Next Article in Journal
Oxygen Levels Affect Macrophage HIV-1 Gene Expression and Delay Resolution of Inflammation in HIV-Tg Mice
Next Article in Special Issue
Comparative Analyses of the Antiviral Activities of IgG and IgA Antibodies to Influenza A Virus M2 Protein
Previous Article in Journal
Comparative Seasonal Respiratory Virus Epidemic Timing in Utah
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Influenza A Virus Antibodies with Antibody-Dependent Cellular Cytotoxicity Function

1
Department of Biology and Microbiology, South Dakota State University, Brookings, SD 57007, USA
2
Zuckerman Institute, Columbia University, New York, NY 10027, USA
3
BioSNTR, Brookings, SD 57007, USA
*
Authors to whom correspondence should be addressed.
Viruses 2020, 12(3), 276; https://doi.org/10.3390/v12030276
Submission received: 24 January 2020 / Revised: 26 February 2020 / Accepted: 26 February 2020 / Published: 1 March 2020
(This article belongs to the Special Issue Influenza Virus Vaccines)

Abstract

:
Influenza causes millions of cases of hospitalizations annually and remains a public health concern on a global scale. Vaccines are developed and have proven to be the most effective countermeasures against influenza infection. Their efficacy has been largely evaluated by hemagglutinin inhibition (HI) titers exhibited by vaccine-induced neutralizing antibodies, which correlate fairly well with vaccine-conferred protection. Contrarily, non-neutralizing antibodies and their therapeutic potential are less well defined, yet, recent advances in anti-influenza antibody research indicate that non-neutralizing Fc-effector activities, especially antibody-dependent cellular cytotoxicity (ADCC), also serve as a critical mechanism in antibody-mediated anti-influenza host response. Monoclonal antibodies (mAbs) with Fc-effector activities have the potential for prophylactic and therapeutic treatment of influenza infection. Inducing mAbs mediated Fc-effector functions could be a complementary or alternative approach to the existing neutralizing antibody-based prevention and therapy. This review mainly discusses recent advances in Fc-effector functions, especially ADCC and their potential role in influenza countermeasures. Considering the complexity of anti-influenza approaches, future vaccines may need a cocktail of immunogens in order to elicit antibodies with broad-spectrum protection via multiple protective mechanisms.

1. Introduction

Influenza viruses cause severe respiratory illness, leading to 290,000–650,000 deaths annually worldwide as estimated by the World Health Organization (WHO) [1]. Influenza viruses consist of four known genera, A, B, C, and D, which belong to the family Orthomyxoviridae [2]. Of these four genera, influenza A virus causes the greatest mortality and is the most common cause of both seasonal and pandemic influenza outbreaks. Influenza B virus can cause seasonal influenza while the influenza C virus can infect children with mild respiratory symptoms. Little is known about the impact of recently discovered influenza D virus on human health [3,4].
Hemagglutinin (HA) and neuraminidase (NA) are the two primary viral surface glycoproteins (Figure 1) involved in critical steps of the influenza life cycle. The trimeric HA protein is comprised of two domains: the head domain and the stalk domain. HA head domain contains the receptor binding site (RBS) that binds to sialic acids (SAs) on the susceptible cells to initiate the virus’ replication cycle. After the virus is endocytosed, the fusion peptide in the HA stalk was exposed to mediate the membrane fusion towards releasing viral ribonucleoprotein (RNP) complex into the cytoplasm and subsequently to the nucleus of infected cells [2]. After the newly assembled influenza virus buds from the infected cell, HA on the virion still interacts with the SA receptors on the host cell membrane. The tetrameric NA spike functions to release the viral progeny through cleaving α-ketosidic linkage between the SA and an adjacent sugar residue [5].
The HA and NA proteins are also highly immunogenic and antibodies targeting both glycoproteins can be isolated after natural infection or vaccination. Through binding to viral surface proteins HA and NA, antibodies can block the essential steps in the virus replication cycle, thereby limiting the spread of infection. Due to the host immune pressure and error-prone RNA polymerase, HA and NA are very plastic and display difference in antigenic properties. According to the antigenic difference, influenza A virus can divide into 18 HA (H1–H18) and 11 NA (N1–N11) subtypes [6]. According to the Weekly U.S. Influenza Surveillance Report released by CDC, H1N1(pdm09) and B/Victoria lineage viruses are equally dominant and responsible for the majority of death cases during the 2019–2020 influenza season [7].
Vaccination is the best countermeasure to prevent and control influenza [8]. Live attenuated, inactivated and recombinant HA vaccines are the three types of licensed seasonal influenza vaccines [9]. These vaccines conferred considerable protection in combating influenza by inducing antibodies that target HA. However, their efficacy can be significantly reduced when novel viruses emerge, or when there is a mismatch between the vaccine strain and the circulating influenza strain [8]. Therefore, the ultimate goal is to develop a universal vaccine that could confer long-lasting protection against multiple influenza strains, including the drifted seasonal influenza viruses and antigenically distinct viruses. Several approaches are employed to achieve that goal, such as stalk-based immunogen [10], chimeric HA immunogen strategies [11] and computationally optimized broadly reactive antigen (COBRA)-based vaccines [12,13]. Elicitation of antibodies exhibiting ADCC activities also contributes to the design of universal vaccines, which are thought to confer broad-spectrum protection [14].
Previous antibody protective efficiency was measured by their capability to prevent HA binding via neutralization assay and hemagglutination inhibition assay [14,15], antibodies without these functions were less well defined. However, increasing evidence suggests that non-neutralizing antibodies (nnAbs) can confer protection via multiple mechanisms without disturbing virus entry or membrane fusion, such as activating complement, increasing phagocytosis, targeting internal viral proteins and eliciting fragment crystallizable (Fc)-effector functions [16,17,18,19]. Unlike neutralizing antibodies, nnAbs do not bind to the RBS in the HA head and fail to inhibit hemagglutination. Therefore, these antibodies demonstrate no detectable in vitro hemagglutination inhibition (HI) titer or neutralizing activity commonly considered as the benchmark standard of vaccine efficacy. Nonetheless, recent studies have validated that nnAbs can also contribute to the vaccine-elicited protection against influenza virus infection in vivo [20,21]. The protective mechanisms employed by nnAbs depend on the engagement with effector cells and consist of antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and antibody-dependent cell-mediated phagocytosis (ADCP). ADCP also assists in the elimination of virus-infected cells and protects against influenza virus infection in a mouse model [22]. Importantly, these nnAbs can be induced by vaccination in humans [18,20,23]. Jegaskanda et al. showed that ADCC-mediated antibodies (ADCC-Abs) can be induced from monovalent inactivated influenza A(H1N1) virus vaccine, high titers of ADCC-Abs can contribute to lower virus replication and reduced clinical symptoms [20]. Zhong and colleagues demonstrated that both ADCC-Abs and neutralizing antibodies can be elicited by influenza vaccination [23].
The mechanism of the antibody-triggered Fc-effector functions is well understood. The antibody Fc region interacts with the FcγRIIIa (CD16a) on the surface of the Fc receptor-bearing cells, primarily natural killer cells (NK cells). After the antibodies bind to their target viral proteins, the combined interactions activate these effector cells. The C-terminal immunoreceptor tyrosine-based activation motif (ITAM) in effector cells is subsequently activated to stimulate the release of granzyme B and perforin by activating signal-transduction molecules and various pathways, such as calcium-dependent pathways and mitogen-activated protein kinase pathway (originally known as extracellular signal-regulated kinases pathway) [24,25]. Details of the signaling pathways initiated by Fc-Fc receptors (FcRs) have been extensively reviewed by Nimmerjahn et al [26]. Perforins can produce pores in the virus-infected cell surface, allowing the entry of granzyme B that cleaves death substrate to initiate apoptosis [27]. Together, these facilitate the elimination of influenza virus-infected cells [28]. This Fc-receptor binding activity was also confirmed to confer protection in animal models in the absence of neutralizing antibodies [29].
Multiple parameters impact the potency of Fc-effector functions. FcRs expression pattern in these cells affects the potency of Fc-effector functions. FcRs are divided into activating and inhibitory receptors based on whether the receptor-mediated signal activates the ITAM or the opposing immunoreceptor tyrosine-based inhibitory motif (ITIM). The magnitude of NK cell responses critically relies on the integration of signals transduced by ITAM and ITIM [30]. Depending on the antibody isotype, these binding receptors can differ. FcγR, FcεR, and FcαR can recognize Fc region of IgG, IgE, and IgA, respectively [31]. FcγRs are the most well-studied FcRs. Different FcγRs can vary significantly from antibody binding affinity to ADCC induction [32]. In human FcRs, FcγRI(CD64), FcγRIIa(CD32a), FcγRIIc(CD32c), and FcγRIIIa(CD16a) are activating receptors, while FcγRIIb (CD32b) is the inhibitory receptor [33]. It is worth noting that no inhibitory receptor is present on NK cells’ surface, only the activating receptor FcγRIIIa is expressed [26]. Without the intervention of FcγRIIb as an inhibitory receptor to negatively regulate cell activation, NK cells serve as the predominant cells that can mediate potent ADCC responses in vivo [28].
Although mouse FcγRs and their counterparts in humans are named similarly, they may differ substantially in their expression pattern, antibody subclass binding affinities and in vivo dominant FcγRs [29,33]. Some FcγRs, such as FcγRIIa, FcγRIIc, and FcγRIIIb, are expressed in human immune cells but not in mice cells [34] and FcγRIV is expressed in mice immune cells but not in human cells [35]. Therefore, a transgenic mouse model consistently expressing human FcRs is needed when investigating human antibody-mediated Fc-effector functions and the human FcγR contribution to antiviral response. Smith et al. generated a mouse model without any murine FcγRs and human FcγRs genes were inserted into the mouse genome to express functional human FcγRs [36]. These human FcγRs were validated to mediate the cytotoxic effects of human IgG antibodies, providing a valuable platform for therapeutic IgG antibodies studies [29].
Most therapeutic antibodies in development are IgG [37]. IgG subclasses (IgG1, IgG2, IgG3, and IgG4 in humans) exhibited different binding affinities to FcγRs as they differ in primary sequence and the associated structure. Of the four IgG subclasses, IgG1, IgG2, and IgG4 are used clinically for therapeutic purposes [38], while the extensive application of IgG3 may be hindered by the its short serum half-life [39,40]. Both IgG1 and IgG3 can mediate potent ADCC activity, while IgG2 and IgG4 have a poor ADCC function [41].
Lastly, antibody glycosylation is essential for maintaining antibody structural conformation which is directly related to the potency of Fc-effector functions [37,42]. Many factors can affect glycosylation patterns, which include age, pregnancy and virus infection [43,44,45]. However, the detailed mechanism of how antibody glycosylation impact Fc-effector functions remains unclear. It has been demonstrated that the reduced content of fucose can enhance ADCC activity [46]. Various approaches, such as alpha-(1,6)-fucosyltransferase gene knockout, have been developed to manipulate antibody fucosylation to optimize ADCC activity [47]. Glyco-engineering and protein engineering have emerged as the two main strategies to harness ADCC activity [48].
Antibodies mediating Fc-effector activities were isolated and characterized in numerous studies, which suggest that these antibodies can play a critical role in tumor clearance [25] and immunity to multiple pathogens such as human immunodeficiency virus (HIV) [49,50], Ebola [51], dengue virus (DENV) [52], West Nile virus (WNV) [53] and influenza virus [16,17]. HIV infection remains a health issue on a global scale and vaccine candidates were developed to address this deteriorating issue. Currently, only one HIV vaccine, RV144, shows modest efficacy (31% efficacy) and this efficacy is not associated with neutralizing antibodies but with potent ADCC-Abs [54,55,56,57,58].

2. Antibodies Induce ADCC Activities through Diverse Innate Effector Cells

As Fc-FcγR interactions are required to induce ADCC, effector cells must express FcγRs on membrane surface to facilitate Fc binding. The NK cell is not the only cell type that has the FcγRIIIa on the membrane, other Fc receptor-bearing cells, like neutrophils, monocytes/macrophages, granulocytes, and dendritic cells (DCs) may also play a crucial role in mediating ADCC or ADCP for viral infected cell clearance [26,59,60]. Broadly neutralizing mAbs targeting HA-stalk can induce ADCP by neutrophils, which indicate the important role of neutrophils in viral clearance [61,62]. Ackerman et al. described a high-throughput flow cytometry-based assay to measure the phagocytic activity by using a monocytic cell line [63]. In addition, Horner et al. reported the critical role of polymorphonuclear granulocyte in mediating ADCC activity in combating malignant diseases [64].

3. In Vitro Assays to Assess Antibody-Induced ADCC Activities

Since the discovery of the ADCC phenomenon in 1965 by Erna Moeller [65], multiple assays have been developed to measure ADCC activity for different purposes and are extensively employed in vaccine study and drug discovery. To measure ADCC activities in vitro, virus-infected target cells, antibodies, and effector cells are needed. The various assays differ in the type of effector cells used or the way to measure the cytotoxicity, which have contributed to high variability among the assays. Considering the complex mechanism of ADCC, it remains questionable which assays are the most physiologically relevant to predict the in vivo efficacy [56,66].

3.1. 51Cr Release Assay

ADCC activity is measured indirectly by the release of 51Chromium(51Cr) following lysis of virus-infected target cells in early studies [67,68,69]. In this assay, the target cells are labeled with 51Cr and subsequently incubated with the appropriate antibody. The quantity of 51Cr released from the lysed cells is detected by a gamma counter as a correlate of ADCC activity [69]. However, the biohazard problem of radioisotopes plus the test’s low sensitivity restricts the wide application of this assay. For these reasons, nonradioactive assays with better sensitivity are later developed to assess ADCC activities [70].

3.2. Target Cell Killing Assay

Flow cytometry-based ADCC assays are developed to measure ADCC activity by using different fluorescent dyes to differentiate living and dead cells [70,71]. In this assay, PKH-67 is used to label both live and dead target cells while dead target cells and effector cells are labeled with 7-amino actinomycin D (7-AAD). Instead of measuring reporter molecules released from target cells, this assay directly measures the lysed target cells. The quantification of lysed target cells is determined by measuring the double fluorescent dyes positive cells by flow cytometry. Target cell death in the absence of antibody is used as a background control. In 1990, Radosevic et al. reported their ADCC assay using different fluorochromes for cell staining with similar principles [72]. Compared to other ADCC measurement assays, this flow cytometry-based assay has a number of advantages such as simplicity, sensitivity, and accessibility, which gains it popularity in many laboratories [72].

3.3. NK Cell Activation Assay

NK cell activation assay is employed extensively to evaluate ADCC activities in vitro due to its high-throughput and precision [16,29,57,73,74]. In this flow cytometry-based assay, recombinant proteins or virus-infected cells are coated in plates followed by the addition of antibody and peripheral blood mononuclear cells (or NK cell lines). Lysosomal-associated membrane protein-1 (LAMP-1 or CD107a) is a cell marker expressed on the NK surface and is employed in this assay to identify NK cells’ functional activities as the expression level of CD107a is highly correlated with cytokine secretion from activated NK cells [75,76]. Therefore, the potency of ADCC activities can be determined indirectly by measuring the activation marker CD107a expression on the NK cell surface. This assay has been widely used to assess ADCC activities evoked by antibodies against the influenza virus and HIV [77,78].

3.4. ADCC Reporter Bioassay

This bioluminescent reporter assay is designed to determine ADCC activities by the measurement of firefly luciferase expression in effector cells [79]. A modified Jurkat stable cell line expressing FcγRIIIa is used as effector cells and expresses firefly luciferase when activated by nuclear factor of activated T cells (NFAT)-response element during the time when FcγRs are bound by Fc portion of the antibody. This cell line is developed in a simple thaw-and-use format which reduces the high inter-assay variability and greatly increases the throughput. This commercially available assay has been employed to quantify ADCC activities against influenza infections [17,80,81,82,83,84].

4. ADCC Induced by anti-HA Antibodies

4.1. HA Stalk-Targeting mAbs Can Mediate Robust ADCC Activities

The HA stalk region, in contrast to the HA head region, is less immunogenic. The antigenic subdominance of HA stalk region may be due to the steric shielding of HA head domain, making it less accessible to the humoral immunity [85]. Therefore, antibodies against the stalk region are not readily stimulated and detected in natural infection of humans. Nevertheless, low levels of stalk-specific B cells are detected in influenza patients [86]. Furthermore, anti-stalk antibodies with broad neutralizing activity have been isolated and characterized [87,88,89,90,91,92]. Since the stalk domain is more conserved than the head domain, these antibodies exhibit broad-spectrum binding and are capable of conferring heterosubtypic protection by interfering with the membrane fusion. HA stalk antibodies were shown to neutralize subtypes in the same group or cross both group 1 and group 2 [93,94]. Ekiert et al. reported that HA stalk-binding human antibody, CR6261, neutralizes multiple influenza subtypes from group 1 [95]. Corti et al. found that one antibody generated from single human plasma targeting fusion sub-domain of the HA stalk could bind and neutralize influenza A viruses from both group 1 and group 2 [94]. Dreyfus et al. reported an HA stalk-binding antibody, CR9114, broad binding to influenza A and B HAs and the antibody was able to confer protection against not only influenza A virus infection, but also influenza B virus infection in mice [89]. It should be noted that some antibodies targeting the stalk region are not highly potent when compared to those against the head region. This deficiency should be addressed in future studies.
Through inhibiting conformational changes essential to the membrane fusion, HA stalk-binding antibodies can disarm influenza virus infectivity [96]. Headless HA and chimeric HA are the two strategies to elicit HA stalk-reactive antibodies [86]. These stalk-reactive antibodies exhibit higher protective potency in vivo than in vitro since these antibodies may be capable of engaging FcγR-bearing cells to confer Fc-dependent protection [97]. Vries et al. showed that anti-H1 stalk ADCC-antibody can be detected after the immunization with a chimeric HA consisting of an H1 stalk domain and an irrelevant head domain [21]. The incorporation of two anti-viral mechanisms has attracted substantial attention in influenza vaccine design. Those HA stalk vaccine candidates were tested in human clinical trials [98,99]. Unfortunately, clinical outcomes of HA stalk-based vaccines are not encouraging so further clinical study was recently discontinued, which emphasizes a critical need to discover novel vaccines that are capable of inducing potent cross-subtype protective antibodies with ADCC activity (https://www.fiercebiotech.com/biotech/gsk-dumps-universal-flu-vaccine-after-interim-data-readout).

4.2. HI-Negative Head-Targeting mAbs Can Mediate Robust ADCC Activities

The anti-stalk antibodies can induce ADCC activities through engaging FcγRs on NK cells, but the role of anti-HA head mAbs in inducing ADCC remains unclear. Dilillo et al. used an FcγR knock-out mice model to show that only anti-HA stalk mAbs can bind to FcγRs and induce ADCC potently whereas anti-HA head mAbs inefficiently induce ADCC activity due to poor engagement of FcγRs on the NK cell surface [29]. Other research groups also reported on these findings independently [73,83]. Interestingly, the examination of anti-HA head mAbs for mediating ADCC in different studies achieved distinct results. Recent studies suggested that some HA-head reactive antibodies (HI-negative) could also engage FcγRIIIa on NK cells and mediate ADCC activity (Figure 2).
Firstly, epitopes in the vestigial esterase domain on HA head have also shown some promise as ADCC-Ab binding targets (Figure 2A). Vestigial esterase domain-binding antibodies were reported to function mainly by blocking viral egress similar to NA inhibitors [89,101]. Recent studies indicated that vestigial esterase domain-binding antibodies also elicit ADCC that may help to protect against viral infection [102]. Bangaru and colleagues found a monoclonal antibody, H3v-47, that not only can neutralize diverse H3N2 viruses and block progeny viruses release, but also induce potent ADCC activity, suggesting that Fc-effector functions are critical for both HA head and stalk targeting antibodies [102]. Interestingly, these researchers found that this multifunctional antibody can recognize a novel epitope that spans the vestigial esterase and receptor-binding subdomains. Similar Fc-mediated ADCC activity was observed in mAb 46B8 that targets the vestigial esterase domain of HA head of the influenza B virus [74]. In addition, the vestigial esterase domain is highly conserved, indicating a potential role in universal vaccine design.
Secondly, antibodies targeting epitopes in the trimer interface may also initiate Fc-effector activities. Watanabe et al. reported that antibody (S5V2-29) binding to a conserved epitope at the HA head trimer interface can also confer potent protection in vivo by activating FcγR-dependent effector activities [103]. Bangaru et al. discovered a broadly protective antibody, FluA-20, which recognized a well-conserved epitope occluded in the trimer interface and also exhibited robust Fc-mediated ADCC in vitro [104]. However, the ADCC may not be essential to the protective role in mice. Historically, it is believed that the trimeric HA protein is so stable that the trimer interface may not be exposed to the host immune system and is likely unreachable by antibody but, contrarily, the epitopes identified by these researchers suggest that the trimer interface may be accessible, at least partially. As such, the trimer interface can be further explored as a potential target for vaccine and therapeutics.
Thirdly, some HA-head reactive antibodies with undefined epitopes were reported to confer protection in vivo via Fc-effector functions. DiLillio et al. demonstrated that the broadly neutralizing HA head-reactive antibodies, 4G05 and 1F05, can induce protection in vivo through Fc-FcγR interactions whereas strain-specific HA head reactive antibodies confer protection in an Fc-FcγR interactions-independent manner in vivo [100]. In addition, anti-H4 monoclonal antibodies binding to the HA head showed high cross-reactivity to avian and mammalian H4 HAs and protected mice from lethal H4N6 challenges through ADCC activity [81]. Two highly conserved epitopes, designated E1 and E2, located in HA head of a pandemic H1N1 virus, were reported to be recognized by ADCC-Ab [70]. The discovery of HA head reactive antibodies and conserved epitopes may assist the future design of universal vaccines and therapeutics.

4.3. HI-Positive Head-Targeting mAbs Cannot Mediate Robust ADCC Activities

As stated above, HA stalk-targeted monoclonal antibodies and HA head-targeted antibodies with no detectable HI titer may evoke potent ADCC activities both in vitro and in vivo. However, HI-positive antibodies that block RBS are commonly demonstrated not to induce robust ADCC activities [82,83,105]. This failure in inducing potent ADCC activity by HI-positive antibodies might be explained by a model proposed by Leon and colleagues [105]. In this model, two synapses between the effector cell and the virus-infected cells are essential for a robust ADCC response (Figure 3A,B). The binding between Fc and FcγRIIIa serves as the first synapse and the interaction between viral HA head and the SA on the effector cell forms the second synapse. These two synapses cooperatively reinforce the connection between the target cell and the effector cell, which mediate robust ADCC activity. The Fc-FcγRIIIa interaction alone is insufficient to induce potent ADCC activity and the interaction of HA to SA on the effector cell is required to induce potent ADCC function [82,83,105]. However, it remains obscure whether the SA-binding of HA activates a signaling pathway to induce ADCC or stabilizes the interaction between antibody and innate immune leukocytes [105]. HI-positive antibodies may compete with the SA on the effector cell in binding to the HA head domain, which as a result may impair the second synapse and hence lead to the reduced ADCC activity eventually (Figure 3C). It was reported that the in vitro ADCC activity induced by a stalk-reactive antibody is diminished after the addition of a HI-positive antibody [83,105]. The blocked or diminished ADCC activity may be due to the competitive role of neutralizing (HI-positive) antibody in binding to the HA. Vaccination can induce both neutralizing mAbs and nnAbs, future vaccine design should consider this delicate balance to achieve the optimal vaccine efficacy.
It is worth noting that neutralization and ADCC are not mutually exclusive. Antibodies with both anti-viral mechanisms have also been identified and characterized in recent studies, which demonstrated that ADCC activity could occur concurrently with a neutralization response [23,106]. Shen et al. reported that one HA head-targeted antibody, C12G6, can not only neutralize Yamagata, Victoria and earlier lineages of influenza B viruses, but also inhibits the membrane fusion, virus egress, and induces an ADCC response [106]. Notably, they also demonstrated that the potency of C12G6-mediated ADCC activity against different viruses followed an opposite trend to its HI activity, in which higher HI titer was found to be associated with relatively lower ADCC activity, and vice versa.
Figure 3. Two synapses between effector cell and virus-infected cell are essential for robust ADCC activities. The interaction between Fc and FcγRIIIa severs as the first synapse and the interaction between viral HA head and the sialic acid on effector cell form the second synapse, which are indispensable for inducing potent ADCC activities (A,B). The HI-positive antibody may interfere with the interaction between viral HA head and the sialic acid on effector cell, leading to weak ADCC activity (C). HI assay was extensively performed to determine the effects of antibodies in blocking the receptor engagement and replication of the viruses. However, this evaluation may be challenged by some emerging influenza viruses that lack the ability to agglutinate red blood cells of various animal origins [107]. In this scenario, the cell-based virus-neutralization assay would be suitable for the characterization of RBS binding antibodies.
Figure 3. Two synapses between effector cell and virus-infected cell are essential for robust ADCC activities. The interaction between Fc and FcγRIIIa severs as the first synapse and the interaction between viral HA head and the sialic acid on effector cell form the second synapse, which are indispensable for inducing potent ADCC activities (A,B). The HI-positive antibody may interfere with the interaction between viral HA head and the sialic acid on effector cell, leading to weak ADCC activity (C). HI assay was extensively performed to determine the effects of antibodies in blocking the receptor engagement and replication of the viruses. However, this evaluation may be challenged by some emerging influenza viruses that lack the ability to agglutinate red blood cells of various animal origins [107]. In this scenario, the cell-based virus-neutralization assay would be suitable for the characterization of RBS binding antibodies.
Viruses 12 00276 g003

5. ADCC to Other Proteins in Influenza Virus

Three influenza proteins are expressed on the viral membrane; HA, NA, and the Matrix 2 protein (M2) and are targets for ADCC-Abs (Figure 4). Although still critical in the viral life cycle, compared to HA, NA and M2 proteins have relatively lower expression on the viral surface and are not heavily targeted by the immune response. For this reason, NA and M2 are similarly less immunogenic and have a slower antigenic drift rate. This makes them attractive vaccine targets and recently more studies have been exploring this avenue [108,109,110].
The NA functions to cleave host SA receptors in the viral life cycle allowing the release of nascent viruses and NA-targeted mAbs have shown a protective effect in animal models by blocking this release mechanism [111]. Antibodies targeting NA are induced at lower level than antibodies targeting HA after vaccination, which may be due to the relatively less expression of NA on infected cells [112]. Some NA-targeted antibodies require Fc-FcγR interactions to induce protection [18,112]. DiLillio et al. reported that the broadly neutralizing NA-reactive antibodies, 3C05, can induce protective immunity in vivo in an FcγR-dependent manner [100]. Recently, Stadlbauer et al. showed that one monoclonal antibody generated from plasmablasts isolated from a human donor naturally infected with the H3N2 virus, designated 1G01, showed the broadest reactive spectrum to a variety of neuraminidases ranging from influenza A group 1 and group 2, to both influenza B virus lineages [113]. These antibodies exhibited multiple anti-viral activities including the blockade of virus particle release and ADCC. The combination of two anti-viral mechanisms contributed to the broad protection from lethal challenges in vivo by these antibodies targeting neuraminidase antigen.
The M2 conducts the proton flux into the virion in the process of virus entry and equilibrates pH across the trans-Golgi network in the viral maturation stage [114,115]. Response against epitopes from the ectodomain of M2 (M2e) has been shown to induce protective immunity in mice [116]. M2e raises the hope for the universal vaccine because of its highly conserved sequence and antibodies targeting M2e with broad protective effects in animal models [117,118]. M2e vaccine candidates with universal vaccine potential are tested in clinical studies [109]. It is worth noting that mAbs targeting the M2e are believed to possess Fc-receptor dependent functions, like ADCC or ADCP [109,119,120,121]. Song et al. generated an M2e-targeted mAb (Z3G1) without detectable neutralizing activity. Interestingly, the Z3G1 antibody was found to be able to induce potent ADCC and CDC activities in vitro as well as demonstrated protective effect in vivo [122]. Bakkouri et al. further demonstrated the essential role of Fc receptors in anti-M2e antibody-induced protection in mice [117]. These studies indicate that M2e-based vaccines evoke cross-protective responses in an FcγR-dependent manner. Generation and characterization of mAbs against M2e can provide critical insights for the development of novel therapeutics against influenza virus infection.
In addition to those proteins expressed on the viral surface, influenza nucleoprotein (NP) possesses highly conserved regions and is also detected on influenza virus-infected host cells surface [123,124]. This makes NP reachable by antibody and substantial efforts have been focused on the antiviral effects of anti-NP mAbs. The host immune response against NP may participate in multi-mechanisms against influenza infection. Cytotoxic T lymphocytes (CTL) can kill target cells via the recognition of NP antigen peptides presented by MHC-I molecules [125,126]. Berthoud et al. reported a T cell-inducing vaccine based on NP and M1, was safe and immunogenic in older adults [127]. These internal viral proteins have not only shown beneficial by inducing host CTL response, but also can serve as a target for protective mAbs. Jegaskanda et al. demonstrated that mAbs targeting M1 and NP can also elicit ADCC activity and these mAbs can be induced by influenza vaccination and natural infection in children [20,128]. However, the protective role of anti-NP ADCC-Abs has not been investigated in animal models. Since internal viral proteins are more conserved, these ADCC-Abs may be considered to be included in future universal vaccine design.

6. ADCC-Based Vaccine

To tackle the threat posed by influenza viruses, developing a universal vaccine that can induce broad immune response against both seasonal drift strains and emerging strains with pandemic potential is of heightening priority of influenza research. Considerable effort has been focused on the development of therapeutic agents and universal vaccines against influenza virus infection in recent decades. Protective antibodies that bind to conserved epitopes in the HA head, HA stalk, NA, NP, M2e have been isolated and characterized in the vaccine research. Most influenza vaccinology studies aim to induce a high titer of broad neutralizing antibodies (bnAbs), which are capable of conferring broad and durable protection against influenza virus infection, but nnAbs are less investigated. However, it is difficult to induce bnAbs through natural infection and the effectiveness of bnAbs was compromised after the emergence of mutant viruses. Therefore, nnAb has become an alternative option and numerous studies have shown the different protective mechanisms of nnAbs and bnAbs [16,20,129]. Although no neutralizing activities can be detected in vitro, nnAbs are competent to mediate Fc-dependent protection in vivo by eliciting Fc-dependent effector activities. The protective property exhibited by nnAb seems to make an equally potential candidate for universal vaccines and therapeutics against influenza infection. Therefore, Fc-dependent effector activities should be considered and recognized when we evaluate the protective effects of influenza therapeutic agents and vaccine candidates.
The ADCC activity serves as a protective mechanism of HA stalk-binding antibodies and ADCC-Abs also exhibit much greater cross-reactivity than classic neutralizing antibodies, defining a promising direction to universal influenza vaccines. Various animal models are used to assess the protective capacity of ADCC based vaccines, including mouse, ferret, and non-human primates. Florek et al. reported that the ADCC-Abs induced by vaccination are capable of inhibiting influenza virus shedding in cynomolgus macaques [130]. A study focusing on severe human infection cases in China and Australia suggested that antibodies from severe influenza survivors were more likely to exhibit potent ADCC activity, which highlights critical role of ADCC in combating influenza virus infections [131].
ADCC represents a promising strategy for vaccine development, but there are still several questions to be addressed. First, the pre-existing neutralizing antibodies may affect the efficacy of the ADCC-based vaccine. He et al reported that the HI-positive antibody can inhibit the potency of ADCC activity induced by HA stalk-specific antibodies by competing in HA binding [83]. Therefore, the efficacy of the ADCC-based vaccine may be affected by pre-existing immunity. Besides, the activation of pre-existing B cells may induce class switching from IgG to non-ADCC isotypes [132]. Second, vaccines that elicit antibodies targeting the M2 or the internal proteins may confer protection against severe influenza diseases, however, may not prevent viral infection since these antibodies mainly recognize proteins expressed on the infected cell membrane [99]. Third, whether sufficient ADCC-Abs can be induced by vaccination is unclear. The antigen needs to be carefully designed and inclusion of an adjuvant is also critical to protective outcome. Four, there are reports of antibody-dependent enhancement (ADE) in ADCC-mediating antibodies [71,133]. In these studies, ADCC epitopes were identified and these epitope-based vaccines were generated and tested in mice. The immunized animals with ADCC epitopes showed robust ADCC activities but had lower survival rates in comparison with the control group. In addition, it was shown that ADCC epitope-based vaccines might have side effects including extra tissue damage as seen in the lungs, which was caused by the high level of cytotoxic granules (perforin) induced by ADCC activation [71].

7. Strategies to Augment ADCC Activity

Augmenting ADCC activity is a novel research direction in cancer research. Target cell killing activities triggered by ADCC mAbs can be substantially increased by the addition of ADCC-promoting agents, which has attracted substantial attention in cancer research [134,135]. Means of augmentation of these responses in cancer research might shed light on influenza research as well. Different strategies for enhancing ADCC were developed and employed in clinical trials in recent studies.
Considering Fc- FcγRIII interaction is required to elicit ADCC activities, the potency of ADCC can be improved by strengthening this interaction. By selecting Fc variants with optimized Fc-FcγRIII affinity, antibodies can elicit a more robust activating signal to the effector cells [59]. Lazar et al. report that Fc variant S239D/I332E has the capacity to optimize Fc-FcγR interaction and enhance effector function both in vitro and in vivo [136].
Other strategies have been attempted to promote the secretion of cytokines by activating effector cells. Reovirus and toll-like receptor (TLR) agonists can increase NK-mediated ADCC by facilitating cytokines secretion [137]. TLR agonists can significantly enhance ADCC by increasing the percentage of activated NK cells. It has been reported that CpG-containing oligodeoxynucleotides (CpG ODN), TLR9 agonists, have the effect of promoting the cytokines secretion from an activated NK cell [138].

8. Conclusions and Future Directions

The protective efficacy of influenza vaccines largely depends on the stimulation of neutralizing antibodies. For a long period of time, vaccine efficacy has been evaluated by HI titers of neutralizing antibodies, which correlate fairly well with traditional views on vaccine-conferred protection. Recently it is becoming increasingly apparent that nnAbs also play a critical role in combating influenza infection even though in vitro antiviral activity is not detected in traditional assays [16,17,18]. Therefore, future evaluation of vaccine efficacy should take Fc-dependent antiviral functions into consideration to optimize vaccine efficacy. In this review, we have summarized multiple anti-viral mechanisms of monoclonal antibodies with ADCC function and reveal that ADCC can overlap substantially with other mechanisms to confer heterosubtypic protection against seasonal and pandemic influenza. The characterization of identified nnAbs constitutes a novel pathway that mayraise hope for the design of universal vaccines. However, there are still questions about ADCC-Abs need to be addressed.
(1)
As mAbs mediated Fc-effector activities are epitope dependent, identifying epitopes that can elicit nnAbs will serve as a good start for the rational design of universal vaccines and therapeutics. Some studies show that ADCC-Abs recognize linear epitopes [70]. Arunkumar et al. demonstrated that antibodies recognizing conformational epitopes exhibit better protection compared to the antibodies targeting linear epitopes [17]. Further investigation should be focused on the elucidation of which epitope type can induce a more potent ADCC response and as a result provide better protection.
(2)
ADCC-Abs may also exert immune pressure on the virus, resulting in escape mutants that evade antibody recognition similar to what is seen with traditional neutralizing antibodies. No influenza studies have reported ADCC-antibody escape mutants currently, but Chung et al. demonstrated the critical amino acids in the linear epitope could mutate to escape ADCC responses in HIV-1 [139]. Lee et al. summarized two potential mechanisms employed by HIV-1 to resist ADCC responses, i. by directly reducing envelope antigen expression on the infected cell surface and ii. by restricting the exposure of epitopes on the envelope to reduce antibody binding [56,140,141]. Similar or different scenarios may present in influenza, which may need further exploration in future studies.
(3)
It remains unclear about the viral replication step(s) in which ADCC occurs. It is indicated that ADCC activity does not intervene in viral entry and fusion and there is a high possibility that ADCC may target the virus-infected cell, which has HA, or other virus protein expressed [142]. This should be better investigated.
(4)
Designing antibodies with enhanced capacity to induce ADCC activities against influenza infection should be pursued vigorously. As stated above, new approaches are being developed to augment ADCC in cancer research. These studies in cancer research may provide valuable insights into influenza research towards developing better vaccines or therapeutics to improve influenza patient outcomes. Therefore, it would be beneficial to measure the ADCC potency threshold for protective immunity. However, it is worth noting that a more potent ADCC response may not guarantee better protection. Arunkumar et at. reported that two non-neutralizing antibodies, KL-BHA-9B9 and KL-BHA-4C10, exhibited uncorrelated ADCC activity with in vivo protection [17]. KL-BHA-9B9 had potent ADCC activity in vitro but poor protection in vivo, whereas KL-BHA-4C10 conferred strong protection in vivo but had almost undetectable ADCC in vitro. A better understanding of ADCC activation may facilitate the mechanistic elucidation of this phenomenon as well as other variabilities emerged in current assays.
(5)
Although diverse assays have been developed to measure ADCC activity in vitro, it is still obscure to what extent these assays can correlate to the efficacy in vivo. It is challenging to predict ADCC-Ab efficacy in humans. The transgenic mouse model expresses human FcγRs may better evaluate the antibody therapeutic effect in humans. However, mice are not natural hosts of influenza and influenza viruses causing human epidemics or pandemics typically replicate inefficiently in this animal model unless adapted viruses are used. Non-human primate models such as rhesus macaques or cynomolgus macaques are used in ADCC-Abs research and research findings from this model collectively indicated that ADCC-Abs are associated with reduced shedding of influenza viruses [16,130]. Considering the different expression patterns of FcγRs in humans and non-human primate animals, it remains uncertain whether the correlation of treatment outcome can be expected in humans. In this regard, clinical studies regarding ADCC-Abs are instrumental to examine the potential role of ADCC-Abs in combating influenza virus infection in humans [143].
(6)
It would be interesting and worthwhile to study the synergy between neutralizing antibody and non-neutralizing antibody in vivo since both classes of antibodies will be elicited after viral infection. The high complexity of each type of antibodies and the lack of an ideal animal model make this investigation relatively challenging. In some cases, the Fc-effector functions induced by nnAb can increase the potency of neutralizing antibodies [14]. In contrast, it is also revealed that Fc-effector functions may not play any role when there is a high dose of neutralizing antibodies [29]. One possible explanation for this is that the function elicited by neutralizing antibodies, such as viral entry blocking or fusion disruption, is potent and sufficient for in vivo protection alone.
(7)
The interplay among ADCC-Abs targeting HA, NA, M2e and NP needs to be investigated. It is critical to identify what antibody combination patterns can induce synergistic protection to augment vaccine efficacy. It is reported that the NA-inhibiting antibody elicited weak ADCC response alone, but it may cooperate with antibodies targeting the HA stem to enhance their potency of ADCC activity [83]. NA inhibitors can enhance Fc-dependent functions induced by anti-stalk antibody, suggesting a therapeutic synergy between NA inhibitors and anti-stalk antibody in humans [144]. Deng et al. demonstrated that the vaccination with double-layered nanoparticles containing HA-stalk and M2e induced potent long-lasting immunity and full protection against divergent influenza A viruses challenge in mice, indicating that the combination of antibodies targeting HA stalk domain and M2e can improve the potency and breadth of vaccine-induced protection [145]. The cocktail of therapeutic antibodies may overcome the emergence of viral escape mutants [37]. Further studies on antibody or inhibitor interplay may guide the better design of future universal anti-influenza countermeasures with broad-spectrum protection via multiple mechanisms.

Author Contributions

Concept and design: D.W. and F.L.; literature search and writing: R.G. and C.C.S.; illustrations: R.G., Z.S. and D.W.; supervision and editing: D.W. and F.L.; final approval of the version submitted: all the authors. All authors have read and agreed to the published version of the manuscript.

Funding

This work was partially supported by NIH grant R01AI141889, by South Dakota State University agricultural experiment station (AES) 3AH-477, by National Science Foundation/EPSCoR (http://www.nsf.gov/od/iia/programs/epscor/index.jsp) award IIA1335423, and by the state of South Dakota’s Governor’s Office of Economic Development as a South Dakota Research Innovation Center.

Acknowledgments

We thank Ian Hauffe for proofreading this manuscript and other members of the Li and Wang laboratories for their input into this work.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. WHO. Up to 650,000 People Die of Respiratory Diseases Linked to Seasonal Flu Each Year; WHO: Ginevra, Switzerland, 2017. [Google Scholar]
  2. Bouvier, N.M.; Palese, P. The biology of influenza viruses. Vaccine 2008, 26 (Suppl. 4), D49–D53. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Hause, B.M.; Collin, E.A.; Liu, R.; Huang, B.; Sheng, Z.; Lu, W.; Wang, D.; Nelson, E.A.; Li, F. Characterization of a novel influenza virus in cattle and Swine: Proposal for a new genus in the Orthomyxoviridae family. MBio 2014, 5, e00031-14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Hause, B.M.; Ducatez, M.; Collin, E.A.; Ran, Z.; Liu, R.; Sheng, Z.; Armien, A.; Kaplan, B.; Chakravarty, S.; Hoppe, A.D.; et al. Isolation of a novel swine influenza virus from Oklahoma in 2011 which is distantly related to human influenza C viruses. PLoS Pathog. 2013, 9, e1003176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Shtyrya, Y.A.; Mochalova, L.V.; Bovin, N.V. Influenza virus neuraminidase: Structure and function. Acta Nat. 2009, 1, 26–32. [Google Scholar] [CrossRef] [Green Version]
  6. Tong, S.; Zhu, X.; Li, Y.; Shi, M.; Zhang, J.; Bourgeois, M.; Yang, H.; Chen, X.; Recuenco, S.; Gomez, J.; et al. New world bats harbor diverse influenza A viruses. PLoS Pathog. 2013, 9, e1003657. [Google Scholar] [CrossRef] [Green Version]
  7. CDC; Weekly, U.S. Influenza Surveillance Report; Key Updates for Week 7, Ending 15 February; CDC: Atlanta, GA, USA, 2020.
  8. Houser, K.; Subbarao, K. Influenza vaccines: Challenges and solutions. Cell Host Microbe 2015, 17, 295–300. [Google Scholar] [CrossRef] [Green Version]
  9. Grohskopf, L.A.; Alyanak, E.; Broder, K.R.; Walter, E.B.; Fry, A.M.; Jernigan, D.B. Prevention and Control of Seasonal Influenza with Vaccines: Recommendations of the Advisory Committee on Immunization Practices-United States, 2019–2020 Influenza Season. MMWR Recomm. Rep. 2019, 68, 1–21. [Google Scholar] [CrossRef] [Green Version]
  10. Margine, I.; Krammer, F.; Hai, R.; Heaton, N.S.; Tan, G.S.; Andrews, S.A.; Runstadler, J.A.; Wilson, P.C.; Albrecht, R.A.; Garcia-Sastre, A.; et al. Hemagglutinin stalk-based universal vaccine constructs protect against group 2 influenza A viruses. J. Virol. 2013, 87, 10435–10446. [Google Scholar] [CrossRef] [Green Version]
  11. Krammer, F.; Pica, N.; Hai, R.; Margine, I.; Palese, P. Chimeric hemagglutinin influenza virus vaccine constructs elicit broadly protective stalk-specific antibodies. J. Virol. 2013, 87, 6542–6550. [Google Scholar] [CrossRef] [Green Version]
  12. Giles, B.M.; Ross, T.M. A computationally optimized broadly reactive antigen (COBRA) based H5N1 VLP vaccine elicits broadly reactive antibodies in mice and ferrets. Vaccine 2011, 29, 3043–3054. [Google Scholar] [CrossRef] [Green Version]
  13. Carter, D.M.; Darby, C.A.; Lefoley, B.C.; Crevar, C.J.; Alefantis, T.; Oomen, R.; Anderson, S.F.; Strugnell, T.; Cortes-Garcia, G.; Vogel, T.U.; et al. Design and Characterization of a Computationally Optimized Broadly Reactive Hemagglutinin Vaccine for H1N1 Influenza Viruses. J. Virol. 2016, 90, 4720–4734. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Jegaskanda, S.; Reading, P.C.; Kent, S.J. Influenza-specific antibody-dependent cellular cytotoxicity: Toward a universal influenza vaccine. J. Immunol. 2014, 193, 469–475. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Von Holle, T.A.; Moody, M.A. Influenza and Antibody-Dependent Cellular Cytotoxicity. Front. Immunol. 2019, 10, 1457. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Jegaskanda, S.; Weinfurter, J.T.; Friedrich, T.C.; Kent, S.J. Antibody-dependent cellular cytotoxicity is associated with control of pandemic H1N1 influenza virus infection of macaques. J. Virol. 2013, 87, 5512–5522. [Google Scholar] [CrossRef] [Green Version]
  17. Asthagiri Arunkumar, G.; Ioannou, A.; Wohlbold, T.J.; Meade, P.; Aslam, S.; Amanat, F.; Ayllon, J.; Garcia-Sastre, A.; Krammer, F. Broadly Cross-Reactive, Nonneutralizing Antibodies against Influenza B Virus Hemagglutinin Demonstrate Effector Function-Dependent Protection against Lethal Viral Challenge in Mice. J. Virol. 2019, 93. [Google Scholar] [CrossRef] [Green Version]
  18. Henry Dunand, C.J.; Leon, P.E.; Huang, M.; Choi, A.; Chromikova, V.; Ho, I.Y.; Tan, G.S.; Cruz, J.; Hirsh, A.; Zheng, N.Y.; et al. Both Neutralizing and Non-Neutralizing Human H7N9 Influenza Vaccine-Induced Monoclonal Antibodies Confer Protection. Cell Host Microbe 2016, 19, 800–813. [Google Scholar] [CrossRef] [Green Version]
  19. Padilla-Quirarte, H.O.; Lopez-Guerrero, D.V.; Gutierrez-Xicotencatl, L.; Esquivel-Guadarrama, F. Protective Antibodies Against Influenza Proteins. Front. Immunol. 2019, 10, 1677. [Google Scholar] [CrossRef] [Green Version]
  20. Jegaskanda, S.; Luke, C.; Hickman, H.D.; Sangster, M.Y.; Wieland-Alter, W.F.; McBride, J.M.; Yewdell, J.W.; Wright, P.F.; Treanor, J.; Rosenberger, C.M.; et al. Generation and Protective Ability of Influenza Virus-Specific Antibody-Dependent Cellular Cytotoxicity in Humans Elicited by Vaccination, Natural Infection, and Experimental Challenge. J. Infect. Dis. 2016, 214, 945–952. [Google Scholar] [CrossRef] [Green Version]
  21. de Vries, R.D.; Nieuwkoop, N.J.; Pronk, M.; de Bruin, E.; Leroux-Roels, G.; Huijskens, E.G.W.; van Binnendijk, R.S.; Krammer, F.; Koopmans, M.P.G.; Rimmelzwaan, G.F. Influenza virus-specific antibody dependent cellular cytoxicity induced by vaccination or natural infection. Vaccine 2017, 35, 238–247. [Google Scholar] [CrossRef]
  22. Huber, V.C.; Lynch, J.M.; Bucher, D.J.; Le, J.; Metzger, D.W. Fc receptor-mediated phagocytosis makes a significant contribution to clearance of influenza virus infections. J. Immunol. 2001, 166, 7381–7388. [Google Scholar] [CrossRef] [Green Version]
  23. Zhong, W.; Liu, F.; Wilson, J.R.; Holiday, C.; Li, Z.N.; Bai, Y.; Tzeng, W.P.; Stevens, J.; York, I.A.; Levine, M.Z. Antibody-Dependent Cell-Mediated Cytotoxicity to Hemagglutinin of Influenza A Viruses After Influenza Vaccination in Humans. Open Forum Infect. Dis. 2016, 3, 102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Liao, F.; Shin, H.S.; Rhee, S.G. Tyrosine phosphorylation of phospholipase C-gamma 1 induced by cross-linking of the high-affinity or low-affinity Fc receptor for IgG in U937 cells. Proc. Natl. Acad. Sci. USA 1992, 89, 3659–3663. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Wang, W.; Erbe, A.K.; Hank, J.A.; Morris, Z.S.; Sondel, P.M. NK Cell-Mediated Antibody-Dependent Cellular Cytotoxicity in Cancer Immunotherapy. Front. Immunol. 2015, 6, 368. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Nimmerjahn, F.; Ravetch, J.V. Fcgamma receptors as regulators of immune responses. Nat. Rev. Immunol. 2008, 8, 34–47. [Google Scholar] [CrossRef]
  27. Salk, H.M.; Haralambieva, I.H.; Ovsyannikova, I.G.; Goergen, K.M.; Poland, G.A. Granzyme B ELISPOT assay to measure influenza-specific cellular immunity. J. Immunol. Methods 2013, 398, 44–50. [Google Scholar] [CrossRef] [Green Version]
  28. Ravetch, J.V.; Bolland, S. IgG Fc receptors. Annu. Rev. Immunol. 2001, 19, 275–290. [Google Scholar] [CrossRef]
  29. DiLillo, D.J.; Tan, G.S.; Palese, P.; Ravetch, J.V. Broadly neutralizing hemagglutinin stalk-specific antibodies require FcgammaR interactions for protection against influenza virus in vivo. Nat. Med. 2014, 20, 143–151. [Google Scholar] [CrossRef]
  30. Weiner, L.M.; Surana, R.; Wang, S. Monoclonal antibodies: Versatile platforms for cancer immunotherapy. Nat. Rev. Immunol. 2010, 10, 317–327. [Google Scholar] [CrossRef] [Green Version]
  31. Schroeder, H.W., Jr.; Cavacini, L. Structure and function of immunoglobulins. J. Allergy Clin. Immunol. 2010, 125, S41–S52. [Google Scholar] [CrossRef] [Green Version]
  32. Anand, S.P.; Finzi, A. Understudied Factors Influencing Fc-Mediated Immune Responses against Viral Infections. Vaccines 2019, 7, 103. [Google Scholar] [CrossRef] [Green Version]
  33. Bruhns, P. Properties of mouse and human IgG receptors and their contribution to disease models. Blood 2012, 119, 5640–5649. [Google Scholar] [CrossRef] [PubMed]
  34. Ravetch, J.V.; Kinet, J.P. Fc receptors. Annu. Rev. Immunol. 1991, 9, 457–492. [Google Scholar] [CrossRef] [PubMed]
  35. Nimmerjahn, F.; Bruhns, P.; Horiuchi, K.; Ravetch, J.V. FcgammaRIV: A novel FcR with distinct IgG subclass specificity. Immunity 2005, 23, 41–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Smith, P.; DiLillo, D.J.; Bournazos, S.; Li, F.; Ravetch, J.V. Mouse model recapitulating human Fcgamma receptor structural and functional diversity. Proc. Natl. Acad. Sci. USA 2012, 109, 6181–6186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Irani, V.; Guy, A.J.; Andrew, D.; Beeson, J.G.; Ramsland, P.A.; Richards, J.S. Molecular properties of human IgG subclasses and their implications for designing therapeutic monoclonal antibodies against infectious diseases. Mol. Immunol. 2015, 67, 171–182. [Google Scholar] [CrossRef] [PubMed]
  38. Wu, M.; Rivkin, A.; Pham, T. Panitumumab: Human monoclonal antibody against epidermal growth factor receptors for the treatment of metastatic colorectal cancer. Clin. Ther. 2008, 30, 14–30. [Google Scholar] [CrossRef]
  39. Krammer, F. The human antibody response to influenza A virus infection and vaccination. Nat. Rev. Immunol. 2019, 19, 383–397. [Google Scholar] [CrossRef]
  40. Vidarsson, G.; Dekkers, G.; Rispens, T. IgG subclasses and allotypes: From structure to effector functions. Front. Immunol. 2014, 5, 520. [Google Scholar] [CrossRef] [Green Version]
  41. An, Z.; Forrest, G.; Moore, R.; Cukan, M.; Haytko, P.; Huang, L.; Vitelli, S.; Zhao, J.Z.; Lu, P.; Hua, J.; et al. IgG2m4, an engineered antibody isotype with reduced Fc function. MAbs 2009, 1, 572–579. [Google Scholar] [CrossRef] [Green Version]
  42. Pincetic, A.; Bournazos, S.; DiLillo, D.J.; Maamary, J.; Wang, T.T.; Dahan, R.; Fiebiger, B.M.; Ravetch, J.V. Type I and type II Fc receptors regulate innate and adaptive immunity. Nat. Immunol. 2014, 15, 707–716. [Google Scholar] [CrossRef]
  43. Bondt, A.; Rombouts, Y.; Selman, M.H.; Hensbergen, P.J.; Reiding, K.R.; Hazes, J.M.; Dolhain, R.J.; Wuhrer, M. Immunoglobulin G (IgG) Fab glycosylation analysis using a new mass spectrometric high-throughput profiling method reveals pregnancy-associated changes. Mol. Cell Proteom. 2014, 13, 3029–3039. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Yamada, E.; Tsukamoto, Y.; Sasaki, R.; Yagyu, K.; Takahashi, N. Structural changes of immunoglobulin G oligosaccharides with age in healthy human serum. Glycoconj. J. 1997, 14, 401–405. [Google Scholar] [CrossRef] [PubMed]
  45. Moore, J.S.; Wu, X.; Kulhavy, R.; Tomana, M.; Novak, J.; Moldoveanu, Z.; Brown, R.; Goepfert, P.A.; Mestecky, J. Increased levels of galactose-deficient IgG in sera of HIV-1-infected individuals. AIDS 2005, 19, 381–389. [Google Scholar] [CrossRef] [PubMed]
  46. Shinkawa, T.; Nakamura, K.; Yamane, N.; Shoji-Hosaka, E.; Kanda, Y.; Sakurada, M.; Uchida, K.; Anazawa, H.; Satoh, M.; Yamasaki, M.; et al. The absence of fucose but not the presence of galactose or bisecting N-acetylglucosamine of human IgG1 complex-type oligosaccharides shows the critical role of enhancing antibody-dependent cellular cytotoxicity. J. Biol. Chem. 2003, 278, 3466–3473. [Google Scholar] [CrossRef] [Green Version]
  47. Kellner, C.; Otte, A.; Cappuzzello, E.; Klausz, K.; Peipp, M. Modulating Cytotoxic Effector Functions by Fc Engineering to Improve Cancer Therapy. Transfus. Med. Hemother. 2017, 44, 327–336. [Google Scholar] [CrossRef] [Green Version]
  48. Monnet, C.; Jorieux, S.; Souyris, N.; Zaki, O.; Jacquet, A.; Fournier, N.; Crozet, F.; de Romeuf, C.; Bouayadi, K.; Urbain, R.; et al. Combined glyco- and protein-Fc engineering simultaneously enhance cytotoxicity and half-life of a therapeutic antibody. MAbs 2014, 6, 422–436. [Google Scholar] [CrossRef] [Green Version]
  49. Gomez-Roman, V.R.; Florese, R.H.; Peng, B.; Montefiori, D.C.; Kalyanaraman, V.S.; Venzon, D.; Srivastava, I.; Barnett, S.W.; Robert-Guroff, M. An adenovirus-based HIV subtype B prime/boost vaccine regimen elicits antibodies mediating broad antibody-dependent cellular cytotoxicity against non-subtype B HIV strains. J. Acquir. Immune Defic. Syndr. 2006, 43, 270–277. [Google Scholar] [CrossRef]
  50. Richard, J.; Prevost, J.; Alsahafi, N.; Ding, S.; Finzi, A. Impact of HIV-1 Envelope Conformation on ADCC Responses. Trends Microbiol. 2018, 26, 253–265. [Google Scholar] [CrossRef]
  51. Saphire, E.O.; Schendel, S.L.; Fusco, M.L.; Gangavarapu, K.; Gunn, B.M.; Wec, A.Z.; Halfmann, P.J.; Brannan, J.M.; Herbert, A.S.; Qiu, X.; et al. Systematic Analysis of Monoclonal Antibodies against Ebola Virus GP Defines Features that Contribute to Protection. Cell 2018, 174, 938–952. [Google Scholar] [CrossRef] [Green Version]
  52. Garcia, G.; Arango, M.; Perez, A.B.; Fonte, L.; Sierra, B.; Rodriguez-Roche, R.; Aguirre, E.; Fiterre, I.; Guzman, M.G. Antibodies from patients with dengue viral infection mediate cellular cytotoxicity. J. Clin. Virol. Off. Publ. Pan Am. Soc. Clin. Virol. 2006, 37, 53–57. [Google Scholar] [CrossRef]
  53. Zhang, M.; Daniel, S.; Huang, Y.; Chancey, C.; Huang, Q.; Lei, Y.F.; Grinev, A.; Mostowski, H.; Rios, M.; Dayton, A. Anti-West Nile virus activity of in vitro expanded human primary natural killer cells. BMC Immunol. 2010, 11, 3. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Rerks-Ngarm, S.; Pitisuttithum, P.; Nitayaphan, S.; Kaewkungwal, J.; Chiu, J.; Paris, R.; Premsri, N.; Namwat, C.; de Souza, M.; Adams, E.; et al. Vaccination with ALVAC and AIDSVAX to prevent HIV-1 infection in Thailand. N. Engl. J. Med. 2009, 361, 2209–2220. [Google Scholar] [CrossRef] [PubMed]
  55. Hsu, D.C.; O’Connell, R.J. Progress in HIV vaccine development. Hum. Vaccines Immunother. 2017, 13, 1018–1030. [Google Scholar] [CrossRef]
  56. Lee, W.S.; Kent, S.J. Anti-HIV-1 antibody-dependent cellular cytotoxicity: Is there more to antibodies than neutralization? Curr. Opin. HIV AIDS 2018, 13, 160–166. [Google Scholar] [CrossRef]
  57. Bruel, T.; Guivel-Benhassine, F.; Lorin, V.; Lortat-Jacob, H.; Baleux, F.; Bourdic, K.; Noel, N.; Lambotte, O.; Mouquet, H.; Schwartz, O. Lack of ADCC Breadth of Human Nonneutralizing Anti-HIV-1 Antibodies. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [Green Version]
  58. Chung, A.W.; Ghebremichael, M.; Robinson, H.; Brown, E.; Choi, I.; Lane, S.; Dugast, A.S.; Schoen, M.K.; Rolland, M.; Suscovich, T.J.; et al. Polyfunctional Fc-effector profiles mediated by IgG subclass selection distinguish RV144 and VAX003 vaccines. Sci. Transl. Med. 2014, 6, 228–238. [Google Scholar] [CrossRef] [PubMed]
  59. Rajasekaran, N.; Chester, C.; Yonezawa, A.; Zhao, X.; Kohrt, H.E. Enhancement of antibody-dependent cell mediated cytotoxicity: A new era in cancer treatment. Immuno Targets Ther. 2015, 4, 91–100. [Google Scholar] [CrossRef] [Green Version]
  60. Kramski, M.; Schorcht, A.; Johnston, A.P.; Lichtfuss, G.F.; Jegaskanda, S.; De Rose, R.; Stratov, I.; Kelleher, A.D.; French, M.A.; Center, R.J.; et al. Role of monocytes in mediating HIV-specific antibody-dependent cellular cytotoxicity. J. Immunol. Methods 2012, 384, 51–61. [Google Scholar] [CrossRef] [PubMed]
  61. Mullarkey, C.E.; Bailey, M.J.; Golubeva, D.A.; Tan, G.S.; Nachbagauer, R.; He, W.; Novakowski, K.E.; Bowdish, D.M.; Miller, M.S.; Palese, P. Broadly Neutralizing Hemagglutinin Stalk-Specific Antibodies Induce Potent Phagocytosis of Immune Complexes by Neutrophils in an Fc-Dependent Manner. MBio 2016, 7. [Google Scholar] [CrossRef] [Green Version]
  62. Barker, E.; Reisfeld, R.A. A mechanism for neutrophil-mediated lysis of human neuroblastoma cells. Cancer Res. 1993, 53, 362–367. [Google Scholar]
  63. Ackerman, M.E.; Moldt, B.; Wyatt, R.T.; Dugast, A.S.; McAndrew, E.; Tsoukas, S.; Jost, S.; Berger, C.T.; Sciaranghella, G.; Liu, Q.; et al. A robust, high-throughput assay to determine the phagocytic activity of clinical antibody samples. J. Immunol. Methods 2011, 366, 8–19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Horner, H.; Frank, C.; Dechant, C.; Repp, R.; Glennie, M.; Herrmann, M.; Stockmeyer, B. Intimate cell conjugate formation and exchange of membrane lipids precede apoptosis induction in target cells during antibody-dependent, granulocyte-mediated cytotoxicity. J. Immunol. 2007, 179, 337–345. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Moeller, E. Contact-Induced Cytotoxicity by Lymphoid Cells Containing Foreign Isoantigens. Science 1965, 147, 873–879. [Google Scholar] [CrossRef] [PubMed]
  66. Lewis, G.K.; Ackerman, M.E.; Scarlatti, G.; Moog, C.; Robert-Guroff, M.; Kent, S.J.; Overbaugh, J.; Reeves, R.K.; Ferrari, G.; Thyagarajan, B. Knowns and Unknowns of Assaying Antibody-Dependent Cell-Mediated Cytotoxicity Against HIV-1. Front. Immunol. 2019, 10, 1025. [Google Scholar] [CrossRef] [Green Version]
  67. Tyler, D.S.; Stanley, S.D.; Nastala, C.A.; Austin, A.A.; Bartlett, J.A.; Stine, K.C.; Lyerly, H.K.; Bolognesi, D.P.; Weinhold, K.J. Alterations in antibody-dependent cellular cytotoxicity during the course of HIV-1 infection. Humoral and cellular defects. J. Immunol. 1990, 144, 3375–3384. [Google Scholar]
  68. Baum, L.L.; Cassutt, K.J.; Knigge, K.; Khattri, R.; Margolick, J.; Rinaldo, C.; Kleeberger, C.A.; Nishanian, P.; Henrard, D.R.; Phair, J. HIV-1 gp120-specific antibody-dependent cell-mediated cytotoxicity correlates with rate of disease progression. J. Immunol. 1996, 157, 2168–2173. [Google Scholar]
  69. Kurane, I.; Hebblewaite, D.; Brandt, W.E.; Ennis, F.A. Lysis of dengue virus-infected cells by natural cell-mediated cytotoxicity and antibody-dependent cell-mediated cytotoxicity. J. Virol. 1984, 52, 223–230. [Google Scholar] [CrossRef] [Green Version]
  70. Srivastava, V.; Yang, Z.; Hung, I.F.; Xu, J.; Zheng, B.; Zhang, M.Y. Identification of dominant antibody-dependent cell-mediated cytotoxicity epitopes on the hemagglutinin antigen of pandemic H1N1 influenza virus. J. Virol. 2013, 87, 5831–5840. [Google Scholar] [CrossRef] [Green Version]
  71. Ye, Z.W.; Yuan, S.; Poon, K.M.; Wen, L.; Yang, D.; Sun, Z.; Li, C.; Hu, M.; Shuai, H.; Zhou, J.; et al. Antibody-Dependent Cell-Mediated Cytotoxicity Epitopes on the Hemagglutinin Head Region of Pandemic H1N1 Influenza Virus Play Detrimental Roles in H1N1-Infected Mice. Front. Immunol. 2017, 8, 317. [Google Scholar] [CrossRef] [Green Version]
  72. Radosevic, K.; Garritsen, H.S.; Van Graft, M.; De Grooth, B.G.; Greve, J. A simple and sensitive flow cytometric assay for the determination of the cytotoxic activity of human natural killer cells. J. Immunol. Methods 1990, 135, 81–89. [Google Scholar] [CrossRef] [Green Version]
  73. de Vries, R.D.; Nieuwkoop, N.J.; van der Klis, F.R.M.; Koopmans, M.P.G.; Krammer, F.; Rimmelzwaan, G.F. Primary Human Influenza B Virus Infection Induces Cross-Lineage Hemagglutinin Stalk-Specific Antibodies Mediating Antibody-Dependent Cellular Cytoxicity. J. Infect. Dis. 2017, 217, 3–11. [Google Scholar] [CrossRef]
  74. Chai, N.; Swem, L.R.; Park, S.; Nakamura, G.; Chiang, N.; Estevez, A.; Fong, R.; Kamen, L.; Kho, E.; Reichelt, M.; et al. A broadly protective therapeutic antibody against influenza B virus with two mechanisms of action. Nat. Commun. 2017, 8, 14234. [Google Scholar] [CrossRef]
  75. Alter, G.; Malenfant, J.M.; Altfeld, M. CD107a as a functional marker for the identification of natural killer cell activity. J. Immunol. Methods 2004, 294, 15–22. [Google Scholar] [CrossRef] [PubMed]
  76. Krzewski, K.; Gil-Krzewska, A.; Nguyen, V.; Peruzzi, G.; Coligan, J.E. LAMP1/CD107a is required for efficient perforin delivery to lytic granules and NK-cell cytotoxicity. Blood 2013, 121, 4672–4683. [Google Scholar] [CrossRef] [PubMed]
  77. Chung, A.W.; Navis, M.; Isitman, G.; Wren, L.; Silvers, J.; Amin, J.; Kent, S.J.; Stratov, I. Activation of NK cells by ADCC antibodies and HIV disease progression. J. Acquir. Immune Defic. Syndr. 2011, 58, 127–131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Madhavi, V.; Navis, M.; Chung, A.W.; Isitman, G.; Wren, L.H.; De Rose, R.; Kent, S.J.; Stratov, I. Activation of NK cells by HIV-specific ADCC antibodies: Role for granulocytes in expressing HIV-1 peptide epitopes. Hum. Vaccines Immunother. 2013, 9, 1011–1018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Cheng, Z.J.; Garvin, D.; Paguio, A.; Moravec, R.; Engel, L.; Fan, F.; Surowy, T. Development of a robust reporter-based ADCC assay with frozen, thaw-and-use cells to measure Fc effector function of therapeutic antibodies. J. Immunol. Methods 2014, 414, 69–81. [Google Scholar] [CrossRef]
  80. Tan, G.S.; Leon, P.E.; Albrecht, R.A.; Margine, I.; Hirsh, A.; Bahl, J.; Krammer, F. Broadly-Reactive Neutralizing and Non-neutralizing Antibodies Directed against the H7 Influenza Virus Hemagglutinin Reveal Divergent Mechanisms of Protection. PLoS Pathog. 2016, 12, e1005578. [Google Scholar] [CrossRef]
  81. Amanat, F.; Meade, P.; Strohmeier, S.; Krammer, F. Cross-reactive antibodies binding to H4 hemagglutinin protect against a lethal H4N6 influenza virus challenge in the mouse model. Emerg. Microbes Infect. 2019, 8, 155–168. [Google Scholar] [CrossRef] [Green Version]
  82. Cox, F.; Kwaks, T.; Brandenburg, B.; Koldijk, M.H.; Klaren, V.; Smal, B.; Korse, H.J.; Geelen, E.; Tettero, L.; Zuijdgeest, D.; et al. HA Antibody-Mediated FcgammaRIIIa Activity Is Both Dependent on FcR Engagement and Interactions between HA and Sialic Acids. Front. Immunol. 2016, 7, 399. [Google Scholar] [CrossRef] [Green Version]
  83. He, W.; Tan, G.S.; Mullarkey, C.E.; Lee, A.J.; Lam, M.M.; Krammer, F.; Henry, C.; Wilson, P.C.; Ashkar, A.A.; Palese, P.; et al. Epitope specificity plays a critical role in regulating antibody-dependent cell-mediated cytotoxicity against influenza A virus. Proc. Natl. Acad. Sci. USA 2016, 113, 11931–11936. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Ermler, M.E.; Kirkpatrick, E.; Sun, W.; Hai, R.; Amanat, F.; Chromikova, V.; Palese, P.; Krammer, F. Chimeric Hemagglutinin Constructs Induce Broad Protection against Influenza B Virus Challenge in the Mouse Model. J. Virol. 2017, 91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Andrews, S.F.; Huang, Y.; Kaur, K.; Popova, L.I.; Ho, I.Y.; Pauli, N.T.; Henry Dunand, C.J.; Taylor, W.M.; Lim, S.; Huang, M.; et al. Immune history profoundly affects broadly protective B cell responses to influenza. Sci. Transl. Med. 2015, 7, 316ra192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Krammer, F.; Palese, P. Influenza virus hemagglutinin stalk-based antibodies and vaccines. Curr. Opin. Virol. 2013, 3, 521–530. [Google Scholar] [CrossRef] [Green Version]
  87. Tan, G.S.; Krammer, F.; Eggink, D.; Kongchanagul, A.; Moran, T.M.; Palese, P. A pan-H1 anti-hemagglutinin monoclonal antibody with potent broad-spectrum efficacy in vivo. J. Virol. 2012, 86, 6179–6188. [Google Scholar] [CrossRef] [Green Version]
  88. Desbien, A.L.; Van Hoeven, N.; Reed, S.J.; Casey, A.C.; Laurance, J.D.; Baldwin, S.L.; Duthie, M.S.; Reed, S.G.; Carter, D. Development of a high density hemagglutinin protein microarray to determine the breadth of influenza antibody responses. Biotechniques 2013, 54, 345–348. [Google Scholar] [CrossRef]
  89. Dreyfus, C.; Laursen, N.S.; Kwaks, T.; Zuijdgeest, D.; Khayat, R.; Ekiert, D.C.; Lee, J.H.; Metlagel, Z.; Bujny, M.V.; Jongeneelen, M.; et al. Highly conserved protective epitopes on influenza B viruses. Science 2012, 337, 1343–1348. [Google Scholar] [CrossRef] [Green Version]
  90. Ekiert, D.C.; Friesen, R.H.; Bhabha, G.; Kwaks, T.; Jongeneelen, M.; Yu, W.; Ophorst, C.; Cox, F.; Korse, H.J.; Brandenburg, B.; et al. A highly conserved neutralizing epitope on group 2 influenza A viruses. Science 2011, 333, 843–850. [Google Scholar] [CrossRef] [Green Version]
  91. Wang, T.T.; Tan, G.S.; Hai, R.; Pica, N.; Petersen, E.; Moran, T.M.; Palese, P. Broadly protective monoclonal antibodies against H3 influenza viruses following sequential immunization with different hemagglutinins. PLoS Pathog. 2010, 6, e1000796. [Google Scholar] [CrossRef] [Green Version]
  92. Yasugi, M.; Kubota-Koketsu, R.; Yamashita, A.; Kawashita, N.; Du, A.; Sasaki, T.; Nishimura, M.; Misaki, R.; Kuhara, M.; Boonsathorn, N.; et al. Human monoclonal antibodies broadly neutralizing against influenza B virus. PLoS Pathog. 2013, 9, e1003150. [Google Scholar] [CrossRef] [Green Version]
  93. Lang, S.; Xie, J.; Zhu, X.; Wu, N.C.; Lerner, R.A.; Wilson, I.A. Antibody 27F3 Broadly Targets Influenza A Group 1 and 2 Hemagglutinins through a Further Variation in VH1-69 Antibody Orientation on the HA Stem. Cell Rep. 2017, 20, 2935–2943. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Corti, D.; Voss, J.; Gamblin, S.J.; Codoni, G.; Macagno, A.; Jarrossay, D.; Vachieri, S.G.; Pinna, D.; Minola, A.; Vanzetta, F.; et al. A neutralizing antibody selected from plasma cells that binds to group 1 and group 2 influenza A hemagglutinins. Science 2011, 333, 850–856. [Google Scholar] [CrossRef] [PubMed]
  95. Ekiert, D.C.; Bhabha, G.; Elsliger, M.A.; Friesen, R.H.; Jongeneelen, M.; Throsby, M.; Goudsmit, J.; Wilson, I.A. Antibody recognition of a highly conserved influenza virus epitope. Science 2009, 324, 246–251. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Laursen, N.S.; Wilson, I.A. Broadly neutralizing antibodies against influenza viruses. Antivir. Res. 2013, 98, 476–483. [Google Scholar] [CrossRef] [Green Version]
  97. Krammer, F. Novel universal influenza virus vaccine approaches. Curr. Opin. Virol. 2016, 17, 95–103. [Google Scholar] [CrossRef] [Green Version]
  98. Nachbagauer, R.; Kinzler, D.; Choi, A.; Hirsh, A.; Beaulieu, E.; Lecrenier, N.; Innis, B.L.; Palese, P.; Mallett, C.P.; Krammer, F. A chimeric haemagglutinin-based influenza split virion vaccine adjuvanted with AS03 induces protective stalk-reactive antibodies in mice. NPJ Vaccines 2016, 1, 1–10. [Google Scholar] [CrossRef]
  99. Nachbagauer, R.; Krammer, F. Universal influenza virus vaccines and therapeutic antibodies. Clin. Microbiol. Infect. 2017, 23, 222–228. [Google Scholar] [CrossRef] [Green Version]
  100. DiLillo, D.J.; Palese, P.; Wilson, P.C.; Ravetch, J.V. Broadly neutralizing anti-influenza antibodies require Fc receptor engagement for in vivo protection. J. Clin. Investig. 2016, 126, 605–610. [Google Scholar] [CrossRef]
  101. Brandenburg, B.; Koudstaal, W.; Goudsmit, J.; Klaren, V.; Tang, C.; Bujny, M.V.; Korse, H.J.; Kwaks, T.; Otterstrom, J.J.; Juraszek, J.; et al. Mechanisms of hemagglutinin targeted influenza virus neutralization. PLoS ONE 2013, 8, e80034. [Google Scholar] [CrossRef] [Green Version]
  102. Bangaru, S.; Zhang, H.; Gilchuk, I.M.; Voss, T.G.; Irving, R.P.; Gilchuk, P.; Matta, P.; Zhu, X.; Lang, S.; Nieusma, T.; et al. A multifunctional human monoclonal neutralizing antibody that targets a unique conserved epitope on influenza HA. Nat. Commun. 2018, 9, 2669. [Google Scholar] [CrossRef] [Green Version]
  103. Watanabe, A.; McCarthy, K.R.; Kuraoka, M.; Schmidt, A.G.; Adachi, Y.; Onodera, T.; Tonouchi, K.; Caradonna, T.M.; Bajic, G.; Song, S.; et al. Antibodies to a Conserved Influenza Head Interface Epitope Protect by an IgG Subtype-Dependent Mechanism. Cell 2019, 177, 1124–1135. [Google Scholar] [CrossRef]
  104. Bangaru, S.; Lang, S.; Schotsaert, M.; Vanderven, H.A.; Zhu, X.; Kose, N.; Bombardi, R.; Finn, J.A.; Kent, S.J.; Gilchuk, P.; et al. A Site of Vulnerability on the Influenza Virus Hemagglutinin Head Domain Trimer Interface. Cell 2019, 177, 1136–1152. [Google Scholar] [CrossRef] [PubMed]
  105. Leon, P.E.; He, W.; Mullarkey, C.E.; Bailey, M.J.; Miller, M.S.; Krammer, F.; Palese, P.; Tan, G.S. Optimal activation of Fc-mediated effector functions by influenza virus hemagglutinin antibodies requires two points of contact. Proc. Natl. Acad. Sci. USA 2016, 113, E5944–E5951. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Shen, C.; Chen, J.; Li, R.; Zhang, M.; Wang, G.; Stegalkina, S.; Zhang, L.; Chen, J.; Cao, J.; Bi, X.; et al. A multimechanistic antibody targeting the receptor binding site potently cross-protects against influenza B viruses. Sci. Transl. Med. 2017, 9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Mostafa, A.; Pleschka, S. Influenza H3N2 Vaccines: Recent Challenges. Trends Microbiol. 2018, 26, 87–89. [Google Scholar] [CrossRef] [PubMed]
  108. Neirynck, S.; Deroo, T.; Saelens, X.; Vanlandschoot, P.; Jou, W.M.; Fiers, W. A universal influenza A vaccine based on the extracellular domain of the M2 protein. Nat. Med. 1999, 5, 1157–1163. [Google Scholar] [CrossRef]
  109. Schotsaert, M.; De Filette, M.; Fiers, W.; Saelens, X. Universal M2 ectodomain-based influenza A vaccines: Preclinical and clinical developments. Expert Rev. Vaccines 2009, 8, 499–508. [Google Scholar] [CrossRef] [Green Version]
  110. Eichelberger, M.C.; Monto, A.S. Neuraminidase, the Forgotten Surface Antigen, Emerges as an Influenza Vaccine Target for Broadened Protection. J. Infect. Dis. 2019, 219, S75–S80. [Google Scholar] [CrossRef] [Green Version]
  111. Wohlbold, T.J.; Nachbagauer, R.; Xu, H.; Tan, G.S.; Hirsh, A.; Brokstad, K.A.; Cox, R.J.; Palese, P.; Krammer, F. Vaccination with adjuvanted recombinant neuraminidase induces broad heterologous, but not heterosubtypic, cross-protection against influenza virus infection in mice. MBio 2015, 6, e02556. [Google Scholar] [CrossRef] [Green Version]
  112. Wohlbold, T.J.; Krammer, F. In the shadow of hemagglutinin: A growing interest in influenza viral neuraminidase and its role as a vaccine antigen. Viruses 2014, 6, 2465–2494. [Google Scholar] [CrossRef]
  113. Stadlbauer, D.; Zhu, X.; McMahon, M.; Turner, J.S.; Wohlbold, T.J.; Schmitz, A.J.; Strohmeier, S.; Yu, W.; Nachbagauer, R.; Mudd, P.A.; et al. Broadly protective human antibodies that target the active site of influenza virus neuraminidase. Science 2019, 366, 499–504. [Google Scholar] [CrossRef] [PubMed]
  114. Cady, S.D.; Luo, W.; Hu, F.; Hong, M. Structure and function of the influenza A M2 proton channel. Biochemistry 2009, 48, 7356–7364. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Pielak, R.M.; Chou, J.J. Influenza M2 proton channels. Biochim. Biophys. Acta 2011, 1808, 522–529. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Zhou, C.; Zhou, L.; Chen, Y.H. Immunization with high epitope density of M2e derived from 2009 pandemic H1N1 elicits protective immunity in mice. Vaccine 2012, 30, 3463–3469. [Google Scholar] [CrossRef] [PubMed]
  117. El Bakkouri, K.; Descamps, F.; De Filette, M.; Smet, A.; Festjens, E.; Birkett, A.; Van Rooijen, N.; Verbeek, S.; Fiers, W.; Saelens, X. Universal vaccine based on ectodomain of matrix protein 2 of influenza A: Fc receptors and alveolar macrophages mediate protection. J. Immunol. 2011, 186, 1022–1031. [Google Scholar] [CrossRef] [Green Version]
  118. Lee, Y.T.; Kim, K.H.; Ko, E.J.; Lee, Y.N.; Kim, M.C.; Kwon, Y.M.; Tang, Y.; Cho, M.K.; Lee, Y.J.; Kang, S.M. New vaccines against influenza virus. Clin. Exp. Vaccine Res. 2014, 3, 12–28. [Google Scholar] [CrossRef] [Green Version]
  119. Schepens, B.; De Vlieger, D.; Saelens, X. Vaccine options for influenza: Thinking small. Curr. Opin. Immunol. 2018, 53, 22–29. [Google Scholar] [CrossRef]
  120. Lee, Y.N.; Lee, Y.T.; Kim, M.C.; Hwang, H.S.; Lee, J.S.; Kim, K.H.; Kang, S.M. Fc receptor is not required for inducing antibodies but plays a critical role in conferring protection after influenza M2 vaccination. Immunology 2014, 143, 300–309. [Google Scholar] [CrossRef]
  121. Kolpe, A.; Schepens, B.; Fiers, W.; Saelens, X. M2-based influenza vaccines: Recent advances and clinical potential. Expert Rev. Vaccines 2017, 16, 123–136. [Google Scholar] [CrossRef]
  122. Song, A.; Myojo, K.; Laudenslager, J.; Harada, D.; Miura, T.; Suzuki, K.; Kuni-Kamochi, R.; Soloff, R.; Ohgami, K.; Kanda, Y. Evaluation of a fully human monoclonal antibody against multiple influenza A viral strains in mice and a pandemic H1N1 strain in nonhuman primates. Antivir. Res. 2014, 111, 60–68. [Google Scholar] [CrossRef]
  123. Virelizier, J.L.; Allison, A.C.; Oxford, J.S.; Schild, G.C. Early presence of ribonucleoprotein antigen on surface of influenza virus-infected cells. Nature 1977, 266, 52–54. [Google Scholar] [CrossRef] [PubMed]
  124. Yewdell, J.W.; Frank, E.; Gerhard, W. Expression of influenza A virus internal antigens on the surface of infected P815 cells. J. Immunol. 1981, 126, 1814–1819. [Google Scholar]
  125. Antrobus, R.D.; Lillie, P.J.; Berthoud, T.K.; Spencer, A.J.; McLaren, J.E.; Ladell, K.; Lambe, T.; Milicic, A.; Price, D.A.; Hill, A.V.; et al. A T cell-inducing influenza vaccine for the elderly: Safety and immunogenicity of MVA-NP+M1 in adults aged over 50 years. PLoS ONE 2012, 7, e48322. [Google Scholar] [CrossRef]
  126. Zheng, M.; Luo, J.; Chen, Z. Development of universal influenza vaccines based on influenza virus M and NP genes. Infection 2014, 42, 251–262. [Google Scholar] [CrossRef] [PubMed]
  127. Berthoud, T.K.; Hamill, M.; Lillie, P.J.; Hwenda, L.; Collins, K.A.; Ewer, K.J.; Milicic, A.; Poyntz, H.C.; Lambe, T.; Fletcher, H.A.; et al. Potent CD8+ T-cell immunogenicity in humans of a novel heterosubtypic influenza A vaccine, MVA-NP+M1. Clin. Infect. Dis. 2011, 52, 1–7. [Google Scholar] [CrossRef] [PubMed]
  128. Jegaskanda, S.; Co, M.D.T.; Cruz, J.; Subbarao, K.; Ennis, F.A.; Terajima, M. Induction of H7N9-Cross-Reactive Antibody-Dependent Cellular Cytotoxicity Antibodies by Human Seasonal Influenza A Viruses that are Directed Toward the Nucleoprotein. J. Infect. Dis. 2017, 215, 818–823. [Google Scholar] [CrossRef]
  129. Carragher, D.M.; Kaminski, D.A.; Moquin, A.; Hartson, L.; Randall, T.D. A novel role for non-neutralizing antibodies against nucleoprotein in facilitating resistance to influenza virus. J. Immunol. 2008, 181, 4168–4176. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Florek, N.W.; Weinfurter, J.T.; Jegaskanda, S.; Brewoo, J.N.; Powell, T.D.; Young, G.R.; Das, S.C.; Hatta, M.; Broman, K.W.; Hungnes, O.; et al. Modified vaccinia virus Ankara encoding influenza virus hemagglutinin induces heterosubtypic immunity in macaques. J. Virol. 2014, 88, 13418–13428. [Google Scholar] [CrossRef] [Green Version]
  131. Vanderven, H.A.; Liu, L.; Ana-Sosa-Batiz, F.; Nguyen, T.H.; Wan, Y.; Wines, B.; Hogarth, P.M.; Tilmanis, D.; Reynaldi, A.; Parsons, M.S.; et al. Fc functional antibodies in humans with severe H7N9 and seasonal influenza. JCI Insight 2017, 2. [Google Scholar] [CrossRef]
  132. Horns, F.; Vollmers, C.; Croote, D.; Mackey, S.F.; Swan, G.E.; Dekker, C.L.; Davis, M.M.; Quake, S.R. Lineage tracing of human B cells reveals the in vivo landscape of human antibody class switching. Elife 2016, 5, e16578. [Google Scholar] [CrossRef]
  133. Wang, J.; Liu, M.; Ding, N.; Li, Y.; Shao, J.; Zhu, M.; Xie, Z.; Sun, K. Vaccine based on antibody-dependent cell-mediated cytotoxicity epitope on the H1N1 influenza virus increases mortality in vaccinated mice. Biochem. Biophys. Res. Commun. 2018, 503, 1874–1879. [Google Scholar] [CrossRef] [PubMed]
  134. Nakajima, T.; Okayama, H.; Ashizawa, M.; Noda, M.; Aoto, K.; Saito, M.; Monma, T.; Ohki, S.; Shibata, M.; Takenoshita, S.; et al. Augmentation of antibody-dependent cellular cytotoxicity with defucosylated monoclonal antibodies in patients with GI-tract cancer. Oncol. Lett. 2018, 15, 2604–2610. [Google Scholar] [CrossRef] [PubMed]
  135. Kohrt, H.E.; Houot, R.; Marabelle, A.; Cho, H.J.; Osman, K.; Goldstein, M.; Levy, R.; Brody, J. Combination strategies to enhance antitumor ADCC. Immunotherapy 2012, 4, 511–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Lazar, G.A.; Dang, W.; Karki, S.; Vafa, O.; Peng, J.S.; Hyun, L.; Chan, C.; Chung, H.S.; Eivazi, A.; Yoder, S.C.; et al. Engineered antibody Fc variants with enhanced effector function. Proc. Natl. Acad. Sci. USA 2006, 103, 4005–4010. [Google Scholar] [CrossRef] [Green Version]
  137. Ming Lim, C.; Stephenson, R.; Salazar, A.M.; Ferris, R.L. TLR3 agonists improve the immunostimulatory potential of cetuximab against EGFR(+) head and neck cancer cells. Oncoimmunology 2013, 2, e24677. [Google Scholar] [CrossRef] [Green Version]
  138. Roda, J.M.; Parihar, R.; Carson, W.E., 3rd. CpG-containing oligodeoxynucleotides act through TLR9 to enhance the NK cell cytokine response to antibody-coated tumor cells. J. Immunol. 2005, 175, 1619–1627. [Google Scholar] [CrossRef]
  139. Chung, A.W.; Isitman, G.; Navis, M.; Kramski, M.; Center, R.J.; Kent, S.J.; Stratov, I. Immune escape from HIV-specific antibody-dependent cellular cytotoxicity (ADCC) pressure. Proc. Natl. Acad. Sci. USA 2011, 108, 7505–7510. [Google Scholar] [CrossRef] [Green Version]
  140. Smalls-Mantey, A.; Doria-Rose, N.; Klein, R.; Patamawenu, A.; Migueles, S.A.; Ko, S.Y.; Hallahan, C.W.; Wong, H.; Liu, B.; You, L.; et al. Antibody-dependent cellular cytotoxicity against primary HIV-infected CD4+ T cells is directly associated with the magnitude of surface IgG binding. J. Virol. 2012, 86, 8672–8680. [Google Scholar] [CrossRef] [Green Version]
  141. Boge, M.; Wyss, S.; Bonifacino, J.S.; Thali, M. Membrane-proximal tyrosine-based signal mediates internalization of the HIV-1 envelope glycoprotein via interaction with the AP-2 clathrin adaptor. J. Biol. Chem. 1998, 273, 15773–15778. [Google Scholar] [CrossRef] [Green Version]
  142. Jegerlehner, A.; Schmitz, N.; Storni, T.; Bachmann, M.F. Influenza A vaccine based on the extracellular domain of M2: Weak protection mediated via antibody-dependent NK cell activity. J. Immunol. 2004, 172, 5598–5605. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Jegaskanda, S. The Potential Role of Fc-Receptor Functions in the Development of a Universal Influenza Vaccine. Vaccines 2018, 6, 27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Kosik, I.; Angeletti, D.; Gibbs, J.S.; Angel, M.; Takeda, K.; Kosikova, M.; Nair, V.; Hickman, H.D.; Xie, H.; Brooke, C.B.; et al. Neuraminidase inhibition contributes to influenza A virus neutralization by anti-hemagglutinin stem antibodies. J. Exp. Med. 2019, 216, 304–316. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Deng, L.; Mohan, T.; Chang, T.Z.; Gonzalez, G.X.; Wang, Y.; Kwon, Y.M.; Kang, S.M.; Compans, R.W.; Champion, J.A.; Wang, B.Z. Double-layered protein nanoparticles induce broad protection against divergent influenza A viruses. Nat. Commun. 2018, 9, 359. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic view of influenza virion. Hemagglutinin (HA), neuraminidase (NA) and matrix protein 2 (M2) are the proteins expressed on virus membrane. Trimeric HA protein consists of HA head and HA stalk. Viral ribonucleoprotein (vRNP) is composed of vRNA and nucleoprotein (NP, light purple). Viral polymerase complex includes polymerase basic proteins 1 (PB1, blue), 2 (PB2, tan) and acidic protein A (PA, light yellow). The matrix protein (M1) is a multi-functional protein involved in influenza virion assembly and infection.
Figure 1. Schematic view of influenza virion. Hemagglutinin (HA), neuraminidase (NA) and matrix protein 2 (M2) are the proteins expressed on virus membrane. Trimeric HA protein consists of HA head and HA stalk. Viral ribonucleoprotein (vRNP) is composed of vRNA and nucleoprotein (NP, light purple). Viral polymerase complex includes polymerase basic proteins 1 (PB1, blue), 2 (PB2, tan) and acidic protein A (PA, light yellow). The matrix protein (M1) is a multi-functional protein involved in influenza virion assembly and infection.
Viruses 12 00276 g001
Figure 2. Antibody-dependent cellular cytotoxicity (ADCC)-epitopes in HA. Antibodies targeting HA stalk, as well as HA head including esterase domain and trimer interface, can induce potent ADCC in vitro and elicit broad-spectrum protection in vivo. The HA structure is generated using PyMOL. A/California 04/2009 HA (PDB code: 3UBE) is used as a representative HA structure to show the location of ADCC-epitopes. Influenza HA is a trimer with each monomer comprised of HA1(wheat), vestigial esterase domain (orange) and receptor binding domain (pink), and HA2 (palegreen). The remaining two monomers are shown in light blue. Sialic acid is colored cyan. Side view of the HA trimer surface is shown in (A), while the top view of the HA trimer interface is shown in the cartoon (B) and surface (C) modes. Besides, there are ADCC-Abs targeting HA head with unknown epitopes [100].
Figure 2. Antibody-dependent cellular cytotoxicity (ADCC)-epitopes in HA. Antibodies targeting HA stalk, as well as HA head including esterase domain and trimer interface, can induce potent ADCC in vitro and elicit broad-spectrum protection in vivo. The HA structure is generated using PyMOL. A/California 04/2009 HA (PDB code: 3UBE) is used as a representative HA structure to show the location of ADCC-epitopes. Influenza HA is a trimer with each monomer comprised of HA1(wheat), vestigial esterase domain (orange) and receptor binding domain (pink), and HA2 (palegreen). The remaining two monomers are shown in light blue. Sialic acid is colored cyan. Side view of the HA trimer surface is shown in (A), while the top view of the HA trimer interface is shown in the cartoon (B) and surface (C) modes. Besides, there are ADCC-Abs targeting HA head with unknown epitopes [100].
Viruses 12 00276 g002
Figure 4. ADCC- mediating antibodies (ADCC-Abs) targeting different influenza antigens. HA, NA and M2 are the surface-exposed proteins and they are capable of inducing ADCC-Abs. Antibodies targeting the conserved epitopes in these proteins confer broad protection against divergent viruses. NP expressed on the surface of the influenza-infected cell serves as a promising target for ADCC-Abs.
Figure 4. ADCC- mediating antibodies (ADCC-Abs) targeting different influenza antigens. HA, NA and M2 are the surface-exposed proteins and they are capable of inducing ADCC-Abs. Antibodies targeting the conserved epitopes in these proteins confer broad protection against divergent viruses. NP expressed on the surface of the influenza-infected cell serves as a promising target for ADCC-Abs.
Viruses 12 00276 g004

Share and Cite

MDPI and ACS Style

Gao, R.; Sheng, Z.; Sreenivasan, C.C.; Wang, D.; Li, F. Influenza A Virus Antibodies with Antibody-Dependent Cellular Cytotoxicity Function. Viruses 2020, 12, 276. https://doi.org/10.3390/v12030276

AMA Style

Gao R, Sheng Z, Sreenivasan CC, Wang D, Li F. Influenza A Virus Antibodies with Antibody-Dependent Cellular Cytotoxicity Function. Viruses. 2020; 12(3):276. https://doi.org/10.3390/v12030276

Chicago/Turabian Style

Gao, Rongyuan, Zizhang Sheng, Chithra C. Sreenivasan, Dan Wang, and Feng Li. 2020. "Influenza A Virus Antibodies with Antibody-Dependent Cellular Cytotoxicity Function" Viruses 12, no. 3: 276. https://doi.org/10.3390/v12030276

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop