Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Apr 6.
Published in final edited form as: Immunol Cell Biol. 2020 Apr 6;98(4):305–317. doi: 10.1111/imcb.12324

Antibody-mediated complement activation in pathology and protection

Benjamin S Goldberg 1, Margaret E Ackerman 1,2,*
PMCID: PMC7293394  NIHMSID: NIHMS1576117  PMID: 32142167

Abstract

Antibody-dependent complement activity is associated with autoimmune morbidity, but also anti-tumor efficacy. In infectious disease, both recombinant monoclonal antibodies and polyclonal antibodies generated in natural adaptive responses can mediate complement activity to protective, therapeutic, or disease-enhancing effect. Recent advances have contributed to the structural resolution of molecular complexes involved in antibody-mediated complement activation, defining the avid nature of participating interactions, and pointing to how antibody isotype, subclass, hinge flexibility, glycosylation state, amino acid sequence, and the contextual nature of the cognate antigen/epitope are all factors that can determine complement activity through impact on antibody multimerization and subsequent recruitment of C1q. Beyond the efficiency of activation, complement activation products interact with various cell types that mediate immune adherence, trafficking, immune education, and innate functions. Similarly, depending on the anatomical location and extent of activation, complement can support homeostatic restoration or be leveraged by pathogens or neoplasms to enhance infection or promote tumorigenic microenvironments, respectively. Advances in means to suppress complement activation by intravenous immunoglobulin (IVIG), IVIG mimetics, and complement-intervening antibodies represent proven and promising exploratory therapeutic strategies, while antibody engineering has likewise offered frameworks to enhance, eliminate, or isolate complement activation to interrogate in vivo mechanisms of action. Such strategies promise to support the optimization of antibody-based drugs that are able to tackle emerging and difficult-to-treat diseases by improving our understanding of the synergistic and antagonistic relationships between antibody mechanisms mediated by Fc receptors, direct binding, and the products of complement activation.

Keywords: Monoclonal antibody, complement, complement effector functions, therapeutics, cancer, infectious disease, antibody engineering, Antibody-dependent complement activation, Antibody-dependent complement-mediated cytotoxicity

Graphical Abstract

Determinants & Outcomes of Antibody-mediated Complement Activation. This review discusses the factors specific to antibodies, antigenic targets, pathogen defenses, and tissue microenvironments that determine the consequences of complement activation. Furthermore, it explores some approaches to capture these complexities in in vitro assays and screens of antibody-mediated complement functions in host-neoplasm, host-graft/auto-host, and host-pathogen interactions. Image reproduced with permission4

graphic file with name nihms-1576117-f0001.jpg

Introduction

The complement system is an evolutionarily ancient defense network that is distributed nearly ubiquitously throughout host extracellular tissues and intracellularly. This integral innate immune system is comprised of more than thirty plasma proteins and cell surface receptors that collectively enhance or complement the ability of antibodies and innate immune cells to clear microbes and host cellular debris, promote inflammation, and directly disrupt the membrane of microbial pathogens and enveloped viruses1 (Figure 1). Following initiation by one of three canonical mechanisms - the alternative, lectin, or classical pathways - a proteolytic cascade involving zymogens and amplifying feedback loops results in the intermediate outcome of coating or opsonizing the target in complement protein fragments that can interface with both innate and adaptive immune elements, and drive a variety of possible terminal outcomes including target lysis1 (Figure 1a).

Figure 1: Overview of Complement Initiation Pathways & Possible Outcomes.

Figure 1:

Figure reproduced with permission1.

(a) The complement cascade be initiated by three defined pathways, which commonly amplify through the C3 convertase, and terminate in target opsonization and/or membrane disruption. The classical pathway denotes activation of the complement cascade by the multi-functional C1q molecule, where C1q-mediated activation was classically defined by an initial recognition event by C1q of target-bound antibody. The Lectin pathway is initiated when specific polysaccharides are directly recognized by mannose-binding lectin (MBL), collectin-11 (CL-11), or by members of the ficolin family (Fcn). The alternative pathway is generally characterized by low levels of continual activation that is typically tempered by host regulatory proteins to protect from self-inflicted tissue damage. (b) Gradually, the products of this low-level activation build up on surfaces lacking host complement regulatory proteins and reach a crescendo, resulting in (c) exponential amplification and cascade progression leading to (d, e) soluble inflammatory molecule generation, target opsonization, lytic insertion of the membrane attack complex (MAC), destruction, adherence and trafficking, adaptive immune education, and host clearance. Ag: antigen; Ab: antibody; C1q: complement component 1q; C1r/s: complement component 1q-associated serine proteases r and s; MBL: mannose binding lectin; MASP: MBL-associated serine proteases; Fcn: ficolins; CL-11: collectin-11; PAMP: pathogen-associated molecular pattern; FP: factor P (properdin); C3(H2O): hydrolyzed C3; FB: factor B, FD: factor D; RCA: regulators of complement activation; C4: complement component 4; C2: complement component 2; FI: factor I; CR1: complement receptor 1 (CD35); C3dg: complement fragment 3dg; C3aR: C3a receptor; C5aR1 and 2: C5a receptor 1 and 2 (CD88 and C5L2, respectively); CRIg: CR of the immunoglobulin family; MAC: membrane attack complex (C5b-9); TCR: T-cell receptor; BCR: B-cell receptor.

The classical complement cascade represents a powerful link between adaptive antibody-mediated immunity and effectors of innate immunity. In fact, target-coated complement fragments can regulate adaptive effector B and T lymphocyte development via immune complex trafficking and antigen presentation, thus augmenting adaptive responses to achieve sustained and robust immune memory1 (Figure 1d). Despite being the subject of decades of investigation, understanding of the complement system is by no means complete, and novel activities and relationships continue to be discovered and defined for diverse complement molecules and activation byproducts. In particular, as monoclonal antibodies now represent a growing and effective class of clinical intervention, understanding the diverse interactions and outcomes of their interactions with the complement system is of high translational relevance.

Antibody-Mediated Complement Activation (AMCA)

Immune complexes (ICs) formed by antigen-bound IgA, IgG, and/or IgM are able to activate complement2. While IgA and IgM isotypes are inherently high avidity immunoglobulins that can recruit pathway initiation molecules and activate complement upon stable surface binding, we will focus on IgG. In its monomeric state, IgG can exhibit bivalent antigen recognition but can only weakly interact with a single globular head of the C1q hexamer3.

Classical and Lectin pathway initiation molecules C1q and mannose-binding lectin (MBL) or ficolins, respectively, share the structural feature of multivalent recognition limbs. Avid recognition functions to limit non-specific engagement by requiring a high local concentration of danger signals, and to increase the stability of the complex by decreasing the off-rate proportional to the number of target-engaged limbs4, 5. In fact, monovalent binding affinity between these pattern recognition molecules and their targets is negligibly low3, 6, requiring antigen clustering for avid engagement of MBL/ficolins, and antigen-driven antibody clustering for avid C1q engagement. Therefore, the structure of these cascade initiation molecules facilitates sustained cascade initiation upon specific target recognition. While MBL and ficolins recognize IgM7, which is in some sense pre-oligomerized, and carbohydrate components of bacterial and fungal cell walls that are usually present in regularly repeating patterns, the same cannot be said of IgG nor all of its targets, respectively. This puts the onus on antibody clustering on the target surface to facilitate avid C1q binding and efficient activation, suggesting that important factors in antigen selection would include expression level, spatial distribution, and membrane mobility. Important antibody factors (Figure 2a) would include antigen-binding and Fc valency as defined by the intrinsic affinity between Fc and C1q, and between antibody and antigen, as well as spatial aspects affected by epitope placement, angle of binding, and hinge length and flexibility between antigen-binding and C1q-binding (Fc) domains5, 810 (Figure 2b).

Figure 2: Structural Determinants of Antibody-mediated Complement Activation (AMCA).

Figure 2:

(a) IgG1 antibody structure denoting antigen-binding (Fab) and Fc Receptor-binding (Fc) domains and antibody-intrinsic factors known to affect the efficiency of AMCA. Boxed regions indicate the site of interactions with C1q globular heads (orange) and between Fc domains (purple). Amino acid residues known to impact Fc-C1q (orange) or Fc-Fc (purple) interactions in the context of IgG1 numbering are indicated. (b) Structure of C1q globular head (orange) and IgG1 Fc (purple) complex. (c). Structure of Fc-Fc dimer complex. (d). (Left) Top down view of Fc hexamer (purple) interacting with six C1q globular heads (orange). (Right) Side view of C1 complex C1r and C1s (blue). IgG1 Fc, PDB 1HZH. IgG1 Fc and gC1q co-complex, PDB 6FCZ. Cryo-EM reconstruction of C1-IgG1 complex, EMD-4232, reproduced with permission130.

Appreciation of such molecular determinants of classical complement activation has been dramatically refined with insights provided by the structure of the C1-(IgG-Fc)6 hexamer complex formed at the binding surface4, which directed attention to Fc-Fc interactions of adjacently-bound antibodies (Figure 2c). While the formation of IgG hexamers was suggested by the crystal structure of an IgG11, Diebolder et al. illustrated that IgG-RGY, IgG containing three point-mutations – E345R, E430G, and S440Y – in the CH2-CH3 interface, enhanced non-covalent Fc-Fc interactions to the point where IgG spontaneously oligomerized in solution in the absence of antigen, recruiting C1 and resulting in classical complement activation. The fact that IgG oligomerization is fundamental to the efficiency of complement activation explains, in part, the dependency of epitope specificity, and the ability of antibody to cluster antigen into lipid rafts on antibody-mediated activation12, 13. A notable observation regarding epitope specificity’s contribution to differential complement activity is the impact of antibody distance from the cell membrane, where membrane-proximal epitopes appear to be favored9.

Beyond such sophisticated spatial factors, a number of antibody-intrinsic factors are known to impact the ability of a given immunoglobulin to exhibit Antibody-Mediated Complement Activation (AMCA). The four subclasses of IgG (e.g. IgG1, IgG2, etc.), display distinct complement activation profiles. In general, IgG1 and IgG3 are considered to be the most efficient activators, while IgG2 can be weakly activating, and IgG4 is often considered to be somewhat incapable of driving complement activation2. Additionally, IgG Fc domains contain a conserved N-linked glycan in the vicinity of the C1q binding interface, which is required for efficient C1 activation2. Interestingly, the effect of deglycosylation on reduced C1q binding has been linked to the glycans’ role in stabilizing the structure of Fc, and mediate interactions impacting IgG oligomerization rather than a direct impact of glycan on C1q-Fc binding affinity2, 5. Beyond this requirement for glycan, controlled studies of monoclonal antibodies with modified glycosylation profiles have enabled mapping of the impact of specific glycoform compositions on complement activation. These studies have revealed that, whereas Fc-galactosylation enhances the C1q-binding, C4b-fixation, and downstream effects, fucosylation and bisection have no effect, and conflicting results have been reported as to the effect of Fc sialylation1416. Interestingly, glycoforms lacking terminal sialic acid and galactose residues have been shown to bind with higher affinity for lectin pathway initiation molecules17.

While Fc features such as these offer useful rules of thumb for anticipating AMCA, it has been long clear that they are not adequate to accurately predict the function of a given antibody of interest. Overall, the diversity of antibody-intrinsic factors that regulate classical complement activation, from structural and Fc glycosylation differences dictating adjacent Fc-to-Fc association to inherent affinity for antigen, antibody valency, and epitope specificity point to difficulty in faithful assessment of AMCA in vitro, even before subsequent impacts are considered.

Downstream Signal Amplification and Outcomes

The magnitude of activation depends on the balance between pathway activation complexes and complement inhibitory factors. The net magnitude and surface distribution of activation will determine the effector outcome(s). A product of classical initiation, and a commonly used tissue biomarker in autoantibody-mediated pathology diagnoses, C4b is covalently attached to functional groups in the near vicinity of C4 cleavage on the target surface via a reactive thioester exposed by C1q-associated serine proteases C1r and C1s. Bound C4b forms the basis of the enzymatically active C4bC2b complex, known as the classical C3 convertase, which itself acts as a serine protease cleaving plasma C3 into the small pro-inflammatory C3a fragment, and the large reactive C3b fragment that, like C4b, also binds to the target surface (Figure 1a). From here, the alternative C3 convertase (C3bBb) can be formed, representing an important step in signal amplification. In addition to amplification, cascade progression occurs through both C3 convertases, which can serve as a foundation for C5 convertase formation (Figure 1a). Thus, cascade amplification, mainly through C3 convertases but also through C5 convertases, plays the dual functional role of generating potent signaling soluble peptides, anaphylatoxins C3a and C5a, and cascade-progressing target surface-reactive and effector function-mediating complement fragments C3b, C4b, and C5b (Figure 1a).

The soluble C3a and C5a anaphylatoxins interact with cognate G-protein-coupled receptors, C5aR and C3aR, which are primarily expressed by myeloid-derived cells. Importantly, anaphylatoxin C5a acts as a chemoattractant for the recruitment of inflammatory cells (Figure 1a, 1e), and acts on immune effector cells to upregulate activating and downregulate inactivating Fc-receptors, resulting in monocyte infiltration and differentiation into macrophages, and recruited effectors of antibody-dependent cellular cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP)18 (Figure 1d). In this way, therapeutic AMCA can orchestrate an amplification loop by attracting additional innate elements to the site of antibody target recognition. The important functions of anaphylatoxins in host defense are carefully balanced against the consequences of misdirected, excessive, or sustained activation resulting in host tissue damage and exacerbation of undesired inflammatory conditions.

Complement fragments C3b, C4b, and C5b covalently coat target IC or particle surfaces and can mediate a variety of functions (Figure 1d). Several distinct species of cell-bound C3 and C4 can exist (i.e. iC3b, C3dg) depending on the levels of regulatory activity encountered during the cascade and in the post-activation microenvironment. These fragments maintain IC solubility and can mediate the recognition, trafficking, processing, and elimination of ICs through engagement with cognate complement receptors (CRs), including CR1 (CD35), CR2 (CD21), CR3 (CD11b/CD18), CR4 (CD11c/CD18), and complement receptor of immunoglobulin family (CRIg), expressed by a variety of leukocytes (Figure 1a). Given this wide expression profile, and the co-expression of antibody receptors on these cells, it is perhaps not surprising that opsonization can affect pro-inflammatory processes of phagocytosis, antigen presentation, cytokine stimulation, neutrophil extracellular trap expulsion, NK activation, and IC adherence and trafficking (Figure 1d). Studies of the impact of various opsonins on other functions make it clear that a variety of phenotypes can be observed, ranging from competitive19 to synergistic20. Without requiring additional proteolytic steps, fragment C5b can readily recruit downstream complement components, C6 through C8 and multiple copies of C9 yielding lytic – including the membrane attack complex (MAC) (Figure 1e) – as well as sub-lytic terminal complement complexes (TCCs). Sub-lytic TCCs can induce intracellular signaling.

Classical Complement Pathologies and Interventions

Given lytic outcomes, the dysregulation of complement or the generation of autoantibodies that activate complement can lead to serious damage to host tissues and organs. In fact, classical complement C4d fragment is a clinical correlate for the diagnosis of, among other autoimmune disorders, systemic lupus erythematosus (SLE)21, lupus nephritis22, IgA nephropathy23, anti-phospholipid antibody (APLA) syndrome24, and C3-glomerulopathies25, 26, as well as in organ transplant rejection27.

Damage generated by autoantibodies can be treated by B cell depletion (e.g. SLE and RA)28. Alternatively, inhibiting the progression to the lytic end of the complement cascade with the anti-C5 mAb eculizumab has been shown to be a clinically effective strategy in preventing complement-mediated hemolysis in paroxysmal nocturnal hemoglobinuria (PNH) patients29, 30, and in preventing hemolysis leading to thrombotic angiopathy and kidney disease in patients with atypical hemolytic-uremic syndrome (aHUS)31. More recently, eculizumab has been shown to reduce the rate of relapse in patients with the autoantibody anti-aquaporin-4-mediated neuromyelitis optica (NMO) spectrum disorder32. Inhibition by eculizumab can be overridden by strong complement activation in vitro, and lytic complement mechanisms are unlikely the sole contributor to autoantibody-dependent complement-mediated disease states which may explain the differential, incomplete, or lack of therapeutic efficacy seen in other diseases3335. Intuitively, blocking terminal complement may leave patients susceptible to infection36. In fact, late complement component deficiency is strongly associated with increased risk of developing meningococcal disease37 where bactericidal complement antibody is a correlate of protection38.

Nonetheless, the clinical successes of eculizumab portends both the scientific utility of such inhibitors in understanding the precise etiology of diseases in which complement is implicated, and the potential value of exploring other suppression strategies. To this end, additional strategies to suppress complement activity include a small-molecule C5 inhibitor39, a single-chain antibody linked to a complement inhibitor40, siRNA against C541, inhibitory anti-C1s mAb42, C3 inhibition43, a variety of IVIG-mimetic IgG1-Fc multimers4447, and disruption of ICs by relatively complement-inert subclasses such as human IgG448. Beyond canonical complement regulatory factors, a role for a histological biomarker in Alzheimer’s disease and atherosclerosis, apolipoprotein-E (ApoE), in attenuation of unresolvable inflammatory diseases was recently identified. ApoE was shown to selectively complex with activated C1q at picomolar affinity and inhibit its activity41. Lastly, intravenous immunoglobulin (IVIG) has been extensively used in the clinic to suppress inflammation across a range of disorders. In autoimmune neuropathies involving anti-ganglioside antibodies, both polyclonal IgM and IgA49, and IVIG50 were shown to inhibit complement activation in a glycoform-dependent manner51, 52. The major critique of strategies to suppress classical complement activity is that the complement system is critical to the systemic defense, clearance of apoptotic debris, and general maintenance of host homeostasis, such as in tumor surveillance. However, while natural complement deficiencies in humans are associated with increased risk of meningococcal infection that can be reduced by vaccination53, prolonged pharmacologic C3 inhibition does not appear to increase risk of infection in otherwise healthy adult non-human primates43.

Role in tumor promotion & elimination

The complement system can be directly activated by neoplastic host cells, as well as indirectly via antibody opsonization. While demonstrated to be antagonistic of some Fc receptor-mediated functions54, complement dependent cytotoxicity is thought to be a main mechanism of action for some monoclonal therapeutics55. In fact, tumor cell vulnerability to complement-mediated lysis is implicated as a predictor of mAb-based therapeutic efficacy56. However, aberrant complement activation and anaphylatoxin generation has been linked to augmented tumorigenesis57 as demonstrated by tumor inhibition by pharmacological blockade of the C5a receptor58. In chronic inflammation, anaphylatoxins - particularly C5a - can promote a pro-tumor microenvironment through a variety of mechanisms, including chemotactic recruitment of tissue-infiltrating myeloid cells that are stimulated to secrete immunosuppressive molecules thereby sustaining a pro-tumor microenvironment58. Additionally, sub-lytic TCCs can have oncogenic effects, including cell cycle modulation and resistance to apoptosis and complement59. Mechanisms of tumor cell complement evasion include antigenic modulation60 and overexpression of complement regulatory proteins (CRPs), of which CD59 (MAC-inhibitory protein, or protectin) inhibits C9 polymerization and can induce internalization of affected areas of membrane in order to escape MAC lysis61. What is known regarding the interplay between complement and various cancers has been reviewed recently62.

Cancer cell populations are inherently heterogeneous in their susceptibility to classical complement fixation, and can be shielded by tumor microenvironmental factors (i.e. hypoxia, pH, etc.) that can impact the complement cascade63. Nonetheless, targets such as CD20 have established the importance of AMCA as a mechanism of action64, as well as point to epitope-dependent factors impacting efficient membrane clustering and Fc-Fc oligomerization12, 65. In fact, the degree to which anti-CD20 antibodies induce classical activation has raised concerns that exhaustion of plasma levels of complement components might be rate- and efficacy-limiting66. Targeting multiple antigens or epitopes simultaneously can also influence the efficiency of activation67, and in some cases, overcome tumor heterogeneity and plasticity68. These cocktails contribute to cross-linking-dependent Fc-effector function enhancement69, but also increase the effective local concentration of IgG Fcs and their orientation heterogeneity on the target surface, likely influencing the probability of hexameric Fc organization70.

Role in Infectious Disease: from elimination to evasion to enhancement

Natural host immunity is critically reliant upon the complement system to protect and eliminate infections from a number of microorganisms71 and viruses; the classical complement initiation pathway has been demonstrated to contribute to anti-viral activity and protection against West Nile Virus (WNV)72, Zika73, Influenza74, vaccinia virus75, CMV76, RSV77, and HIV78, 79. Additionally, classical complement has been implicated in host defense against the pathogenic bacteria Salmonella71, Gonorrhea80, as well as against parasites8183. However, genetic variation affecting antigen identity and character can dramatically influence the susceptibility of a virus or pathogen to classical complement neutralization or lysis, even in the instance of closely related viruses84. When confronted with an invasive pathogen, it is commonly understood that antibodies can directly interfere with mechanisms of infection (e.g. host cell entry by virions, replication, bacterial quorum-sensing, etc.), usually by steric interference. Similarly, complement opsoninization can theoretically potentiate the ability of an antibody to neutralize pathogens by steric mechanisms, as has been experimentally observed76, 85.

Unfortunately, evolutionary competition with host immunity has equipped infectious agents with the means to evade, and in some cases, even repurpose the complement system to gain a fitness advantage through a variety of mechanisms. Herpes simplex virus type 1 expresses a glycoprotein, gE, that mimics human Fcγ receptor, and has been demonstrated to block human IgG Fc-mediated complement activation86. Streptococcus pyogenes binds IgG from serum, which increases C4b-binding protein (C4BP) recruitment, and enhances bacterial virulence87. Other complement evasion mechanisms include the incorporation of host membrane CRPs by budding enveloped virions88, molecular mimicry89 or recruitment of such host regulatory factors87, 90, and production of other complement-disrupting factors such as complement-cleaving enzymes91, and interference with C1q-associated serine proteases92. Another mechanism of complement evasion, and evasion of antibody-mediated effector function more generally, is antibody-induced antigen internalization on virally-infected host cells93, 94. These pathogen evasion mechanisms, and others, have been extensively reviewed recently95. Understanding such mechanisms of evasion and enhancement of infection offers the prospect of aiding the development and optimization of next-generation vaccines and antibody drugs.

Despite controversy around the existence or degree of infection enhancement influenced by antibody-mediated complement activation, evidence indeed exists for such a phenomenon across a range of infectious agents, including HIV96. Mechanistic explanations of infection enhancement include instances in which host complement regulators directly facilitate entry97, but also sub-lytic opsonization can result in CR-mediated entry directly98 or via phagocytic uptake99. In this way, sub-lytic AMCA can enhance infectivity by increasing the efficiency of intracellular invasion, for example, by transporting infectious ICs to susceptible target cells100. Several bacterial pathogens, such as F. tularensis, exploit CR3-mediated macrophage internalization and downstream anti-inflammatory signaling to achieve intracellular persistence101. Other mechanisms of complement enhanced infection have been reported, but these activities often occur alongside protective mechanisms. In fact, the difference between WNV neutralization and infection-enhancement depends on factors that dictate the degree of complement activation102. This balance between protective and pathogenic outcomes of AMCA represents yet another challenge to in vitro models and in vivo understanding.

Ab engineering

Modulating AMCA, whether beneficial or detrimental to antibody drug efficacy, is a logical strategy to improve antibody drugs. Downstream pathway interventions include inhibition of regulatory factor function, such as of Factor I, a strategy which has been demonstrated to increase the lytic efficiency of rituximab103. Complement regulatory factor-neutralizing and tumor-targeting antibodies have been delivered in mAb cocktails104, as well as combined in bi-specific mAbs105. A novel bi-specific, targeting both CD20 antigen and the C1q globular head, was recently reported that provides a direct means of C1q recruitment that may additionally provide enhanced flexibility and relieve geometric constraints tied to antigen and epitope specificity106. Direct modification of the antibody molecule in bispecific strategies such as this, or more commonly by Fc engineering (Table 1) not only contributes to drug optimization, but has also served as a key tool for determining antibody mechanism of action.

Table 1:

Engineered antibodies and their complement phenotypes

ID Mutations / Alterations Complement Phenotype Sources and Important Citations Comments
C1q-binding CDC potency
Complement Phenotype Reduced LALA L234A, L235A n.d. - - - 125 Xu et al. Cell Immunol. (2000) Significant reduction in ADCC; has been used in vivo in nonhuman primates, 126 Hessell Nature (2007)
127 Hezareh M., et al. J. Virol. (2001)
LALA-PG L234A, L235A, P329G - - - n.d. 128 Schlothauer et al. PEDS (2016) Residual FcgRI binding by SPR. This, and related variants are being used in human clinical trials for complete effector knockout.
KA K322A n.d. - - - CDC potency: 4 Diebolder, C. A., et al. Science (2014); ADCC: 127 Hezereh M. et al. J. Virol. (2001) Significant reduction in ADCC
Glycoform N297Q, aglycosylation - - - - - - 129 Sazinsky S. et al. PNAS (2008); CDC potency: 130 Ugurlar D. et al. Science (2018) n.b. to low affinity FcgRs
Glycoform Sialylation - - - - 15 Quast I. et al., J Clin Invest. (2015);
Enhanced Glycoform Sialylation + + 16 Dekkers G. et al. Front Immunol. (2017)
Glycoform Galactosylation + + 14 Peschke B., et al. Front Immunol. (2017)
IgG Fc-μzTP L309C (IgG1-Fc + IgM tailpiece) 44 Spirig, R. et al., J Immunol. (2018) IVIG mimetic - depletes classical complement pathway components to ameliorate inflammation & autoimmune disorders. Evaluated in mice in immune thrombocytopenia and inflammatory arthritis.
RGY E345R, E430G, S440Y n.d. n.d. 4 Diebolder, C. A., et al. Science (2014) Triple-mutant that forms solution-phase hexamers, recruits and activates C1. Seminal paper in elucidating structure of classical complement activation complex
EG E430G no change ++ 108 de Jong, R. N. et al., PLoS Biology (2016) Point mutation that facilitates IgG1 oligomerization at the target surface, but not in solution at relevant concentration levels.
EFT S267E, H268F, S324T +++ ++ 131 Moore L. et al. mAbs (2010) Ablated ADCC
EFT+AE S267E, H268F, S324T + G236A, I332E n.d. +++ Broad enhancement to low affinity FcgRs
K326A, E333A ++ + 107 Idusogie et al. J Immunol. (2001) Unaltered ADCC
113F IgG1 CH1 & Hinge; IgG3 Fc; part of C-term. CH3 substituted with IgG1 residues; See ref. + + Source - 132 Natsume et al. Cancer Res (2008); CDC potency - 131 Moore L. et al. mAbs (2010) Selection of mutations from paper demonstrating importance of K326 & E333 residues in C1q-IgG interaction. Enhanced B-cell depletion in cynomolgus monkeys.
IgGA IgG1-CH2, IgA1-CH3; See ref. ++ + 133 Kelton W., Georgiou G., et al. Cell Chem. Bio. (2014) Chosen from panel of IgG1/IgG3 chimeras for being the most potent complement activator that retained protein A binding.
Selective/Enhanced IgG G801 K320E, Q386R Glycosylated ++ n.d. 134 Lee C.H., Georgiou G., et al. Nature Immuno. (2017) Residual binding to FcgRI (G801); No detectable FcgR binding (A801). Allows for differentiation between FcR-mediated and CR-mediated cellular cytotoxicity and phagocytosis. Evaluated in mice.
IgG A801 K320E, Q386R Aglycosylated +++ +

Not determined (n.d.)

Fc domains lacking complement binding, and/or other effector functions entirely have been used extensively in in vivo studies in animal models to parse the effects of complement and FcγR functions versus Fab-mediated activities alone, and some modifications are even advancing clinically.

Conversely, other mutations have been explored to increase the efficiency of AMCA activation—both through avidity modulation based on engineering of the Fc-Fc interface and propensity to hexamerize, and through affinity for the C1q globular head (Figure 2b)107. Such potentiating strategies can have the effect of overcoming the action of complement regulatory factors, effectively driving the cascade towards terminal complex formation with direct lytic and signaling impacts. The triple mutant RGY used in the structural studies enhanced Fc-to-Fc affinity to the point where antibody hexamerization occurred in solution, complexing with C1q in the absence of antigen. A point mutation E430G, representing the ‘G’ in the RGY triple mutant, demonstrated enhanced hexamerization on the target surface and not in solution (Table 1). Additionally, this variant was shown to exhibit reduced immunogenicity and possess other traits desirable for biopharmaceutical development108. E430G was shown to convert an anti-EGFR IgG1, 2F8/Zalutumumab, from complement-inert to -activating109. This mutation has been used to demonstrate the contribution of AMCA effector mechanisms to the antitumor and antimicrobial efficacy of specific antibodies80, 109111, and as a means to demonstrate inherent susceptibility of a given target to terminal classical complement functions109. Efficiency-enhancing mutations can result in lysis of targets even in the context of late-complement-component-depleted and C9-deficient serum, perhaps indicating that swift and sustained complement activation is important and sufficient for efficient lysis112, 113. An early stage clinical trial (NCT03576131) of dual non-competing anti-DR5 combination therapy utilizing this avidity-modulating mutation is ongoing111.

Modeling the Contribution of Complement

Collectively, the library of ADCA-modifying Fc mutations is further supported by a suite of in vitro assays with diverse readouts, and animal models useful to evaluation of the contribution of complement activation to pathology or protection driven by antibodies. The detection of complement fixation products has provided clinically-useful biomarkers for numerous autoimmune diseases24, 114 and cancer progression115, as well as in the prognosis of tissue/organ transplant tolerability and post-transplant monitoring27, 116, 117. While detection of complement activation end-products on cells and tissues can be correlated with certain autoimmune disease states, pre-clinical characterization of drug candidates AMCA requires recapitulating the context of natural systems in which interventional AMCA would take place.

Given the physiological complexity and highly interconnected nature of the complement system, it would appear to be difficult to rely on in vitro experimentation as a model of in vivo outcomes. In order to best approximate their biological relevance, the design of in vitro experiments must take into consideration a number of factors. For example, there are individual- and tissue-specific differences in the concentration of complement components. To this end, reconstitution of the classical and alternative pathway by a defined mixture of the individual components might be useful in the standardization of complement assays, and may also reduce artefacts originating from other serum components that vary between serum sources118.

Recapitulating native contexts is likely to produce more relevant in vitro results, but such target presentations and assay readouts are often less amenable to high-throughput screening. A common means to observe complement activation is through the use of C3 fragment-specific monoclonal detection reagents119, which is adaptable to bead-based antigen-multiplexed antibody screening120. The throughput of these methods come at the expense of capturing the functional consequences of complement activation, whether opsonophagocytosis, immune adherence and trafficking, lysis, or others, which can be characterized through lower-throughput specialized assays121. Even specialized assessment of complement functions in vitro may fail to fully capture the clinical context, as they are often performed on lab time-scales in the absence of effector cells, and using susceptible or sensitized targets. In vivo models can substantially increase the clinical relevance when characterizing the varied outcomes of AMCA, or lack thereof.

Animal models have been extensively used to parse the mechanisms of action of pathogenic or protective antibodies with a diversity of genetic knockouts122, 123, human cascade component knock-ins106, and complement-modifying interventions including C5 and/or C3 depletion using cobra venom factors124. While animal models have demonstrated an indispensable role here, species-specific and human allelic differences in each of the many complement factors can also influence outcomes. Once a relevant difference is identified, models can be generated to more closely reflect native interactions. For example, a humanized C1q mouse, in which mouse C1qA, C1qB, and C1qC genes were replaced with chimeric versions containing human globular head domains, has been generated106.

One certainty remains, determining when, where, and how complement matters in disease and antibody-based interventions is complicated. If carefully designed, experiments modulating AMCA in vitro and in vivo can provide unique insights that lead to the identification of druggable targets or assist in clinical translation of antibody drugs.

Conclusions

The complement system is an amazingly complex, potent, and intricately regulated aspect of immunity. In part encouraged by the high-profile therapeutic success of anti-CD20 therapy, the factors controlling activation efficiency are being more deeply explored in the context of infectious diseases and autoimmunity. Continued development of assays, models, novel interventions, and engineered antibodies promise to elucidate how complement-dependent antibody mechanisms contribute to disease neutralization, eradication, and in some cases, enhancement as the diversity of interventions and models that have been engineered to explore, exploit, or suppress antibody-mediated complement activity are deployed across diverse disease states.

ACKNOWLEDGMENTS

This work was supported by the National Institute of General Medical Sciences and National Institute of Allergy and Infectious Disease under R01 AI131975 and by the National Institute of Allergy and Infectious Disease under P01 AI120756.

Footnotes

CONFLICTS OF INTEREST

The authors declare no financial conflicts of interest.

REFERENCES

  • 1.Ricklin D, Reis ES, Mastellos DC, Gros P, Lambris JD. Complement component C3 - The “Swiss Army Knife” of innate immunity and host defense. Immunol Rev 2016; 274: 33–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Jefferis R Isotype and glycoform selection for antibody therapeutics. Arch Biochem Biophys 2012; 526: 159–166. [DOI] [PubMed] [Google Scholar]
  • 3.Hughes-Jones NC, Gardner B. Reaction between the isolated globular sub-units of the complement component C1q and IgG-complexes. Mol Immunol 1979; 16: 697–701. [DOI] [PubMed] [Google Scholar]
  • 4.Diebolder CA, Beurskens FJ, de Jong RN, et al. Complement is Activated by IgG Hexamers Assembled at the Cell Surface. Science 2014; 343: 1260–1263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Wang G, de Jong RN, van den Bremer ET, et al. Molecular Basis of Assembly and Activation of Complement Component C1 in Complex with Immunoglobulin G1 and Antigen. Mol Cell 2016; 63: 135–145. [DOI] [PubMed] [Google Scholar]
  • 6.Feinstein A, Richardson N, Taussig MI. Immunoglobulin flexibility in complement activation. Immunol Today 1986; 7: 169–174. [DOI] [PubMed] [Google Scholar]
  • 7.Lei X, Liu C, Azadzoi K, et al. A novel IgM-H-ficolin complement pathway to attack allogenic cancer cells in vitro. Sci Rep 2015; 5: 7824. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Bindon CI, Hale G, Waldmann H. Importance of antigen specificity for complement-mediated lysis by monoclonal antibodies. Eur J Immunol 1988; 18: 1507–1514. [DOI] [PubMed] [Google Scholar]
  • 9.Cleary KLS, Chan HTC, James S, Glennie MJ, Cragg MS. Antibody Distance from the Cell Membrane Regulates Antibody Effector Mechanisms. J Immunol 2017; 198: 3999–4011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Tan LK, Shopes RJ, Oi VT, Morrison SL. Influence of the hinge region on complement activation, C1q binding, and segmental flexibility in chimeric human immunoglobulins. Proc Natl Acad Sci U S A 1990; 87: 162–166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Saphire EO, Parren PW, Pantophlet R, et al. Crystal Structure of a Neutralizing Human IgG Against HIV-1: A Template for Vaccine Design. Science 2001; 293: 1155–1159. [DOI] [PubMed] [Google Scholar]
  • 12.Teeling JL, Mackus WJ, Wiegman LJ, et al. The biological activity of human CD20 monoclonal antibodies is linked to unique epitopes on CD20. J Immunol 2006; 177: 362–371. [DOI] [PubMed] [Google Scholar]
  • 13.Chan HT, Hughes D, French RR, et al. CD20-induced Lymphoma Cell Death is Independent of Both Caspases and Its Redistribution into Triton X-100 Insoluble Membrane Rafts. Cancer Res 2003; 63: 5480–5489. [PubMed] [Google Scholar]
  • 14.Peschke B, Keller CW, Weber P, Quast I, Lunemann JD. Fc-Galactosylation of Human Immunoglobulin Gamma Isotypes Improves C1q Binding and Enhances Complement-Dependent Cytotoxicity. Front Immunol 2017; 8: 646. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Quast I, Keller CW, Maurer MA, et al. Sialylation of IgG Fc domain impairs complement-dependent cytotoxicity. J Clin Invest 2015; 125: 4160–4170. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Dekkers G, Treffers L, Plomp R, et al. Decoding the Human Immunoglobulin G-Glycan Repertoire Reveals a Spectrum of Fc-Receptor- and Complement-Mediated-Effector Activities. Front Immunol 2017; 8: 877. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.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 2007; 104: 8433–8437. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Monk PN, Scola AM, Madala P, Fairlie DP. Function, structure and therapeutic potential of complement C5a receptors. Br J Pharmacol 2007; 152: 429–448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Wang SY, Racila E, Taylor RP, Weiner GJ. NK-cell activation and antibody-dependent cellular cytotoxicity induced by rituximab-coated target cells is inhibited by the C3b component of complement. Blood 2008; 111: 1456–1463. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Palmer LJ, Damgaard C, Holmstrup P, Nielsen CH. Influence of complement on neutrophil extracellular trap release induced by bacteria. J Periodontal Res 2016; 51: 70–76. [DOI] [PubMed] [Google Scholar]
  • 21.Kalunian KC, Chatham WW, Massarotti EM, et al. Measurement of Cell-Bound Complement Activation Products Enhances Diagnostic Performance in Systemic Lupus Erythematosus. Arthritis Rheum 2012; 64: 4040–4047. [DOI] [PubMed] [Google Scholar]
  • 22.Martin M, Smolag KI, Bjork A, et al. Plasma C4d as marker for lupus nephritis in systemic lupus erythematosus. Arthritis Res Ther 2017; 19: 266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Nasri H, Ahmadi A, Rafieian-Kopaei M, Bashardoust B, Nasri P, Mubarak M. Association of glomerular C4d deposition with various demographic data in IgA nephropathy patients; a preliminary study. J Nephropathol 2015; 4: 19–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Lonati PA, Scavone M, Gerosa M, et al. Blood Cell-Bound C4d as a Marker of Complement Activation in Patients With the Antiphospholipid Syndrome. Front Immunol 2019; 10: 773. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Sethi S, Nasr SH, De Vriese AS, Fervenza FC. C4d as a Diagnostic Tool in Proliferative GN. J Am Soc Nephrol 2015; 26: 2852–2859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Singh G, Singh SK, Nalwa A, et al. Glomerular C4d Staining Does Not Exclude a C3 Glomerulopathy. Kidney Int Rep 2019; 4: 698–709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Liao T, Liu X, Ren J, et al. Noninvasive and quantitative measurement of C4d deposition for the diagnosis of antibody-mediated cardiac allograft rejection. EBioMedicine 2018; 37: 236–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Perez-Sanchez C, Cecchi I, Barbarroja N, et al. Early restoration of immune and vascular phenotypes in systemic lupus erythematosus and rheumatoid arthritis patients after B cell depletion. J Cell Mol Med 2019; 23: 6308–6318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Hillmen P, Elebute M, Kelly R, et al. Long-term effect of the complement inhibitor eculizumab on kidney function in patients with paroxysmal nocturnal hemoglobinuria. Am J Hematol 2010; 85: 553–559. [DOI] [PubMed] [Google Scholar]
  • 30.Hillmen P, Young NS, Schubert J, et al. The complement inhibitor eculizumab in paroxysmal nocturnal hemoglobinuria. N Engl J Med 2006; 355: 1233–1243. [DOI] [PubMed] [Google Scholar]
  • 31.Legendre CM, Licht C, Muus P, et al. Terminal complement inhibitor eculizumab in atypical hemolytic-uremic syndrome. N Engl J Med 2013; 368: 2169–2181. [DOI] [PubMed] [Google Scholar]
  • 32.Pittock SJ, Berthele A, Fujihara K, et al. Eculizumab in Aquaporin-4-Positive Neuromyelitis Optica Spectrum Disorder. N Engl J Med 2019; 381: 614–625. [DOI] [PubMed] [Google Scholar]
  • 33.Harder MJ, Hochsmann B, Dopler A, et al. Different Levels of Incomplete Terminal Pathway Inhibition by Eculizumab and the Clinical Response of PNH Patients. Front Immunol 2019; 10: 1639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Lin Z, Schmidt CQ, Koutsogiannaki S, et al. Complement C3dg-mediated erythrophagocytosis: implications for paroxysmal nocturnal hemoglobinuria. Blood 2015; 126: 891–894. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Howard J Jr., Utsugisawa K, Benatar M, et al. Safety and efficacy of eculizumab in anti-acetylcholine receptor antibody-positive refractory generalised myasthenia gravis (REGAIN): a phase 3, randomised, double-blind, placebo-controlled, multicentre study. Lancet Neurol 2017; 16: 976–986. [DOI] [PubMed] [Google Scholar]
  • 36.Ram S, Lewis LA, Rice PA. Infections of people with complement deficiencies and patients who have undergone splenectomy. Clin Microbiol Rev 2010; 23: 740–780. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Hellerud BC, Aase A, Herstad TK, et al. Critical roles of complement and antibodies in host defense mechanisms against Neisseria meningitidis as revealed by human complement genetic deficiencies. Infect Immun 2010; 78: 802–809. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Frasch CE, Borrow R, Donnelly J. Bactericidal antibody is the immunologic surrogate of protection against meningococcal disease. Vaccine 2009; 27 Suppl 2: B112–116. [DOI] [PubMed] [Google Scholar]
  • 39.Jendza K, Kato M, Salcius M, et al. A small-molecule inhibitor of C5 complement protein. Nat Chem Biol 2019; 15: 666–668. [DOI] [PubMed] [Google Scholar]
  • 40.Alawieh A, Langley EF, Tomlinson S. Targeted complement inhibition salvages stressed neurons and inhibits neuroinflammation after stroke in mice. Sci Transl Med 2018; 10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Yin C, Ackermann S, Ma Z, et al. ApoE attenuates unresolvable inflammation by complex formation with activated C1q. Nat Med 2019; 25: 496–506. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Jager U, D’Sa S, Schorgenhofer C, et al. Inhibition of complement C1s improves severe hemolytic anemia in cold agglutinin disease: a first-in-human trial. Blood 2019; 133: 893–901. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Reis ES, Berger N, Wang X, et al. Safety profile after prolonged C3 inhibition. Clin Immunol 2018; 197: 96–106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Spirig R, Campbell IK, Koernig S, et al. rIgG1 Fc Hexamer Inhibits Antibody-Mediated Autoimmune Disease via Effects on Complement and FcγRs. J Immunol 2018; 200: 2542–2553. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Zhou H, Olsen H, So E, et al. A fully recombinant human IgG1 Fc multimer (GL-2045) inhibits complement-mediated cytotoxicity and induces iC3b. Blood Adv 2017; 1: 504–515. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Stephen-Victor E, Bayry J. Multimerized IgG1 Fc molecule as an anti-inflammatory agent. Nat Rev Rheumatol 2018; 14: 390–392. [DOI] [PubMed] [Google Scholar]
  • 47.Tradtrantip L, Felix CM, Spirig R, Morelli AB, Verkman AS. Recombinant IgG1 Fc hexamers block cytotoxicity and pathological changes in experimental in vitro and rat models of neuromyelitis optica. Neuropharmacology 2018; 133: 345–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Lilienthal GM, Rahmoller J, Petry J, Bartsch YC, Leliavski A, Ehlers M. Potential of Murine IgG1 and Human IgG4 to Inhibit the Classical Complement and Fc? Receptor Activation Pathway. Front Immunol 2018; 9: 958. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Sudo M, Miyaji K, Spath PJ, Morita-Matsumoto K, Yamaguchi Y, Yuki N. Polyclonal IgM and IgA block in vitro complement deposition mediated by anti-ganglioside antibodies in autoimmune neuropathies. Int Immunopharmacol 2016; 40: 11–15. [DOI] [PubMed] [Google Scholar]
  • 50.Yuki N, Watanabe H, Nakajima T, Spath PJ. IVIG blocks complement deposition mediated by anti-GM1 antibodies in multifocal motor neuropathy. J Neurol Neurosurg Psychiatry 2011; 82: 87–91. [DOI] [PubMed] [Google Scholar]
  • 51.Sudo M, Yamaguchi Y, Spath PJ, et al. Different IVIG glycoforms affect in vitro inhibition of anti-ganglioside antibody-mediated complement deposition. PLoS One 2014; 9: e107772. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pagan JD, Kitaoka M, Anthony RM. Engineered Sialylation of Pathogenic Antibodies In Vivo Attenuates Autoimmune Disease. Cell 2018; 172: 564–577 e513. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Morgan BP, Orren A. Vaccination against meningococcus in complement-deficient individuals. Clin Exp Immunol 1998; 114: 327–329. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Wang SY, Veeramani S, Racila E, et al. Depletion of the C3 component of complement enhances the ability of rituximab-coated target cells to activate human NK cells and improves the efficacy of monoclonal antibody therapy in an in vivo model. Blood 2009; 114: 5322–5330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Rogers LM, Veeramani S, Weiner GJ. Complement in monoclonal antibody therapy of cancer. Immunol Res 2014; 59: 203–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Di Gaetano N, Cittera E, Nota R, et al. Complement activation determines the therapeutic activity of rituximab in vivo. J Immunol 2003; 171: 1581–1587. [DOI] [PubMed] [Google Scholar]
  • 57.Corrales L, Ajona D, Rafail S, et al. Anaphylatoxin C5a creates a favorable microenvironment for lung cancer progression. J Immunol 2012; 189: 4674–4683. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Markiewski MM, DeAngelis RA, Benencia F, et al. Modulation of the antitumor immune response by complement. Nat Immunol 2008; 9: 1225–1235. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Vlaicu SI, Tatomir A, Rus V, Rus H. Role of C5b-9 and RGC-32 in Cancer. Front Immunol 2019; 10: 1054. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Beers SA, French RR, Chan HT, et al. Antigenic modulation limits the efficacy of anti-CD20 antibodies: implications for antibody selection. Blood 2010; 115: 5191–5201. [DOI] [PubMed] [Google Scholar]
  • 61.Moskovich O, Fishelson Z. Live cell imaging of outward and inward vesiculation induced by the complement C5b-9 complex. J Biol Chem 2007; 282: 29977–29986. [DOI] [PubMed] [Google Scholar]
  • 62.Afshar-Kharghan V The role of the complement system in cancer. J Clin Invest 2017; 127: 780–789. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Dantas E, Erra Diaz F, Pereyra Gerber P, et al. Low pH impairs complement-dependent cytotoxicity against IgG-coated target cells. Oncotarget 2016; 7: 74203–74216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Glennie MJ, French RR, Cragg MS, Taylor RP. Mechanisms of killing by anti-CD20 monoclonal antibodies. Mol Immunol 2007; 44: 3823–3837. [DOI] [PubMed] [Google Scholar]
  • 65.Teeling JL, French RR, Cragg MS, et al. Characterization of new human CD20 monoclonal antibodies with potent cytolytic activity against non-Hodgkin lymphomas. Blood 2004; 104: 1793–1800. [DOI] [PubMed] [Google Scholar]
  • 66.Beurskens FJ, Lindorfer MA, Farooqui M, et al. Exhaustion of cytotoxic effector systems may limit monoclonal antibody-based immunotherapy in cancer patients. J Immunol 2012; 188: 3532–3541. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Dechant M, Weisner W, Berger S, et al. Complement-Dependent Tumor Cell Lysis Triggered by Combinations of Epidermal Growth Factor Receptor Antibodies. Cancer Res 2008; 68: 4998–5003. [DOI] [PubMed] [Google Scholar]
  • 68.Jacobsen HJ, Poulsen TT, Dahlman A, et al. Pan-HER, an Antibody Mixture Simultaneously Targeting EGFR, HER2, and HER3, Effectively Overcomes Tumor Heterogeneity and Plasticity. Clin Cancer Res 2015; 21: 4110–4122. [DOI] [PubMed] [Google Scholar]
  • 69.Chao MP, Alizadeh AA, Tang C, et al. Anti-CD47 Antibody Synergizes with Rituximab to Promote Phagocytosis and Eradicate Non-Hodgkin Lymphoma. Cell 2010; 142: 699–713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Oostindie SC, van der Horst HJ, Lindorfer MA, et al. CD20 and CD37 antibodies synergize to activate complement by Fc-mediated clustering. Haematologica 2019; 104: 1841–1852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Rossi O, Coward C, Goh YS, et al. The essential role of complement in antibody-mediated resistance to Salmonella. Immunology 2019; 156: 69–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Vogt MR, Dowd KA, Engle M, et al. Poorly neutralizing cross-reactive antibodies against the fusion loop of West Nile virus envelope protein protect in vivo via Fc receptor and complement-dependent effector mechanisms. J Virol 2011; 85: 11567–11580. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Schiela B, Bernklau S, Malekshahi Z, et al. Active Human Complement Reduces the Zika Virus Load via Formation of the Membrane-Attack Complex. Front Immunol 2018; 9: 2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Wu Y, Cho M, Shore D, et al. A potent broad-spectrum protective human monoclonal antibody crosslinking two haemagglutinin monomers of influenza A virus. Nat Commun 2015; 6: 7708. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Benhnia MR, McCausland MM, Laudenslager J, et al. Heavily isotype-dependent protective activities of human antibodies against vaccinia virus extracellular virion antigen B5. J Virol 2009; 83: 12355–12367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Li F, Freed DC, Tang A, et al. Complement enhances in vitro neutralizing potency of antibodies to human cytomegalovirus glycoprotein B (gB) and immune sera induced by gB/MF59 vaccination. NPJ Vaccines 2017; 2: 36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Bukreyev A, Yang L, Collins PL. The Secreted G Protein of Human Respiratory Syncytial Virus Antagonizes Antibody-Mediated Restriction of Replication Involving Macrophages and Complement. J Virol 2012; 86: 10880–10884. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Ackerman ME, Mikhailova A, Brown EP, et al. Polyfunctional HIV-Specific Antibody Responses Are Associated with Spontaneous HIV Control. PLoS Pathog 2016; 12: e1005315. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Alter G, Dowell KG, Brown EP, et al. High-resolution definition of humoral immune response correlates of effective immunity against HIV. Mol Syst Biol 2018; 14: e7881. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Gulati S, Beurskens FJ, de Kreuk BJ, et al. Complement alone drives efficacy of a chimeric antigonococcal monoclonal antibody. PLoS Biol 2019; 17: e3000323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Boyle MJ, Reiling L, Feng G, et al. Human Antibodies Fix Complement to Inhibit Plasmodium falciparum Invasion of Erythrocytes and Are Associated with Protection against Malaria. Immunity 2015; 42: 580–590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Reiling L, Boyle MJ, White MT, et al. Targets of complement-fixing antibodies in protective immunity against malaria in children. Nat Commun 2019; 10: 610. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Oyong DA, Wilson DW, Barber BE, et al. Induction and Kinetics of Complement-Fixing Antibodies Against Plasmodium vivax Merozoite Surface Protein 3α and Relationship With Immunoglobulin G Subclasses and Immunoglobulin M. J Infect Dis 2019; 220: 1950–1961. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Johnson JB, Capraro GA, Parks GD. Differential mechanisms of complement-mediated neutralization of the closely related paramyxoviruses simian virus 5 and mumps virus. Virology 2008; 376: 112–123. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Bottermann M, Foss S, Caddy SL, et al. Complement C4 Prevents Viral Infection through Capsid Inactivation. Cell Host Microbe 2019; 25: 617–629 e617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lubinski JM, Lazear HM, Awasthi S, Wang F, Friedman HM. The herpes simplex virus 1 IgG fc receptor blocks antibody-mediated complement activation and antibody-dependent cellular cytotoxicity in vivo. J Virol 2011; 85: 3239–3249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Ermert D, Weckel A, Magda M, et al. Human IgG Increases Virulence of Streptococcus pyogenes through complement evasion. J Immunol 2018; 200: 3495–3505. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Yang K, Lan J, Shepherd N, et al. Blockage of CD59 Function Restores Activities of Neutralizing and Nonneutralizing Antibodies in Triggering Antibody-Dependent Complement-Mediated Lysis of HIV-1 Virions and Provirus-Activated Latently Infected Cells. J Virol 2015; 89: 9393–9406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Isaacs SN, Kotwal GJ, Moss B. Vaccinia virus complement-control protein prevents antibody-dependent complement-enhanced neutralization of infectivity and contributes to virulence. Proc Natl Acad Sci U S A 1992; 89: 628–632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Johnson JB, Borisevich V, Rockx B, Parks GD. A Novel Factor I Activity in Nipah Virus Inhibits Human Complement Pathways through Cleavage of C3b. J Virol 2015; 89: 989–998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Sorbara MT, Foerster EG, Tsalikis J, et al. Complement C3 Drives Autophagy-Dependent Restriction of Cyto-invasive Bacteria. Cell Host Microbe 2018; 23: 644–652 e645. [DOI] [PubMed] [Google Scholar]
  • 92.Xie J, Zhi H, Garrigues RJ, Keightley A, Garcia BL, Skare JT. Structural determination of the complement inhibitory domain of Borrelia burgdorferi BBK32 provides insight into classical pathway complement evasion by Lyme disease spirochetes. PLoS Pathog 2019; 15: e1007659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Leemans A, De Schryver M, Van der Gucht W, et al. Antibody-Induced Internalization of the Human Respiratory Syncytial Virus Fusion Protein. J Virol 2017; 91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Anand SP, Grover JR, Tolbert WD, et al. Antibody-Induced Internalization of HIV-1 Env Proteins Limits Surface Expression of the Closed Conformation of Env. J Virol 2019; 93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ermert D, Ram S, Laabei M. The hijackers guide to escaping complement: Lessons learned from pathogens. Mol Immunol 2019; 114: 49–61. [DOI] [PubMed] [Google Scholar]
  • 96.Senaldi G, Peakman M, McManus T, Davies ET, Tee DE, Vergani D. Activation of the complement system in human immunodeficiency virus infection: relevance of the classical pathway to pathogenesis and disease severity. J Infect Dis 1990; 162: 1227–1232. [DOI] [PubMed] [Google Scholar]
  • 97.Stein KR, Gardner TJ, Hernandez RE, et al. CD46 facilitates entry and dissemination of human cytomegalovirus. Nat Commun 2019; 10: 2699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Biryukov S, Angov E, Landmesser ME, Spring MD, Ockenhouse CF, Stoute JA. Complement and Antibody-mediated Enhancement of Red Blood Cell Invasion and Growth of Malaria Parasites. EBioMedicine 2016; 9: 207–216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Schlesinger LS, Bellinger-Kawahara CG, Payne NR, Horwitz MA. Phagocytosis of Mycobacterium tuberculosis is mediated by human monocyte complement receptors and complement component C3. J Immunol 1990; 144: 2771–2780. [PubMed] [Google Scholar]
  • 100.Heesters BA, Lindqvist M, Vagefi PA, et al. Follicular Dendritic Cells Retain Infectious HIV in Cycling Endosomes. PLoS Pathog 2015; 11: e1005285. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Steiner DJ, Furuya Y, Metzger DW. Host-pathogen interactions and immune evasion strategies in Francisella tularensis pathogenicity. Infect Drug Resist 2014; 7: 239–251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Pierson TC, Xu Q, Nelson S, et al. The stoichiometry of antibody-mediated neutralization and enhancement of West Nile virus infection. Cell Host Microbe 2007; 1: 135–145. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Lindorfer M, Beum P, Taylor R. CD20 mAb-Mediated Complement Dependent Cytotoxicity of Tumor Cells is Enhanced by Blocking the Action of Factor I. Antibodies 2013; 2: 598–616. [Google Scholar]
  • 104.Macor P, Tripodo C, Zorzet S, et al. In vivo targeting of human neutralizing antibodies against CD55 and CD59 to lymphoma cells increases the antitumor activity of rituximab. Cancer Res 2007; 67: 10556–10563. [DOI] [PubMed] [Google Scholar]
  • 105.Gelderman KA, Lam S, Gorter A. Inhibiting complement regulators in cancer immunotherapy with bispecific mAbs. Expert Opin Biol Ther 2005; 5: 1593–1601. [DOI] [PubMed] [Google Scholar]
  • 106.Cruz JW, Damko E, Modi B, et al. A novel bispecific antibody platform to direct complement activity for efficient lysis of target cells. Sci Rep 2019; 9: 12031. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Idusogie EE, Presta LG, Gazzano-Santoro H, et al. Mapping of the C1q binding site on rituxan, a chimeric antibody with a human IgG1 Fc. J Immunol 2000; 164: 4178–4184. [DOI] [PubMed] [Google Scholar]
  • 108.de Jong RN, Beurskens FJ, Verploegen S, 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 2016; 14: e1002344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Tammen A, Derer S, Schwanbeck R, et al. Monoclonal Antibodies against Epidermal Growth Factor Receptor Acquire an Ability To Kill Tumor Cells through Complement Activation by Mutations That Selectively Facilitate the Hexamerization of IgG on Opsonized Cells. J Immunol 2017; 198: 1585–1594. [DOI] [PubMed] [Google Scholar]
  • 110.Oostindie SC, van der Horst HJ, Lindorfer MA, et al. CD20 and CD37 antibodies synergize to activate complement by Fc-mediated clustering. Haematologica 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.van der Horst HJ, Overdijk MB, Breij EC, Chamuleau M, Lokhorst HM, Mutis T. Potent Ex Vivo Anti-Tumor Activity in Relapsed Refractory Multiple Myeloma Using Novel DR5-Specific Antibodies with Enhanced Capacity to Form Hexamers upon Target Binding. Am Soc Hematology; 2017. [Google Scholar]
  • 112.Cook EM, Lindorfer MA, van der Horst H, et al. Antibodies That Efficiently Form Hexamers upon Antigen Binding Can Induce Complement-Dependent Cytotoxicity under Complement-Limiting Conditions. J Immunol 2016; 197: 1762–1775. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Taylor RP, Lindorfer MA, Cook EM, et al. Hexamerization-enhanced CD20 antibody mediates complement-dependent cytotoxicity in serum genetically deficient in C9. Clin Immunol 2017; 181: 24–28. [DOI] [PubMed] [Google Scholar]
  • 114.Cohen D, Colvin RB, Daha MR, et al. Pros and cons for C4d as a biomarker. Kidney Int 2012; 81: 628–639. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Gallenkamp J, Spanier G, Worle E, et al. A novel multiplex detection array revealed systemic complement activation in oral squamous cell carcinoma. Oncotarget 2018; 9: 3001–3013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Moreno Gonzales MA, Mitema DG, Smith BH, et al. Comparison Between Total IgG, C1q, and C3d Single Antigen Bead Assays in Detecting Class I Complement-Binding Anti-HLA Antibodies. Transplant Proc 2017; 49: 2031–2035. [DOI] [PubMed] [Google Scholar]
  • 117.Lan JH, Tinckam K. Clinical Utility of Complement Dependent Assays in Kidney Transplantation. Transplantation 2018; 102: S14–S22. [DOI] [PubMed] [Google Scholar]
  • 118.Mutti M, Ramoni K, Nagy G, Nagy E, Szijarto V. A New Tool for Complement Research: In vitro Reconstituted Human Classical Complement Pathway. Front Immunol 2018; 9: 2770. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Thurman JM, Kulik L, Orth H, et al. Detection of complement activation using monoclonal antibodies against C3d. J Clin Invest 2013; 123: 2218–2230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Fischinger S, Fallon JK, Michell AR, et al. A high-throughput, bead-based, antigen-specific assay to assess the ability of antibodies to induce complement activation. J Immunol Methods 2019; 473: 112630. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Thomas SR, Leung S, Knox K, et al. Development of flow cytometric opsonophagocytosis and antibody-mediated complement deposition assays for non-typeable Haemophilus influenzae. BMC Microbiol 2018; 18: 167. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Mastellos D, Papadimitriou JC, Franchini S, Tsonis PA, Lambris JD. A novel role of complement: mice deficient in the fifth component of complement (C5) exhibit impaired liver regeneration. J Immunol 2001; 166: 2479–2486. [DOI] [PubMed] [Google Scholar]
  • 123.Zhi H, Xie J, Skare JT. The Classical Complement Pathway Is Required to Control Borrelia burgdorferi Levels During Experimental Infection. Front Immunol 2018; 9: 959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Benhnia MR, McCausland MM, Moyron J, et al. Vaccinia virus extracellular enveloped virion neutralization in vitro and protection in vivo depend on complement. J Virol 2009; 83: 1201–1215. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125.Xu D, Alegre ML, Varga SS, et al. In vitro characterization of five humanized OKT3 effector function variant antibodies. Cell Immunol 2000; 200: 16–26. [DOI] [PubMed] [Google Scholar]
  • 126.Hessell AJ, Hangartner L, Hunter M, et al. Fc receptor but not complement binding is important in antibody protection against HIV. Nature 2007; 449: 101–104. [DOI] [PubMed] [Google Scholar]
  • 127.Hezareh M, Hessell AJ, Jensen RC, van de Winkel JG, Parren PW. Effector function activities of a panel of mutants of a broadly neutralizing antibody against human immunodeficiency virus type 1. J Virol 2001; 75: 12161–12168. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Schlothauer T, Herter S, Koller CF, et al. Novel human IgG1 and IgG4 Fc-engineered antibodies with completely abolished immune effector functions. Protein Eng Des Sel 2016; 29: 457–466. [DOI] [PubMed] [Google Scholar]
  • 129.Sazinsky SL, Ott RG, Silver NW, Tidor B, Ravetch JV, Wittrup KD. Aglycosylated immunoglobulin G1 variants productively engage activating Fc receptors. Proc Natl Acad Sci U S A 2008; 105: 20167–20172. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Ugurlar D, Howes SC, de Kreuk BJ, et al. Structures of C1-IgG1 provide insights into how danger pattern recognition activates complement. Science 2018; 359: 794–797. [DOI] [PubMed] [Google Scholar]
  • 131.Moore GL, Chen H, Karki S, Lazar GA. Engineered Fc variant antibodies with enhanced ability to recruit complement and mediate effector functions. MAbs 2010; 2: 181–189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Natsume A, In M, Takamura H, et al. Engineered antibodies of IgG1/IgG3 mixed isotype with enhanced cytotoxic activities. Cancer Res 2008; 68: 3863–3872. [DOI] [PubMed] [Google Scholar]
  • 133.Kelton W, Mehta N, Charab W, et al. IgGA: a “cross-isotype” engineered human Fc antibody domain that displays both IgG-like and IgA-like effector functions. Chem Biol 2014; 21: 1603–1609. [DOI] [PubMed] [Google Scholar]
  • 134.Lee CH, Romain G, Yan W, et al. IgG Fc domains that bind C1q but not effector Fcγ receptors delineate the importance of complement-mediated effector functions. Nat Immunol 2017; 18: 889–898. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES