptors to drive enhanced functionality. Thus, the selection of a production cell line and evaluation of the resulting antibody glycans should be considered in order to maximize functional activity by therapeutic antibodies. Systematic and comprehensive mapping of the individual Fc-glycan–functional profiles may provide a roadmap to improve protection in next-generation therapeutic design.

Figure 5.

Figure 5.

Specific glycan profiles are linked to distinct functional activity. (a) Hierarchical clustering of VIC mAbs using Fc glycan content revealed 17 clusters of antibodies within the panel. The dashed line across the dendogram indicates the cut-off used to define the clusters. Each cluster is represented by a different color, and cluster number is listed below each cluster. (b) Unsupervised principal component analysis of Fc glycan content of VIC mAbs. The mAbs are color-coded according to the clusters identified in (A) in the plot at left, and the loading plot showing the glycan features driving separation of the mAbs is illustrated at right. (c) Heatmap of the average total galactose (G0, G1, or G2), fucose, bisecting GlcNAc, or sialic acid content of the mAbs within each cluster. Dark blue represents the row minimum and red represents the row maximum. (d) The functional activity of mAbs within each cluster was averaged for each effector function measured, and categorized into high, medium, or low/negligible based on assay cutoffs. Each wedge is color coded by effector function, and the size of the wedge indicates the magnitude of response.

Strategy going forward

Overall, the finding that a relatively high number of anti-Ebola GP antibodies can confer protection either solely or significantly through effector mechanisms was unexpected. Moreover, the number of effector-driven antibodies in the VIC panel suggests that lack of neutralization does not necessarily indicate a lack of protective activity. The antibodies that strongly drive such effector functions, together with control antibodies that do not, will provide focused sets for further study. These study sets will allow dissection of which immune effector functions are important, and determination of how the importance of particular functions differs by epitope recognized (e.g., upper vs. lower on the viral antigen), and if the significance of effector function is retained in other animal models.

How should protective antibodies be identified in the first place? In a rapid selection process intended to identify potential therapeutic candidates quickly, the VIC study indicates that cell culture neutralization is indeed an effective downselection mechanism. Therefore, if time only permits one assay, neutralization alone would likely provide an effective rough cut, and would likely identify features that can translate from in vitro to in vivo studies. Indeed, if we had selected among the VIC pool based on neutralization alone, we would have selected 60% of the protective antibodies.

However, if we initially downselected the VIC pool based instead on Fc polyfunctionality (i.e., sum of the scores for the seven separate immune effector measures), we similarly would have selected about 60% of the protective antibodies. A two-pronged selection approach, using both neutralization and Fc function, would have predicted 96% of the protective antibodies.

The ability of an antibody to promote killing of infected cells should be actively sought in antibody discovery programs, rather than being simply a parallel feature during in vivo testing. We find, in particular, that NK effector function is likely the feature that tips the balance in favor of greater likelihood of protection. Screens for NK activity and substitutions to enhance NK activity may be particularly effective in identifying and enhancing protective activity. A better understanding of how to evaluate large panels of antibodies for Fc-mediated protection could reduce the number of animals required for eventual in vivo experimentation, and support the three Rs of in vivo research: Replacement, Reduction and Refinement90.

Although Fc-mediated protection is more challenging to understand in vitro and may not readily translate from cell culture to different animal species, these functions can be engineered using well-characterized point mutations that affect FcR affinity and the type of glycan modification8,10,91. Substitutions in the Fc such as L234A/L235A92, G236R/L236R93,94 and deletion of the glycosylation sequon at N29795,96, for example, diminish effector functions. Modification of the glycan linked to N297 can enhance functions. Reduced fucosylation and addition of bisecting GlcNAc addition improve ADCC91,97, and several afucosylated therapeutic antibodies have been approved or in human clinical trials for oncology applications98101. Amino acid substitutions in the Fc, such as S239D/I332E102, E345R103 and others8,102,104106, increase Fc affinity for FcγRIIa, FcγRIIIa and complement as well as enhance cell targeting. Multiple antibodies bearing these substitutions have also advanced to clinical trials for oncology107112. Substitutions such as S267E, H268F and S324T, which improve affinity for complement component C1q, and substitutions like E345K or E340G, which enhance IgG hexamerization103,113, stimulate complement-dependent cytotoxicity. Other substitutions, such as M252Y/S254T/T256E114 or M428L/N434S115, extend antibody half-life. Engineered antibodies against Staphylococcus aureus alpha-toxin116 and HIV-1117 bearing these substitutions have been evaluated in clinical trials. In contrast to the well-characterized Fc modifications, Fab-mediated neutralization is difficult to engineer, and there are no universal point mutations that can confer neutralizing activity to a non-neutralizing antibody. Attachment of an engineered Fc, however, to a potently neutralizing Fab could improve in vivo performance by improving targeting and destruction of infected cells so they do not continue to be viral factories, and by improving potential bioactivity at lower antibody concentrations, required for long-term protection and control. Which Fc modifications are ideal, and whether the optimal modifications differ according epitope on Ebola virus GP are still unclear.

Information gathered from studying these antibodies will help characterize which specific features beyond neutralization increase in vivo efficacy of antibodies, and will provide roadmaps for rational engineering and improvement of immunotherapeutics. A greater understanding of the features of these non-neutralizing, yet highly protective samples may also provide strategies to identify such antibodies in the sera of vaccinees. Such studies could be broadly applicable as recent work suggests that induction of cell-targeting antibodies is desirable against a variety of pathogens that threaten human health118122, particularly when neutralizing antibodies are difficult to elicit.

ACKNOWLEDGEMENTS

The authors gratefully acknowledge U19AI107962, a Center of Excellence in Translational Research (CETR) award from NIH/NIAID, and the many members of the Viral Hemorrhagic Fever Immunotherapeutic Consortium who contributed antibodies (many of which were unpublished) and analytical effort to the project, including K.G. Andersen, A.B. Ward and D.R. Burton, R. Ahmed and C.W. Davis (Emory University), M.J. Aman (Integrated BioTherapeutics), A. Bukreyev (University of Texas Medical Branch), K. Chandran and J.R. Lai (Albert Einstein College of Medicine), J.E. Crowe, Jr. (Vanderbilt University School of Medicine), J.M. Dye (USAMRIID), B. Doranz (Integral Molecular), H. Feldmann (NIAID Rocky Mountain Lab), G. Georgiou (University of Texas), G.G. Olinger and L. Hensley (NIAID Integrated Research Facility), Y. Kawaoka (University of Wisconsin), G.P. Kobinger (Université Laval), K. Wagh and B. Korber (Los Alamos National Labs), F. Krammer (Icahn School of Medicine at Mt. Sinai), C.A. Kyratsous (Regeneron), C. Nykiforuk (Emergent Biosciences), X. Qiu (Public Health Agency of Canada), A. Sprecher (Médecins Sans Frontières), A.R. Townsend (University of Oxford), V. Volchkov (Université Lyon), L.M. Walker (Adimab), C.-I. Wang (A*STAR), L. Zeitlin (Mapp Biopharmaceutical) and the students, postdoctoral fellows, technicians and staff scientists in their labs, as well as J.V. Ravetch (Rockefeller University) for valuable discussions. This is manuscript #29662 from Scripps Research.

Footnotes

COMPETING FINANCIAL INTERESTS

The authors declare no competing interests.

REFERENCES

  • 1.Carter PJ & Lazar GA Next generation antibody drugs: pursuit of the ‘high-hanging fruit’. Nat. Rev. Drug Discov 17, 197–223 (2018). [DOI] [PubMed] [Google Scholar]
  • 2.Salazar G, Zhang N, Fu T-M & An Z Antibody therapies for the prevention and treatment of viral infections. NPJ Vaccines 2, 19 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Lu LL, Suscovich TJ, Fortune SM & Alter G Beyond binding: antibody effector functions in infectious diseases. Nat. Rev. Immunol 18, 46–61 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Casadevall A Antibody-based therapies for emerging infectious diseases. Emerg. Infect. Dis 2, 200–208 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Corti D et al. Prophylactic and postexposure efficacy of a potent human monoclonal antibody against MERS coronavirus. Proc. Natl. Acad. Sci. U. S. A 112, 10473–10478 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Burton DR & Saphire EO Swift antibodies to counter emerging viruses. Proc. Natl. Acad. Sci. U. S. A 112, 10082–10083 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.PREVAIL II Writing Group et al. A Randomized, Controlled Trial of ZMapp for Ebola Virus Infection. N. Engl. J. Med 375, 1448–1456 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Sondermann P & Szymkowski DE Harnessing Fc receptor biology in the design of therapeutic antibodies. Curr. Opin. Immunol 40, 78–87 (2016). [DOI] [PubMed] [Google Scholar]
  • 9.Nimmerjahn F Translating Inhibitory Fc Receptor Biology into Novel Therapeutic Approaches. J. Clin. Immunol 36 Suppl 1, 83–87 (2016). [DOI] [PubMed] [Google Scholar]
  • 10.Hogarth PM & Pietersz GA Fc receptor-targeted therapies for the treatment of inflammation, cancer and beyond. Nat. Rev. Drug Discov 11, 311–331 (2012). [DOI] [PubMed] [Google Scholar]
  • 11.Howes SC, Koning RI & Koster AJ Correlative microscopy for structural microbiology. Curr. Opin. Microbiol 43, 132–138 (2018). [DOI] [PubMed] [Google Scholar]
  • 12.Hampton CM et al. Correlated fluorescence microscopy and cryo-electron tomography of virus-infected or transfected mammalian cells. Nat. Protoc 12, 150–167 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Kellner C, Derer S, Valerius T & Peipp M Boosting ADCC and CDC activity by Fc engineering and evaluation of antibody effector functions. Methods 65, 105–113 (2014). [DOI] [PubMed] [Google Scholar]
  • 14.Bournazos S, DiLillo DJ & Ravetch JV The role of Fc-FcγR interactions in IgG-mediated microbial neutralization. J. Exp. Med 212, 1361–1369 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Lewis GK Role of Fc-mediated antibody function in protective immunity against HIV-1. Immunology 142, 46–57 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Treffers LW et al. Genetic variation of human neutrophil Fcγ receptors and SIRPα in antibody-dependent cellular cytotoxicity towards cancer cells. Eur. J. Immunol 48, 344–354 (2018). [DOI] [PubMed] [Google Scholar]
  • 17.Bakema JE & van Egmond M Fc receptor-dependent mechanisms of monoclonal antibody therapy of cancer. Curr. Top. Microbiol. Immunol 382, 373–392 (2014). [DOI] [PubMed] [Google Scholar]
  • 18.Ram S et al. Utilizing complement evasion strategies to design complement-based antibacterial immunotherapeutics: Lessons from the pathogenic Neisseriae. Immunobiology 221, 1110–1123 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Bournazos S et al. Broadly neutralizing anti-HIV-1 antibodies require Fc effector functions for in vivo activity. Cell 158, 1243–1253 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Almagro JC, Daniels-Wells TR, Perez-Tapia SM & Penichet ML Progress and Challenges in the Design and Clinical Development of Antibodies for Cancer Therapy. Front. Immunol 8, 1751 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Maruyama T et al. Ebola virus can be effectively neutralized by antibody produced in natural human infection. J. Virol 73, 6024–6030 (1999). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Lee JE et al. Structure of the Ebola virus glycoprotein bound to an antibody from a human survivor. Nature 454, 177–182 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Parren PWHI, Geisbert TW, Maruyama T, Jahrling PB & Burton DR Pre- and postexposure prophylaxis of Ebola virus infection in an animal model by passive transfer of a neutralizing human antibody. J. Virol 76, 6408–6412 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Oswald WB et al. Neutralizing antibody fails to impact the course of Ebola virus infection in monkeys. PLoS Pathog 3, e9 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Dye JM et al. Postexposure antibody prophylaxis protects nonhuman primates from filovirus disease. Proc. Natl. Acad. Sci. U. S. A 109, 5034–5039 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Qiu X et al. Ebola GP-specific monoclonal antibodies protect mice and guinea pigs from lethal Ebola virus infection. PLoS Negl. Trop. Dis 6, e1575 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Qiu X et al. Successful treatment of ebola virus-infected cynomolgus macaques with monoclonal antibodies. Sci. Transl. Med 4, 138ra81 (2012). [DOI] [PubMed] [Google Scholar]
  • 28.Murin CD et al. Structures of protective antibodies reveal sites of vulnerability on Ebola virus. Proc. Natl. Acad. Sci. U. S. A 111, 17182–17187 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Saphire EO, Schendel SL, Fusco ML, Gangavarapu K, Gunn BM, Wec AZ, Halfmann PJ, Brannan JM, Herbert AS, Qiu X, Wagh K, He S, Giorgi EE, Theiler J, Pommert KBJ, Krause TB, Turner HL, Murin CD, Pallesen J, Davidson E, Ahmed R, Aman MJ, Bukreyev A, Burton DR, Crowe JE, Davis CW, Georgiou G, Krammer F, Kyratsous FA, Lai JR, Nykiforuk C, Pauly MH, Rijal P, Takada A, Townsend AR, Volchkov V, Walker LM, Wang CI, Zeitlin L, Doranz BJ, Ward AB, Korber BK, Kobinger GP, Andersen KG, Kawaoka Y, Alter A, Chandran K, and Dye JM for the Viral Hemorrhagic Fever Immunotherapeutic Consortium. Systematic analysis of monoclonal antibodies against Ebola virus GP defines features that contribute to protection. Cell 174, 938–952 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Audet J et al. Molecular characterization of the monoclonal antibodies composing ZMAb: a protective cocktail against Ebola virus. Sci. Rep 4, 6881 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Olinger GG Jr et al. Delayed treatment of Ebola virus infection with plant-derived monoclonal antibodies provides protection in rhesus macaques. Proc. Natl. Acad. Sci. U. S. A 109, 18030–18035 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pettitt J et al. Therapeutic intervention of Ebola virus infection in rhesus macaques with the MB-003 monoclonal antibody cocktail. Sci. Transl. Med 5, 199ra113 (2013). [DOI] [PubMed] [Google Scholar]
  • 33.Pallesen J et al. Structures of Ebola virus GP and sGP in complex with therapeutic antibodies. Nat Microbiol 1, 16128 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wilson JA et al. Epitopes involved in antibody-mediated protection from Ebola virus. Science 287, 1664–1666 (2000). [DOI] [PubMed] [Google Scholar]
  • 35.Davidson E et al. Mechanism of Binding to Ebola Virus Glycoprotein by the ZMapp, ZMAb, and MB-003 Cocktail Antibodies. J. Virol 89, 10982–10992 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Hood CL et al. Biochemical and structural characterization of cathepsin L-processed Ebola virus glycoprotein: implications for viral entry and immunogenicity. J. Virol 84, 2972–2982 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Schornberg K et al. Role of endosomal cathepsins in entry mediated by the Ebola virus glycoprotein. J. Virol 80, 4174–4178 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zeitlin L et al. Enhanced potency of a fucose-free monoclonal antibody being developed as an Ebola virus immunoprotectant. Proc. Natl. Acad. Sci. U. S. A 108, 20690–20694 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Strasser R et al. Generation of glyco-engineered Nicotiana benthamiana for the production of monoclonal antibodies with a homogeneous human-like N-glycan structure. Plant Biotechnol. J 6, 392–402 (2008). [DOI] [PubMed] [Google Scholar]
  • 40.Ackerman ME et al. Natural variation in Fc glycosylation of HIV-specific antibodies impacts antiviral activity. J. Clin. Invest 123, 2183–2192 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Mahan AE et al. Antigen-Specific Antibody Glycosylation Is Regulated via Vaccination. PLoS Pathog 12, e1005456 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Nimmerjahn F, Anthony RM & Ravetch JV Agalactosylated IgG antibodies depend on cellular Fc receptors for in vivo activity. Proc. Natl. Acad. Sci. U. S. A 104, 8433–8437 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Arnold JN, Wormald MR, Sim RB, Rudd PM & Dwek RA The impact of glycosylation on the biological function and structure of human immunoglobulins. Annu. Rev. Immunol 25, 21–50 (2007). [DOI] [PubMed] [Google Scholar]
  • 44.Anthony RM & Nimmerjahn F The role of differential IgG glycosylation in the interaction of antibodies with FcγRs in vivo. Curr. Opin. Organ Transplant 16, 7–14 (2011). [DOI] [PubMed] [Google Scholar]
  • 45.Jefferis R Glycosylation of recombinant antibody therapeutics. Biotechnol. Prog 21, 11–16 (2005). [DOI] [PubMed] [Google Scholar]
  • 46.Popp O et al. Development of a pre-glycoengineered CHO-K1 host cell line for the expression of antibodies with enhanced Fc mediated effector function. MAbs 10, 290–303 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Jiang X-R et al. Advances in the assessment and control of the effector functions of therapeutic antibodies. Nat. Rev. Drug Discov 10, 101–111 (2011). [DOI] [PubMed] [Google Scholar]
  • 48.Schuster M et al. Improved effector functions of a therapeutic monoclonal Lewis Y-specific antibody by glycoform engineering. Cancer Res 65, 7934–7941 (2005). [DOI] [PubMed] [Google Scholar]
  • 49.Yamane-Ohnuki N & Satoh M Production of therapeutic antibodies with controlled fucosylation. MAbs 1, 230–236 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Qiu X et al. Reversion of advanced Ebola virus disease in nonhuman primates with ZMapp. Nature 514, 47–53 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Corti D et al. Protective monotherapy against lethal Ebola virus infection by a potently neutralizing antibody. Science 351, 1339–1342 (2016). [DOI] [PubMed] [Google Scholar]
  • 52.Pascal KE et al. Development of clinical-stage human monoclonal antibodies that treat advanced Ebola virus disease in non-human primates. J. Infect. Dis (2018). doi: 10.1093/infdis/jiy285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Saphire EO et al. How to turn competitors into collaborators. Nature 541, 283–285 (2017). [DOI] [PubMed] [Google Scholar]
  • 54.Halfmann P et al. Generation of biologically contained Ebola viruses. Proc. Natl. Acad. Sci. U. S. A 105, 1129–1133 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Wong AC, Sandesara RG, Mulherkar N, Whelan SP & Chandran K A forward genetic strategy reveals destabilizing mutations in the Ebolavirus glycoprotein that alter its protease dependence during cell entry. J. Virol 84, 163–175 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Ackerman ME, Barouch DH & Alter G Systems serology for evaluation of HIV vaccine trials. Immunol. Rev 275, 262–270 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Alter G, Malenfant JM & Altfeld M CD107a as a functional marker for the identification of natural killer cell activity. J. Immunol. Methods 294, 15–22 (2004). [DOI] [PubMed] [Google Scholar]
  • 58.Shedlock DJ et al. Antibody-mediated neutralization of Ebola virus can occur by two distinct mechanisms. Virology 401, 228–235 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Flyak AI et al. Mechanism of human antibody-mediated neutralization of Marburg virus. Cell 160, 893–903 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.King LB et al. The Marburgvirus-Neutralizing Human Monoclonal Antibody MR191 Targets a Conserved Site to Block Virus Receptor Binding. Cell Host Microbe 23, 101–109.e4 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Hashiguchi T et al. Structural basis for Marburg virus neutralization by a cross-reactive human antibody. Cell 160, 904–912 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Chandran K, Sullivan NJ, Felbor U, Whelan SP & Cunningham JM Endosomal proteolysis of the Ebola virus glycoprotein is necessary for infection. Science 308, 1643–1645 (2005). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Miller EH et al. Ebola virus entry requires the host-programmed recognition of an intracellular receptor. EMBO J 31, 1947–1960 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Gong X et al. Structural Insights into the Niemann-Pick C1 (NPC1)-Mediated Cholesterol Transfer and Ebola Infection. Cell 165, 1467–1478 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Kaletsky RL, Simmons G & Bates P Proteolysis of the Ebola virus glycoproteins enhances virus binding and infectivity. J. Virol 81, 13378–13384 (2007). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Mohan GS, Li W, Ye L, Compans RW & Yang C Antigenic subversion: a novel mechanism of host immune evasion by Ebola virus. PLoS Pathog 8, e1003065 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Barba-Spaeth G et al. Structural basis of potent Zika-dengue virus antibody cross-neutralization. Nature 536, 48–53 (2016). [DOI] [PubMed] [Google Scholar]
  • 68.Corti D et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 333, 850–856 (2011). [DOI] [PubMed] [Google Scholar]
  • 69.Corti D et al. Cross-neutralization of four paramyxoviruses by a human monoclonal antibody. Nature 501, 439–443 (2013). [DOI] [PubMed] [Google Scholar]
  • 70.de Alwis R et al. Identification of human neutralizing antibodies that bind to complex epitopes on dengue virions. Proc. Natl. Acad. Sci. U. S. A 109, 7439–7444 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Huang J et al. Identification of a CD4-Binding-Site Antibody to HIV that Evolved Near-Pan Neutralization Breadth. Immunity 45, 1108–1121 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Burton DR & Mascola JR Antibody responses to envelope glycoproteins in HIV-1 infection. Nat. Immunol 16, 571–576 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Zhou Y & Sullivan NJ Immunology and evolvement of the adenovirus prime, MVA boost Ebola virus vaccine. Curr. Opin. Immunol 35, 131–136 (2015). [DOI] [PubMed] [Google Scholar]
  • 74.Stanley DA et al. Chimpanzee adenovirus vaccine generates acute and durable protective immunity against ebolavirus challenge. Nat. Med 20, 1126–1129 (2014). [DOI] [PubMed] [Google Scholar]
  • 75.Qiu X et al. Mucosal immunization of cynomolgus macaques with the VSVDeltaG/ZEBOVGP vaccine stimulates strong ebola GP-specific immune responses. PLoS One 4, e5547 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Liu Q et al. Antibody-dependent-cellular-cytotoxicity-inducing antibodies significantly affect the post-exposure treatment of Ebola virus infection. Sci. Rep 7, 45552 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Cimini E et al. Different features of Vδ2 T and NK cells in fatal and non-fatal human Ebola infections. PLoS Negl. Trop. Dis 11, e0005645 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Gillis CM et al. Mechanisms of anaphylaxis in human low-affinity IgG receptor locus knock-in mice. J. Allergy Clin. Immunol 139, 1253–1265.e14 (2017). [DOI] [PubMed] [Google Scholar]
  • 79.Gillis C, Gouel-Chéron A, Jönsson F & Bruhns P Contribution of Human FcγRs to Disease with Evidence from Human Polymorphisms and Transgenic Animal Studies. Front. Immunol 5, 254 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Smith P, DiLillo DJ, Bournazos S, Li F & Ravetch JV Mouse model recapitulating human Fcγ receptor structural and functional diversity. Proceedings of the National Academy of Sciences 109, 6181–6186 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Bournazos S, DiLillo DJ & Ravetch JV humanized mice to study FcγR function. Curr. Top. Microbiol. Immunol 382, 237–248 (2014). [DOI] [PubMed] [Google Scholar]
  • 82.Nimmerjahn F & Ravetch JV Fcgamma receptors as regulators of immune responses. Nat. Rev. Immunol 8, 34–47 (2008). [DOI] [PubMed] [Google Scholar]
  • 83.Sheeley DM, Merrill BM & Taylor LC Characterization of monoclonal antibody glycosylation: comparison of expression systems and identification of terminal alpha-linked galactose. Anal. Biochem 247, 102–110 (1997). [DOI] [PubMed] [Google Scholar]
  • 84.Hills AE, Patel A, Boyd P & James DC Metabolic control of recombinant monoclonal antibody N-glycosylation in GS-NS0 cells. Biotechnol. Bioeng 75, 239–251 (2001). [DOI] [PubMed] [Google Scholar]
  • 85.Baker KN et al. Metabolic control of recombinant protein N-glycan processing in NS0 and CHO cells. Biotechnol. Bioeng 73, 188–202 (2001). [DOI] [PubMed] [Google Scholar]
  • 86.Gunn BM, Yu W-H, Karim MM, Brannan J, Wec AZ, Fusco ML, Schendel SL, Gangavarapu K, Das J, Suscovich T, Andersen KG, Chandran K, Zeitlin L, Lauffenburger D, Dye JM, Saphire EO, and Alter GA. Dissecting the role of Fc and Fab mediated functions in Ebola virus specific immunity. Cell Host Microbe 24, 221–233 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Shields RL et al. Lack of fucose on human IgG1 N-linked oligosaccharide improves binding to human Fcgamma RIII and antibody-dependent cellular toxicity. J. Biol. Chem 277, 26733–26740 (2002). [DOI] [PubMed] [Google Scholar]
  • 88.Chung AW et al. Identification of antibody glycosylation structures that predict monoclonal antibody Fc-effector function. AIDS 28, 2523–2530 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Mahan AE et al. Correction: Antigen-Specific Antibody Glycosylation Is Regulated via Vaccination. PLoS Pathog 12, e1005694 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Sneddon LU, Halsey LG & Bury NR Considering aspects of the 3Rs principles within experimental animal biology. J. Exp. Biol 220, 3007–3016 (2017). [DOI] [PubMed] [Google Scholar]
  • 91.Jefferis R Isotype and glycoform selection for antibody therapeutics. Arch. Biochem. Biophys 526, 159–166 (2012). [DOI] [PubMed] [Google Scholar]
  • 92.Hessell AJ et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature 449, 101–104 (2007). [DOI] [PubMed] [Google Scholar]
  • 93.Horton HM et al. Antibody-mediated coengagement of FcγRIIb and B cell receptor complex suppresses humoral immunity in systemic lupus erythematosus. J. Immunol 186, 4223–4233 (2011). [DOI] [PubMed] [Google Scholar]
  • 94.Chu SY et al. Reduction of total IgE by targeted coengagement of IgE B-cell receptor and FcγRIIb with Fc-engineered antibody. J. Allergy Clin. Immunol 129, 1102–1115 (2012). [DOI] [PubMed] [Google Scholar]
  • 95.Nose M & Wigzell H Biological significance of carbohydrate chains on monoclonal antibodies. Proc. Natl. Acad. Sci. U. S. A 80, 6632–6636 (1983). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Leatherbarrow RJ et al. Effector functions of a monoclonal aglycosylated mouse IgG2a: binding and activation of complement component C1 and interaction with human monocyte Fc receptor. Mol. Immunol 22, 407–415 (1985). [DOI] [PubMed] [Google Scholar]
  • 97.Tobinai K, Klein C, Oya N & Fingerle-Rowson G A Review of Obinutuzumab (GA101), a Novel Type II Anti-CD20 Monoclonal Antibody, for the Treatment of Patients with B-Cell Malignancies. Adv. Ther 34, 324–356 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Beck A & Reichert JM Marketing approval of mogamulizumab: a triumph for glyco-engineering. MAbs 4, 419–425 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Hoy SM Obinutuzumab: A Review of Its Use in Patients with Chronic Lymphocytic Leukaemia. Drugs 75, 285–296 (2015). [DOI] [PubMed] [Google Scholar]
  • 100.Ghazi A, Trikha A & Calhoun WJ Benralizumab--a humanized mAb to IL-5Rα with enhanced antibody-dependent cell-mediated cytotoxicity--a novel approach for the treatment of asthma. Expert Opin. Biol. Ther 12, 113–118 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Robak T GA-101, a third-generation, humanized and glyco-engineered anti-CD20 mAb for the treatment of B-cell lymphoid malignancies. Curr. Opin. Investig. Drugs 10, 588–596 (2009). [PubMed] [Google Scholar]
  • 102.Lazar GA et al. Engineered antibody Fc variants with enhanced effector function. Proc. Natl. Acad. Sci. U. S. A 103, 4005–4010 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Diebolder CA et al. Complement is activated by IgG hexamers assembled at the cell surface. Science 343, 1260–1263 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Shields RL et al. 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. J. Biol. Chem 276, 6591–6604 (2001). [DOI] [PubMed] [Google Scholar]
  • 105.Stavenhagen JB et al. Fc Optimization of Therapeutic Antibodies Enhances Their Ability to Kill Tumor Cells In vitro and Controls Tumor Expansion In vivo via Low-Affinity Activating Fcγ Receptors. Cancer Res 67, 8882–8890 (2007). [DOI] [PubMed] [Google Scholar]
  • 106.Richards JO et al. Optimization of antibody binding to FcγRIIa enhances macrophage phagocytosis of tumor cells. Mol. Cancer Ther 7, 2517–2527 (2008). [DOI] [PubMed] [Google Scholar]
  • 107.Jurczak W et al. Phase IIa Study of Single-Agent MOR208 in Patients with Relapsed or Refractory B-Cell Non-Hodgkin’s Lymphoma. Blood 126, 1528–1528 (2015). [Google Scholar]
  • 108.Kellner C et al. The Fc-engineered CD19 antibody MOR208 (XmAb5574) induces natural killer cell-mediated lysis of acute lymphoblastic leukemia cells from pediatric and adult patients. Leukemia 27, 1595–1598 (2013). [DOI] [PubMed] [Google Scholar]
  • 109.Zalevsky J et al. The impact of Fc engineering on an anti-CD19 antibody: increased Fcγ receptor affinity enhances B-cell clearing in nonhuman primates. Blood 113, 3735–3743 (2009). [DOI] [PubMed] [Google Scholar]
  • 110.Kumar A et al. A phase 1 dose-escalation study of XmAb® 2513 in patients with relapsed or refractory Hodgkin lymphoma. Br. J. Haematol 168, 902–904 (2015). [DOI] [PubMed] [Google Scholar]
  • 111.Lee EM et al. Efficacy of an Fc-modified anti-CD123 antibody (CSL362) combined with chemotherapy in xenograft models of acute myelogenous leukemia in immunodeficient mice. Haematologica 100, 914–926 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Li G et al. Dual functional monoclonal antibody PF-04605412 targets integrin alpha5beta1 and elicits potent antibody-dependent cellular cytotoxicity. Cancer Res 70, 10243–10254 (2010). [DOI] [PubMed] [Google Scholar]
  • 113.de Jong RN et al. A Novel Platform for the Potentiation of Therapeutic Antibodies Based on Antigen-Dependent Formation of IgG Hexamers at the Cell Surface. PLoS Biol 14, e1002344 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114.Dall’Acqua WF, Kiener PA & Wu H Properties of human IgG1s engineered for enhanced binding to the neonatal Fc receptor (FcRn). J. Biol. Chem 281, 23514–23524 (2006). [DOI] [PubMed] [Google Scholar]
  • 115.Borrok MJ et al. pH-dependent binding engineering reveals an FcRn affinity threshold that governs IgG recycling. J. Biol. Chem 290, 4282–4290 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Yu X-Q et al. Safety, Tolerability, and Pharmacokinetics of MEDI4893, an Investigational, Extended-Half-Life, Anti-Staphylococcus aureus Alpha-Toxin Human Monoclonal Antibody, in Healthy Adults. Antimicrob. Agents Chemother 61, (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Gaudinski MR et al. Safety and pharmacokinetics of the Fc-modified HIV-1 human monoclonal antibody VRC01LS: A Phase 1 open-label clinical trial in healthy adults. PLoS Med 15, e1002493 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Lewis GK et al. Beyond Viral Neutralization. AIDS Res. Hum. Retroviruses 33, 760–764 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Corey L et al. Immune correlates of vaccine protection against HIV-1 acquisition. Sci Transl Med 2015; 7 (310): 310rv7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Henry Dunand CJ et al. Both Neutralizing and Non-Neutralizing Human H7N9 Influenza Vaccine-Induced Monoclonal Antibodies Confer Protection. Cell Host Microbe 19, 800–813 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Bootz A et al. Protective capacity of neutralizing and non-neutralizing antibodies against glycoprotein B of cytomegalovirus. PLoS Pathog 13, e1006601 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Horwitz JA et al. Non-neutralizing Antibodies Alter the Course of HIV-1 Infection In Vivo. Cell 170, 637–648.e10 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Lee JE et al. Crystal structure of the 13F6–1-2 Fab fragment bound to its Ebola virus glycoprotein peptide epitope (2008). doi: 10.2210/pdb2qhr/pdb [DOI] [Google Scholar]
  • 124.Misasi J et al. Structural and molecular basis for Ebola virus neutralization by protective human antibodies. Science 351, 1343–1346 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES