This article was downloaded by: [University of Manitoba Libraries] On: 07 September 2015, At: 11:04 Publisher: Taylor & Francis Informa Ltd Registered in England and Wales Registered Number: 1072954 Registered office: 5 Howick Place, London, SW1P 1WG

mAbs Publication details, including instructions for authors and subscription information: http://www.tandfonline.com/loi/kmab20

IgG4 can induce an M2-like phenotype in human monocyte-derived macrophages through FcγRI a

a

a

a

Jennifer FA Swisher , Devin A Haddad , Anna G McGrath , Gunther H Boekhoudt & Gerald M a

Feldman a

Laboratory of Molecular and Developmental Immunology; Division of Monoclonal Antibodies; Office of Biotechnology Products; Center for Drug Evaluation and Research; Food and Drug Administration; Bethesda, MD USA Accepted author version posted online: 30 Oct 2014.

Click for updates To cite this article: Jennifer FA Swisher, Devin A Haddad, Anna G McGrath, Gunther H Boekhoudt & Gerald M Feldman (2014) IgG4 can induce an M2-like phenotype in human monocyte-derived macrophages through FcγRI, mAbs, 6:6, 1377-1384, DOI: 10.4161/19420862.2014.975657 To link to this article: http://dx.doi.org/10.4161/19420862.2014.975657

PLEASE SCROLL DOWN FOR ARTICLE Taylor & Francis makes every effort to ensure the accuracy of all the information (the “Content”) contained in the publications on our platform. However, Taylor & Francis, our agents, and our licensors make no representations or warranties whatsoever as to the accuracy, completeness, or suitability for any purpose of the Content. Any opinions and views expressed in this publication are the opinions and views of the authors, and are not the views of or endorsed by Taylor & Francis. The accuracy of the Content should not be relied upon and should be independently verified with primary sources of information. Taylor and Francis shall not be liable for any losses, actions, claims, proceedings, demands, costs, expenses, damages, and other liabilities whatsoever or howsoever caused arising directly or indirectly in connection with, in relation to or arising out of the use of the Content. This article may be used for research, teaching, and private study purposes. Any substantial or systematic reproduction, redistribution, reselling, loan, sub-licensing, systematic supply, or distribution in any form to anyone is expressly forbidden. Terms & Conditions of access and use can be found at http:// www.tandfonline.com/page/terms-and-conditions

BRIEF REPORT mAbs 6:6, 1377--1384; November/December 2014

IgG4 can induce an M2-like phenotype in human monocyte-derived macrophages through FcgRI Jennifer FA Swisher, Devin A Haddad, Anna G McGrath, Gunther H Boekhoudt, and Gerald M Feldman* Laboratory of Molecular and Developmental Immunology; Division of Monoclonal Antibodies; Office of Biotechnology Products; Center for Drug Evaluation and Research; Food and Drug Administration; Bethesda, MD USA

Keywords: IgG4, Fc gamma receptors, immune complex, monocyte-derived macrophage, alternatively activated M2 macrophage

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

Abbreviations: IC, immune complexes; ITAM, immunoreceptor tyrosine-based activation motif; ITIM, immunoreceptor tyrosine-based inhibitory motif; TLR, Toll Like Receptor; IP-10, Interferon Inducible Protein-10

Antibodies evoke cellular responses through the binding of their Fc region to Fc receptors, most of which contain immunoreceptor tyrosine-based activation motif domains and are thus considered “activating.” However, there is a growing appreciation of these receptors for their ability to deliver an inhibitory signal as well. We previously described one such phenomenon whereby interferon (IFN)g signaling is inhibited by immune complex signaling through FcgRI. To understand the implications of this in the context of therapeutic antibodies, we assessed individual IgG subclasses to determine their ability to deliver this anti-inflammatory signal in monocyte-derived macrophages. Like IgG1, we found that IgG4 is fully capable of inhibiting IFNg-mediated events. In addition, F(ab’)2 fragments that interfere with FcgRI signaling reversed this effect. For mAbs developed with either an IgG1 or an IgG4 constant region for indications where inflammation is undesirable, further examination of a potential Fc-dependent contribution to their mechanism of action is warranted.

Introduction Antibodies elicit a variety of cellular responses through the binding of their Fc region to cognate receptors. Members of the Fcg receptor family (FcgRs) are found on most immune cell subtypes, as well as a growing number of non-immune cells,1-3 enabling a variety of cellular responses depending on the antibody and on the context. There are currently hundreds of mAbs under clinical development, including both intact mAbs as well as Fc fusion proteins, and the majority possess a human IgG Fc region capable of binding to one or more of the known FcgRs. Many of these molecules have been designed to harness activating effector functions such as antibody-dependent cell-mediated cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC) to aid or enhance their mechanism of action.4 Other antibody-related therapeutics are designed to work primarily through neutralization of their target by Fab-region binding; the Fc region is assumed to be “silent,” other than its contribution to FcRn binding that grants IgG its long half-life. Quite frequently, these socalled neutralizing antibodies are directed toward inflammatory cytokines, their receptors, or other molecules involved in the inflammation that underlies many chronic pathologies.5 However, anecdotal evidence of Fc-region involvement in the mechanism of action of some members of this latter class has begun to emerge.

Human antibodies of the IgG class, either in monovalent form or in multivalent form complexed with antigen, bind various members of the family of FcgRs. It is thought that most often, these interactions result in activation of the immune system by triggering the immunoreceptor tyrosine-based activation motif (ITAM)-containing Fcg receptors or accessory molecules.6 Only one member of this family, FcgRIIb, signals instead through an immunoreceptor tyrosine-based inhibitory motif (ITIM) domain and has been shown in mice to play a role in inhibition of inflammatory responses and in the establishment of tolerance.7,8 However, there is growing evidence that some ITAM-containing activating receptors have an intrinsic ability to deliver an alternate inhibitory signal as well.9 Our lab previously described one such phenomenon whereby IFNg signaling events are inhibited by immune complex (IC)-mediated signaling through FcgRI.10 These lines of inquiry have contributed to a growing appreciation of the complexity of Fc-encoded functions present in therapeutic monoclonal antibodies. In addition, each IgG subclass has a different specificity toward each of the different FcgRI that is also dependent on the type and size of the immune complex in which it is presented.11 To further our understanding of these inhibitory pathways, we extended our earlier studies to human macrophages because they display more complete expression of all of the human FcgRI than monocytes and are found in nearly every tissue.12 We looked not only at the ability of IC to inhibit IFNg receptor

This article not subject to US copyright law. *Correspondence to: Gerald M Feldman; Email: [email protected] Submitted: 07/16/2014; Revised: 09/09/2014; Accepted: 09/14/2014 http://dx.doi.org/10.4161/19420862.2014.975657

www.landesbioscience.com

mAbs

1377

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

signal transduction, but also whether other inflammatory receptor systems were affected by examining the effect of IC on Toll Like Receptor (TLR)4-mediated events in the macrophage. Finally, as therapeutic monoclonal antibodies create ICs of a single IgG subtype by binding their antigen or a cell bearing that antigen on its surface, we analyzed the effect of individual IgG subtypes as well as whole human IgG in our IC model. We found that the IC model using human IgG evoked equally strong inhibition of IFNg signaling in macrophages as was previously seen in monocytes, and that although interleukin (IL)4 receptor signaling remained unaffected, TLR4-dependent events were dramatically inhibited. Most surprisingly, however, we found that complexed IgG4 was fully capable of inhibiting IFNg-mediated events in primary monocyte-derived macrophages. Although it has been known that IgG4 is able to bind the high affinity FcgRI, this is the first report that IgG4 is capable of transducing any type of signal through an Fcg receptor.

Results IC-mediated inhibition of IFNg signaling has previously been studied using freshly elutriated human monocytes.10 However, in vivo, macrophages may represent the more relevant and functionally mature population responsible for establishing inflammation in response to invasion or injury.13,14 More importantly, they are responsible for the establishment and maintenance of the chronic inflammation underlying numerous pathologies targeted by therapeutic intervention strategies.15 We therefore extended our studies to assess IC-mediated inhibition of IFNg signaling in mature macrophages and whether it affected other macrophage inflammatory receptor systems, such as TLR4. Although previous studies demonstrated a lack of effect on signaling downstream of more immunomodulatory cytokines such as GM-CSF and IL4,16 it was of interest to determine whether additional inflammatory pathways, such as the Toll-like receptors (TLRs), responsible for microbial pattern recognition, are affected by the presence of IC in this model. Interferon Inducible Protein 10 (IP10) is rapidly and vigorously produced in response to IFNg:17 However, in IC treated monocytes, the amount of IP10 produced is dramatically inhibited.10 In macrophages, the inhibition of IP10 mRNA accumulation and protein secretion was likewise inhibited to a similar degree in the presence of IC (Fig. 1A). IP10 mRNA and protein are also induced following TLR4 triggering by lipopolysaccharide (LPS); this IP10 production was decreased to an even greater degree by immune complex pretreatment than was seen downstream of IFNg (Fig. 1B). Indeed, the levels of other inflammatory chemokines and cytokines downstream of LPS treatment were similarly reduced, including RANTES and IL8 (Fig. 1C); this was also true for the macrophage activation marker CD80 as well as the scavenger receptor CD163 (Fig. 1D). These data demonstrate that Fcdependent inhibitory mechanisms are intact and even augmented in differentiated human monocyte-derived macrophages, reducing the production of IP10 and other cytokines, chemokines, and cell surface molecules that activate the immune system in

1378

response to host or microbial inflammatory activation signals such as IFNg or LPS. In contrast, IL-10 production was induced by IC independent of the second stimulus (Fig. 1C), in agreement with other studies reporting IC-dependent production of this anti-inflammatory cytokine.18-20 IL-10 is known to induce an alternatively activated, regulatory or “M2” macrophage phenotype that, in turn, leads other cells toward a more Th2-type immune response.18 In keeping with these observations, IC also induced the expression of CD105, a transforming growth factor b receptor family member, on the macrophage surface (Fig. 1D). Many studies performed to date have used pooled human IgG to mimic normal immune complexes, as would happen in the normal course of infection. However, treatment with therapeutic antibodies presumably results in the formation of immune complexes- involving the therapeutic and its target- that are composed of antibodies of a single IgG subclass. To investigate the relevance of these immunomodulatory mechanisms to therapeutic monoclonal antibodies, we tested the ability of myelomaderived IgG1, IgG2, and IgG4 to mediate IC-dependent inhibition of IFNg and LPS signaling. These immunoglobulins were chosen as they represent those currently found among licensed therapeutic monoclonal antibodies (http://www.antibodysociety. org/news/approved_mabs.php). IgG1 has been shown to display superior binding to all FcgRs, whereas IgG2 is thought to only bind to the H131 allotype of the FcgR2a.21 In contrast, IgG4 is commonly believed to be devoid of appreciable FcgR binding.22,23 Surprisingly, our results demonstrate significant concentrationdependent immunomodulatory behavior of each IgG subclass, including IgG4 and, to a lesser degree, IgG2 (Fig. 2). IgG4 antibodies have never been previously shown capable of transducing a signal through any Fcg receptor, including FcgRI. It is frequently stated in the literature that glycosylation of the IgG Fc region is required for Fcg receptor family binding.21, 24 To determine whether IC comprising either IgG1 or IgG4 bore this dependence, antibody of each subclass was subjected to complete deglycosylation with PNGase F. Upon testing deglycosylated IgG1 and IgG4 against their native (mock-treated) counterparts as well as whole human IgG, we found that the ability of IgG1 to inhibit IFNg signaling in this model was completely independent of glycosylation, whereas the ability of IgG4 to affect this outcome was only minimally affected by removal of Fc-associated glycans (Fig. 3). As glycosylation dependence of FcgR binding is widely accepted, our data suggest that either a different receptor complex is responsible for this ability to inhibit inflammatory signaling in macrophages, or that glycosylation was not absolutely required for FcgRI binding by these IgG subtypes in this IC model. Whereas ICs derived from pooled IgG inhibit IFNg signaling in monocytes specifically through FcgRI,10 it was not clear that this would be the case in macrophages. Not only do macrophages display the broadest FcgR expression of any cell type and higher expression of the FcgRs that are common to monocytes,21 but published reports suggest that all 3 classes of ITAM-containing Fcg receptors can evoke inhibitory events.10,25,26 IgG4 antibodies (either monomeric or as

mAbs

Volume 6 Issue 6

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

Figure 1. IC-dependent changes in macrophage cytokine production and surface expression in response to inflammatory stimuli. One-day old monocytes or 7-day-old macrophages were plated on IC-treated or untreated plates for 1 hour prior to treatment with 10 ng/mL IFNg or LPS, as indicated. (A and B) RNA was isolated after 1.5 hours at 37 C, reverse transcribed, and levels of the IP10 were analyzed by real-time polymerase chain reaction. Alternatively, supernatants were removed and clarified after 6 hours at 37 C and analyzed for IP10. (C) Supernatants collected after 20 hours were assessed for IL-10, RANTES, and IL-8 by multiplex analysis. Results are the mean § SEM of 5 experiments with separate donors, performed in triplicate. For A and B, the data for each donor are normalized to the amount of IP10 RNA or protein induced by IFNg or LPS treatment alone (100%). For C, the data for each donor are normalized to the amount of secreted protein induced by LPS treatment (100%). (D) Macrophages exposed to IFNg or LPS treatment for 20 hours were stained with Alexa 647 mouse anti-human CD163, PE-Cy5 conjugated mouse anti-human CD80, or APC conjugated mouse anti-human CD105, as indicated. Results are the mean § SEM of 3 experiments with separate donors.

immune complexes), however, have never previously been demonstrated to transduce a signal through any Fcg receptor. To determine the dependence of IC-mediated inhibition of IFNg signaling on FcgR, blocking F(ab’)2 fragments specific to each FcgR class were used alone and in combination to assess the relative contribution of each (Fig. 4A). Consistent with previous data from human monocytes, the reduction of IFNg signaling by polyclonal IgG or IgG1 IC showed greater dependence on FcgRI than the other Fcg receptors.

www.landesbioscience.com

Examination of the raw data suggests that FcgRII or FcgRIII may be responsible for significant contributions to IP10 reduction in individual donors when IC are composed of IgG1 (data not shown). However, when data are combined across multiple donors, only the F(ab’)2 that interferes with FcgRI demonstrates significant reversal of IC-dependent signal inhibition. In contrast, IgG4-mediated inhibition of IFNinduced signaling was reduced only when FcgRI was blocked, with no similar variable dependence on the other FcgRs,

mAbs

1379

inhibition of IP10 mRNA accumulation that was invoked by IgG4 IC was indeed reversed by treatment of macrophages with the highly specific Syk inhibitor Bay 61–3606 prior to IgG4 IC exposure (Fig. 4B). IgG4 IC treatment was also able to reduce inflammatory cytokine secretion (Fig. 4C) and surface marker expression (Fig. 4D) in response to LPS, providing further evidence that IgG4 works through the same general pathway as whole IgG from human serum.

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

Discussion

Figure 2. IgG subclass specificity of IC inhibition of IFNg-dependent signaling. Seven-day-old macrophages were trypsinized and plated on ICtreated or untreated plates for 1 hour prior to treatment with 10 ng/mL IFNg as indicated. Five concentrations of each indicated IgG subtype were used to coat the wells: 50.0, 16.6, 5.5, 1.9, 0.7 mg/mL, from left to right. Supernatants were removed and clarified after 6 hours at 37 C and analyzed for IP10 by multiplex. Results are the mean § SEM of 3 experiments with separate donors, performed in triplicate. The data for each donor are normalized to the amount of IP10 protein induced by IFNg treatment alone (100%).

indicating the differences in the binding behavior of IC from these 2 subclasses for the different FcgRs. To examine whether the ability of IgG4 to inhibit IFNinduced IP10 transcription indeed depends on signaling components necessary for the activating pathways of FcgRs, the effect of Syk inhibitors on this phenomenon were investigated. The

Figure 3. IC-dependent inhibition of IFN-dependent IP10 production does not require Fc glycosylation. Myeloma-derived IgG1 and IgG4 were either treated with PNGase for 48 hrs or mock-treated in buffer under the same conditions but without enzyme. Seven-day-old macrophages were trypsinized and plated on plates coated with the indicated antibody preparation for 1h prior to treatment with 10 ng/mL IFNg as indicated. Supernatants were removed and clarified after 6 hours at 37 C and analyzed for IP10 by multiplex. Results are the mean § SEM of 3 experiments performed in triplicate, normalized to IP10 protein induced by IFNg treatment alone (100%).

1380

Decades of research into how intravenous immunoglobulin (IVIG) exerts its therapeutic effect in numerous pathologies have uncovered multiple mechanisms whereby immunoglobulins can suppress inflammation.27,28 One of these involves antibody and immune complex binding to the ITIM-containing inhibitory receptor FcgRIIb, which acts by recruiting phosphatases to counter nearby signaling events.8 Sialylated IgGs can in turn bind glycosylation-dependent receptors such as DC-SIGN in humans or SIGN-R1 in mice, which causes increased expression of this inhibitory receptor, presumably skewing the overall effects of antibodies and immune complexes away from immune activation and toward tolerance.7, 29 Although the importance of this inhibitory circuit has been convincingly demonstrated in mouse models of inflammation,30 its relative contribution in humans is not yet as clear. The neonatal Fc receptor, FcRn, is responsible for the endosomal recycling of antibodies that imparts their long half-life.31,32 Another proposed mechanism of action of IVIG is competition with problematic auto-antibodies for FcRn binding, resulting in their accelerated clearance. Related to this is the notion that IVIG may compete with autoantibody immune complexes for activating FcgR binding.28 The fact that IVIG needs to be used at much higher doses to treat autoimmune pathologies compared with those used to complement immune deficiencies is in agreement with these mechanisms of general competition that involve the antibody Fc region. In addition, Fc-mediated inhibitory signaling has been described downstream of multiple ITAM-containing, or “activating” Fcg receptors. Work from our lab and others have made clear that each of the human FcgRs has the capacity to affect this balance in either direction.9, 10, 33 Previous results from our lab demonstrated that IC made of human IgG dampen the response of human monocytes to IFNg through FcgRI.10 The data reported in this paper extend this finding to confirm that the response to IFNg is similarly inhibited in primary human monocyte-derived macrophages (Fig. 1A). We further confirm the veracity of our model system by demonstrating that IC treatment diminishes inflammatory cytokine production (Fig. 1B and 1C) and activation marker expression (Fig. 1D) following LPS treatment. Conversely, IC treatment increases secretion of anti-inflammatory IL-10 and the surface expression of TGF-b receptor family member CD105 (Fig. 1B and 1C). These changes are consistent with the proposed immune complex-dependent “M2b” macrophage phenotype that has been previously described.34 Interestingly, although increased CD163 surface expression is a marker for some types of

mAbs

Volume 6 Issue 6

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

M2 macrophages, its expression has been shown to be decreased by immune complexes,35 in keeping with the idea that the M2 phenotype induced by immune complexes represents a hybrid between classical and alternative macrophage activation. It would appear as though the low affinity activating receptors FcgRIIa (through its own ITAM) and FcgRIIIa (through the ITAM-containing accessory molecule, the common gamma chain) achieve inhibitory signaling through a different mechanism than FcgRI; it requires low signal strength due to monovalent or low avidity/affinity ligand.9 In contrast, occupancy of FcgRI on macrophages by monovalent IgG produces a tonic signal required for full IFNg receptor signal strength.36 In agreement with this paradigm, monovalent IgG has no significant effect on IFNg signaling in our system (unpubFigure 4. IgG4 IC delivers an inhibitory signal through FcgRI. (A) Seven-day-old macrophages were lished data) whereas there is a direct postrypsinized and treated with the indicated F(ab’)2 fragments (at 10 mg/mL each) for 300 on ice prior itive correlation between the density of to plating on indicated IC for 1 hour prior to treatment with 10 ng/mL IFNg as indicated. Supernathe IC and the degree of inhibition of tants were removed and clarified after 6 hours at 37 C and analyzed for IP10 by multiplex. Results IP10 production (Fig. 3). Independent are the mean § SEM of 5 experiments with separate donors performed in triplicate, normalized for each individual donor to the amount of IP10 protein induced by IFNg treatment alone (100%). *p  evidence from other work suggests that 0.05, **p  0.005. (B) Macrophages were treated with the Syk inhibitor Bay 61–3606 (10 mM) for this may be a hallmark of inhibitory 1 hour prior to plating on IC-treated or untreated plates for one hour. Cells were then treated with immune complex signaling through 10 ng/mL IFNg or LPS for 1.5 hours followed by RNA isolation, reverse transcription, and analysis of FcgRI,37 especially in monocyte-derived IP10 mRNA levels by real-time polymerase chain reaction. (C) Supernatants of these macrophages color tissue resident macrophages. lected after 20 hours were analyzed for RANTES and IL-12 p40 by multiplex. Results are the mean § SEM of 3 experiments with separate donors, performed in triplicate. The data for each donor are norFcgRI is often ignored when general malized to the amount of protein induced by LPS treatment alone (100%). (D) Macrophages incustatements are made about antibodybated with LPS for 20 hours were stained with Alexa 647 mouse anti-human CD163 or PE-Cy5 dependent cellular effector function. conjugated mouse anti-human CD80. Results are the mean § SEM of 3 experiments with separate One reason for this may be the outdated donors. perception that since the high-affinity FcgRI can bind monovalent IgG, it will always be occupied due to the high circulating concentration of interaction of the Fc region of the therapeutic mAb with the monovalent IgG and thus unavailable for immune complex bind- FcgR on the human macrophage. ing. In truth, the off-rate of monomeric IgG1, the subclass that Phagocytosis of immune complexes as a result of FcgRI binds FcgRI with the highest affinity, is such that it is constantly engagement on macrophages is also known to be a trigger leaving the receptor available for higher-avidity interactions with toward adoption of an “alternatively activated” or M2 phenoimmune complex.38 In this regard, it has been shown that type42 that results in reduction of inflammatory cytokine and immune complexes are able to effectively compete for FcgRI chemokine levels and induction of immunomodulatory ILbinding and signaling in the presence of physiologically-relevant 10. This might be important not only for antibodies designed concentrations of IgG.39 It has also been repeatedly demonstrated simply to neutralize inflammatory signaling pathways. It has that IC binding to macrophages induces IL-10 secretion and can been shown that M2 macrophages, especially those induced induce a regulatory macrophage phenotype,18,40 with the data by IL-10 treatment, are superior to classically activated M1 suggesting that these effects occur downstream of FcgRI.37 A macrophages for the ingestion of Rituximab-coated cancer recent investigation of TNF antagonists, 2 human antibodies, an cells.43 There is also a growing appreciation for the role of Fc-receptor fusion, and a PEGylated Fab’, demonstrated that the macrophages in the phagocytosis of therapeutic antibodytherapeutics containing a fully functional human Fc region coated tumor cells,44,45 and if the M2 fate is superior for this induced the formation of regulatory macrophages and inhibited task, Fc binding to FcgRI may encourage its development. T cell proliferation in a mixed-lymphocyte reaction to a greater Taken together, these data suggest that the phagocytic effecdegree.41 All of these observed responses would depend on the tor function (or “frustrated phagocytosis,” in the case of our

www.landesbioscience.com

mAbs

1381

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

IC model) is the normal consequence of CD64-dependent FcRg-chain ITAM stimulation, which produces an alternatively activated- or M2- fate in macrophages, the only cell that naturally expresses FcgRI. Human IgG4 antibodies (either soluble or as immune complexes) have never been previously demonstrated to transduce a signal through any Fcg receptor, despite reports of their relatively high affinity for FcgRI.46 Our data demonstrates that immobilized (or “complexed”) IgG4 can effectively inhibit IFNg-stimulated IP10 production (Fig. 2) or inflammatory events downstream of LPS treatment (Fig. 4), and that these phenomena are dependent on the activation of FcgRI through Syk (Fig. 4). Although the incomplete ability of the FcgRI blocking F(ab’)2 fragments (clone 10.1) to prevent the action of IgG4 leaves room for the possibility of involvement of another receptor, a more likely explanation would be that these results are due to an artifact of this particular antibody.47 The similarly surprising finding that glycosylation is not required for induction of this inhibitory pathway, either by IgG1 or IgG4, is potentially explained by recent findings following careful investigation of the size- and glycosylation-dependence of FcgRI binding by IC: it was demonstrated that for IgG1 IC binding to FcgRI, glycosylation is relatively unimportant, whereas for IgG4, the larger the IC, the less important Fc glycosylation was for FcgRI binding.11 Consistent with our general findings regarding IgG4, a recent report showed that a human IgG4 form of a therapeutic antiCD20 antibody is capable of B cell depletion in a humanized mouse model, whereas F(ab’)2 fragments of the same antibody are not.48 Data from this work also suggest that macrophages are the critical effector cell for B cell depletion in the bone marrow. IgG4 is often selected as the backbone of choice for those therapeutic antibodies designed to neutralize an inflammatory mediator or its receptor without unwanted activation of the immune system.4 Our data suggest that therapeutic IgG4s could in fact actively contribute to the inhibition of inflammation through Fcdependent signaling. The presumed inability of IgG4 to invoke any FcgR-dependent signal has likely been based both on an incomplete understanding of its affinity for FcgRI and the functionally monovalent nature of endogenous IgG4.49,50 However, updates describing its high affinity to FcgRI are accompanied by new crystal structures of the IgG4 Fc that suggest the possibility of a unique additional interaction site with this receptor. 51,52 Additionally, although natural IgG4s undergo Fab-arm exchange that renders them hetero-bivalent in vivo, therapeutic IgG4s are now frequently stabilized by mutations that assure their integrity during manufacture and subsequent administration. These new findings highlight how therapeutic IgG4s could presumably form immune complexes with an increased ability for functional multivalent FcgRI engagement, perhaps nearly as capable as IgG1s. Growing evidence suggests that a major outcome of FcgRIdependent immune complex signaling in macrophages is antiinflammatory,53 and this new evidence that complexed IgG4 may be capable of inducing an M2 macrophage phenotype in vivo should be more thoroughly investigated. Such studies would improve understanding of all Fc-mediated contributions to therapeutic antibody efficacy.

1382

Materials and Methods Isolation and treatment of human monocytes Human peripheral blood monocytes were obtained from healthy volunteers by leukophoresis and further purified from mononuclear cells by Ficoll-Hypaque sedimentation, followed by countercurrent centrifugal elutriation, as described previously.16 The use of human volunteers for this purpose was reviewed and approved by both the National Institutes of Health and Food and Drug Administration internal review boards. Monocytes used for experiments were allowed to adhere for 1 hr in serum-free Dulbecco modified Eagle medium (DMEM; Lonza, #12–733F) onto untreated plates or plates coated with serum-derived human IgG or human myeloma-derived IgG1 or IgG4 (Athens Research and Technology, #16-16-090707 (IgG), #16-16-090707-1M (IgG1) or # 16-16-090707-4M (IgG4)) at 50 mg/ml. Alternatively, human monocyte-derived macrophages were cultured by plating freshly elutriated monocytes in DMEM supplemented with 10 mg/mL gentamicin (Invitrogen, #15750-060) at 6 £ 106/ 2 mL/well in 6-well plates. After 4 to 6 hours, heat-inactivated fetal bovine serum (Valley Biomedical, #BS3032HI) was added to 10%, and monocytes were cultured for 7 d to enable differentiation into mature macrophages. Macrophages were then removed using Accutase (Innovative Cell Technologies, #AT104) and allowed to recover in DMEM with 10% FBS for 3h at 37 C. Blocking F(ab’)2 (Ancell, #216–520 (a-FcgRI, clone 10.1), #181–520 (a-FcgRII, clone 7.3), # 165–520 (a-FcgRIII, clone 3G8) were applied to macrophages at 10 mg/mL each for 450 on ice. Where indicated, cells were treated with Syk inhibitor Bay 61–3606 (Santa Cruz, #4152655) at 10 mM for 1 hour in suspension. Cells were then plated on IC-coated plates and or uncoated plates for 1 hour and subsequently treated with human recombinant IFNg (kindly provided by Genentech, Inc., South San Francisco, CA) or ultrapurified LPS from E. coli K12 (Invivogen, #tlrl-peklps) at a concentration of 10 ng/ml. Antibody deglycosylation Enzymatic deglycosylation of serum-derived human IgG and myeloma-derived IgG1 and IgG4 was performed as adapted from the literature.54 Briefly, 5 ml (2500 units) PNGase F (New England Biolabs #P07045) was added to 100 ml IgG at 20 mg/ ml in 50 mM Tris, 150 mM NaCl pH 8.5 and incubated at 37 C for 42 h. Untreated control antibody was incubated in this mixture in the absence of PNGase under the same conditions. The samples were then added to 200 ml 0.01 M HEPES pH 7.4, 0.15 M NaCl, 0.005% v/v Surfactant P20 (HBS-P buffer) and 200 ml pre-washed Protein A/G beads (Santa Cruz, SC2003). Samples were rotated for 30 min at RT, spun down at 2500 rpm for 2 min, and washed 2£ with HBS-P. IgG was then sequentially eluted with 10 mM glycine pH 3.0, 2.5, and 2.0, each for 5 min at RT, and 1M Tris pH 8 was added to eluates to bring them to pH 7.4. RNA isolation and analysis Total RNA was isolated from cells using the RNeasy kit (Qiagen, #74106) according to the manufacturer’s instructions.

mAbs

Volume 6 Issue 6

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

Reverse transcription with random hexamers was conducted using the SuperScriptTM II Reverse Transcriptase (Invitrogen, #18064-014) as per the manufacturer’s established protocols. Two micrograms of RNA per sample were used and a control parallel reaction was included where no reverse transcriptase was added. One-fortieth of the reverse transcriptase reaction material was analyzed by Quantitative RT-PCR using SYBR Green and detection with the real-time PCR System 7900 (Applied Biosystems). Quantitative RT-PCR primers were as follows: for IFNinducible protein 10 (IP-10), 50 -GGAACCTCCAGTCTCAGCACC-30 and 50 -CAGCCTCTGTGTGGTCCATCC-30 , and for b-actin, 50 - TCGTCGACAACGGCTCCGGCATGTGC-30 and 50 -TTCTGCAGGGAGGAGCTGGAAGCAGC-30 . PCR conditions (45 cycles) included 15 s at 95 C, 30 s at 60 C, and 30 s at 72 C. The threshold cycle number for hIP-10 was normalized to that of the b-actin transcripts and then set to a percentage of the maximum transcription detected in samples treated with 10 ng/ml IFNg. Relative fold increases were determined using the 2-DDCT method as described elsewhere.55 Two-tailed Student’s T tests were performed where statisticallysignificant differences are noted. Multiplex analysis After indicated treatments, supernatants were clarified and diluted 10 to 20 times such that values obtained fit in the linear range of the standard curve for each cytokine reported. Assays were performed in duplicate using multiplex bead-based kits for the indicated human cytokines and chemokines (Panomics, #PC1008) per manufacturers’ instructions. Fluorescence of beads was measured using a Bioplex 200 analyzer (Bio-Rad). Cytokine data analysis was performed using the BioPlex Manager software (Bio-Rad). Concentrations were determined with a 5-parametric logistic nonlinear regression curve-fitting algorithm, and results were entered into GraphPad Prism 5 software (GraphPad Software) for plotting and statistical analysis. Flow cytometry For surface marker staining, cells were detached using Accutase (Innovative Cell Technologies, #3155493), washed, and References 1. Okun E, Mattson MP, Arumugam TV. Involvement of Fc receptors in disorders of the central nervous system. Neuromol Med 2010; 12:164-78; PMID:19844812; http://dx.doi.org/10.1007/ s12017-009-8099-5 2. Takai T. Fc receptors and their role in immune regulation and autoimmunity. J Clin Immunol 2005; 25:118; PMID:15742153; http://dx.doi.org/10.1007/ s10875-005-0353-8 3. Nimmerjahn F, Ravetch JV. Fcgamma receptors as regulators of immune responses. Nat Rev Immunol 2008; 8:34-47; PMID:18064051; http://dx.doi.org/10.1038/ nri2206 4. Jiang XR, Song A, Bergelson S, Arroll T, Parekh B, May K, Chung S, Strouse R, Mire-Sluis A, Schenerman M. Advances in the assessment and control of the effector functions of therapeutic antibodies. Nat Rev Drug Disc 2011; 10:101-11; PMID:21283105; http://dx. doi.org/10.1038/nrd3365 5. Chan AC, Carter PJ. Therapeutic antibodies for autoimmunity and inflammation. Nat Rev Immunol 2010;

www.landesbioscience.com

6.

7.

8.

9.

10.

stained with Alexa 647 mouse anti-human CD163 (BD; # 562669), PE-Cy5 conjugated mouse anti-human CD80 (# 559370), or APC conjugated mouse anti-human CD105 (#562408) for 30 minutes at 4 C. Samples were resuspended in phosphate-buffered saline containing 1.0% paraformaldehyde, and analyzed on a FACSCalibur flow cytometer (BD Biosciences), using a gate set for single, intact macrophages.

Disclosure of Potential Conflicts of Interest

No potential conflicts of interest were disclosed.

Acknowledgments

This project was supported in part by an appointment of DAH and AGM to the Research Participation Program at the Center for Biologics Evaluation and Research administered by the Oak Ridge Institute for Science and Education through an interagency agreement between the U.S. Department of Energy and the U.S. Food and Drug Administration. We thank Silvia Bacot and members of Dr. Kathleen Clouse’s lab for elutriation of human monocytes and other technical support, as well as Drs. Peter Adams and Tao Xie for critical review of the manuscript. The findings and conclusions in this paper have not been formally disseminated by the U.S. Food and Drug Administration and should not be construed to represent any Agency determination or policy.

Authors’ Contributions

JFAS designed the experiments, performed the statistical analyses, helped analyze and interpret the data, and helped write the manuscript. DAH and AGM performed most of the experiments, and helped analyze the data. GHB helped analyze and interpret the data. GMF helped design the experiments, analyzed and interpreted the data, and wrote the manuscript.

10:301-16; PMID:20414204; http://dx.doi.org/ 10.1038/nri2761 Powell MS, Hogarth PM. Fc receptors. Adv Exp Med Biol 2008; 640:22-34; PMID:19065781; http://dx.doi. org/10.1007/978-0-387-09789-3_3 Li F, Smith P, Ravetch JV. Inhibitory Fcgamma receptor is required for the maintenance of tolerance through distinct mechanisms. J Immunol 2014; 192:3021-8; PMID:24563255; http://dx.doi.org/10.4049/ jimmunol.1302934 Ravetch JV, Bolland S. IgG Fc receptors. Ann Rev Immunol 2001; 19:275-90; PMID:11244038; http:// dx.doi.org/10.1146/annurev.immunol.19.1.275 Blank U, Launay P, Benhamou M, Monteiro RC. Inhibitory ITAMs as novel regulators of immunity. Immunol Rev 2009; 232:59-71; PMID:19909356; http://dx.doi.org/10.1111/j.1600-065X.2009.00832.x Boekhoudt GH, Frazier-Jessen MR, Feldman GM. Immune complexes suppress IFN-gamma signaling by activation of the FcgammaRI pathway. J Leukocyte Biol 2007; 81:1086-92; PMID:17227821; http://dx. doi.org/10.1189/jlb.0906543

mAbs

11. Lux A, Yu X, Scanlan CN, Nimmerjahn F. Impact of immune complex size and glycosylation on IgG binding to human FcgammaRs. J Immunol 2013; 190:4315-23; PMID:23509345; http://dx.doi.org/ 10.4049/jimmunol.1200501 12. Davies LC, Jenkins SJ, Allen JE, Taylor PR. Tissue-resident macrophages. Nat Immunol 2013; 14:986-95; PMID:24048120; http://dx.doi.org/10.1038/ni.2705 13. Shi C, Pamer EG. Monocyte recruitment during infection and inflammation. Nat Rev Immunol 2011; 11:762-74; PMID:21984070; http://dx.doi.org/ 10.1038/nri3070 14. Medzhitov R. Origin and physiological roles of inflammation. Nature 2008; 454:428-35; PMID:18650913; http://dx.doi.org/10.1038/nature07201 15. Hamilton JA, Tak PP. The dynamics of macrophage lineage populations in inflammatory and autoimmune diseases. Arthritis Rheum 2009; 60:1210-21; PMID: 19404968; http://dx.doi.org/10.1002/art.24505 16. Feldman GM, Chuang EJ, Finbloom DS. IgG immune complexes inhibit IFN-gamma-induced transcription of the Fc gamma RI gene in human monocytes by

1383

17.

18.

19.

Downloaded by [University of Manitoba Libraries] at 11:04 07 September 2015

20.

21.

22.

23.

24.

25.

26.

27.

28.

29.

preventing the tyrosine phosphorylation of the p91 (Stat1) transcription factor. J Immunol 1995; 154:31825; PMID:7995951 Luster AD, Unkeless JC, Ravetch JV. Gamma-interferon transcriptionally regulates an early-response gene containing homology to platelet proteins. Nature 1985; 315:672-6; PMID:3925348; http://dx.doi.org/ 10.1038/315672a0 Anderson CF, Gerber JS, Mosser DM. Modulating macrophage function with IgG immune complexes. J Endotoxin Res 2002; 8:477-81; PMID:12697094; http://dx.doi.org/10.1177/09680519020080060501 Halstead SB, Mahalingam S, Marovich MA, Ubol S, Mosser DM. Intrinsic antibody-dependent enhancement of microbial infection in macrophages: disease regulation by immune complexes. Lancet Infect Dis 2010; 10:712-22; PMID:20883967; http://dx.doi.org/ 10.1016/S1473-3099(10)70166-3 Miles SA, Conrad SM, Alves RG, Jeronimo SM, Mosser DM. A role for IgG immune complexes during infection with the intracellular pathogen Leishmania. J Exp Med 2005; 201:747-54; PMID:15753208; http:// dx.doi.org/10.1084/jem.20041470 Jefferis R, Lund J. Interaction sites on human IgG-Fc for FcgammaR: current models. Immunol Lett 2002; 82:57-65; PMID:12008035; http://dx.doi.org/ 10.1016/S0165-2478(02)00019-6 Canfield SM, Morrison SL. The binding affinity of human IgG for its high affinity Fc receptor is determined by multiple amino acids in the CH2 domain and is modulated by the hinge region. J Exp Med 1991; 173:1483-91; PMID:1827828; http://dx.doi. org/10.1084/jem.173.6.1483 Stone JH, Zen Y, Deshpande V. IgG4-related disease. New Engl J Med 2012; 366:539-51; PMID:22316447; http://dx.doi.org/10.1056/NEJMra1104650 Krapp S, Mimura Y, Jefferis R, Huber R, Sondermann P. Structural analysis of human IgG-Fc glycoforms reveals a correlation between glycosylation and structural integrity. J Mol Biol 2003; 325:979-89; PMID:12527303; http://dx.doi.org/10.1016/S00222836(02)01250-0 Dunn-Siegrist I, Leger O, Daubeuf B, Poitevin Y, Depis F, Herren S, Kosco-Vilbois M, Dean Y, Pugin J, Elson G. Pivotal involvement of Fcgamma receptor IIA in the neutralization of lipopolysaccharide signaling via a potent novel anti-TLR4 monoclonal antibody 15C1. J Biol Chem 2007; 282:34817-27; PMID:17921137; http://dx.doi.org/10.1074/jbc.M706440200 Pinheiro da Silva F, Aloulou M, Skurnik D, Benhamou M, Andremont A, Velasco IT, Chiamolera M, Verbeek JS, Launay P, Monteiro RC. CD16 promotes Escherichia coli sepsis through an FcR gamma inhibitory pathway that prevents phagocytosis and facilitates inflammation. Nat Med 2007; 13:1368-74; PMID:17934470; http://dx.doi.org/10.1038/nm1665 Ballow M. The IgG molecule as a biological immune response modifier: mechanisms of action of intravenous immune serum globulin in autoimmune and inflammatory disorders. J Allergy Clin Immun 2011; 127:31523; PMID:21185071; http://dx.doi.org/10.1016/j. jaci.2010.10.030 Schwab I, Nimmerjahn F. Intravenous immunoglobulin therapy: how does IgG modulate the immune system? Nat Rev Immunol 2013; 13:176-89; PMID:23411799; http://dx.doi.org/10.1038/nri3401 Anthony RM, Ravetch JV. A novel role for the IgG Fc glycan: the anti-inflammatory activity of sialylated IgG Fcs. J Clin Immunol 2010; 30 Suppl 1:S9-14;

1384

30.

31.

32.

33.

34.

35.

36.

37.

38.

39.

40.

41.

42.

43.

PMID:20480216; http://dx.doi.org/10.1007/s10875010-9405-6 Takai T, Ono M, Hikida M, Ohmori H, Ravetch JV. Augmented humoral and anaphylactic responses in Fc gamma RII-deficient mice. Nature 1996; 379:346-9; PMID:8552190; http://dx.doi.org/10.1038/379346a0 Junghans RP, Anderson CL. The protection receptor for IgG catabolism is the beta2-microglobulin-containing neonatal intestinal transport receptor. Proc Nat Acad Sci U S A 1996; 93:5512-6; PMID:8643606; http://dx.doi.org/10.1073/pnas.93.11.5512 Ghetie V, Hubbard JG, Kim JK, Tsen MF, Lee Y, Ward ES. Abnormally short serum half-lives of IgG in beta 2-microglobulin-deficient mice. Eur J immunol 1996; 26:690-6; PMID:8605939; http://dx.doi.org/ 10.1002/eji.1830260327 Ivashkiv LB. How ITAMs Inhibit Signaling. Sci Signaling 2011; 4:pe20; PMID:21505184; http://dx.doi.org/ 10.1126/scisignal.2001917 Martinez FO, Gordon S. The M1 and M2 paradigm of macrophage activation: time for reassessment. F1000prime Rep 2014; 6:13; PMID:24669294; http:// dx.doi.org/10.12703/P6-13 Ambarus CA, Santegoets KC, van Bon L, Wenink MH, Tak PP, Radstake TR, Baeten DL. Soluble immune complexes shift the TLR-induced cytokine production of distinct polarized human macrophage subsets towards IL-10. PloS One 2012; 7:e35994; PMID:22563430; http://dx.doi.org/10.1371/journal. pone.0035994 Bezbradica JS, Rosenstein RK, Demarco RA, Brodsky I, Medzhitov R. A role for the ITAM signaling module in specifying cytokine-receptor functions. Nat Immunol 2014; 15:333-42; PMID:24608040; http://dx.doi. org/10.1038/ni.2845 Sutterwala FS, Noel GJ, Salgame P, Mosser DM. Reversal of proinflammatory responses by ligating the macrophage Fc gamma receptor type I. J Exp Med 1998; 188:217-22; PMID:9653099; http://dx.doi.org/ 10.1084/jem.188.1.217 Mancardi DA, Iannascoli B, Hoos S, England P, Daeron M, Bruhns P. FcgammaRIV is a mouse IgE receptor that resembles macrophage FcepsilonRI in humans and promotes IgE-induced lung inflammation. J Clin Invest 2008; 118:3738-50; PMID:18949059; http://dx.doi.org/10.1172/JCI36452 van der Poel CE, Karssemeijer RA, Boross P, van der Linden JA, Blokland M, de Winkel JGJV, Blokland M, van de Winkel JG, Leusen JH. Cytokine-induced immune complex binding to the high-affinity IgG receptor, Fc gamma RI, in the presence of monomeric IgG. Blood 2010; 116:5327-33; PMID:20805361; http://dx.doi.org/10.1182/blood-2010-04-280214 Gallo P, Goncalves R, Mosser DM. The influence of IgG density and macrophage Fc (gamma) receptor cross-linking on phagocytosis and IL-10 production. Immunol Lett 2010; 133:70-7; PMID:20670655; http://dx.doi.org/10.1016/j.imlet.2010.07.004 Vos AC, Wildenberg ME, Duijvestein M, Verhaar AP, van den Brink GR, Hommes DW. Anti-tumor necrosis factor-alpha antibodies induce regulatory macrophages in an Fc region-dependent manner. Gastroenterology 2011; 140:221-30; PMID:20955706; http://dx.doi. org/10.1053/j.gastro.2010.10.008 Sica A, Mantovani A. Macrophage plasticity and polarization: in vivo veritas. J Clin Invest 2012; 122:787-95; PMID:22378047; http://dx.doi.org/10.1172/JCI59643 Leidi M, Gotti E, Bologna L, Miranda E, Rimoldi M, Sica A, Roncalli M, Palumbo GA, Introna M, Golay J.

mAbs

44.

45.

46.

47.

48.

49.

50.

51.

52.

53.

54.

55.

M2 macrophages phagocytose rituximab-opsonized leukemic targets more efficiently than m1 cells in vitro. J Immunol 2009; 182:4415-22; PMID:19299742; http://dx.doi.org/10.4049/jimmunol.0713732 Braster R, O’Toole T, van Egmond M. Myeloid cells as effector cells for monoclonal antibody therapy of cancer. Methods 2014; 65:28-37; PMID:23811299; http://dx.doi.org/10.1016/j.ymeth.2013.06.020 Gul N, Babes L, Siegmund K, Korthouwer R, Bogels M, Braster R, Vidarsson G, ten Hagen TL, Kubes P, van Egmond M. Macrophages eliminate circulating tumor cells after monoclonal antibody therapy. J Clin Invest 2014; 124:812-23; PMID:24430180; http://dx. doi.org/10.1172/JCI66776 Lu J, Ellsworth JL, Hamacher N, Oak SW, Sun PD. Crystal structure of Fcgamma receptor I and its implication in high affinity gamma-immunoglobulin binding. J Biol Chem 2011; 286:40608-13; PMID: 21965667; http://dx.doi.org/10.1074/jbc. M111.257550 Dougherty GJ, Selvendran Y, Murdoch S, Palmer DG, Hogg N. The human mononuclear phagocyte highaffinity Fc receptor, Fcri, defined by a monoclonal-antibody, 10.1. Eur J Immunol 1987; 17:1453-9; PMID:3500057; http://dx.doi.org/10.1002/ eji.1830171011 Lux A, Seeling M, Baerenwaldt A, Lehmann B, Schwab I, Repp R, Meidenbauer N, Mackensen A, Hartmann A, Heidkamp G, et al. A humanized mouse identifies the bone marrow as a niche with low therapeutic IgG activity. Cell Rep 2014; 7:236-48; PMID:24685130; http://dx.doi.org/10.1016/j.celrep.2014.02.041 Aalberse RC, Stapel SO, Schuurman J, Rispens T. Immunoglobulin G4: an odd antibody. Clin Exp Allergy: J Brit Soc Allergy Clin Immunol 2009; 39:469-77; PMID:19222496; http://dx.doi.org/ 10.1111/j.1365-2222.2009.03207.x Nirula A, Glaser SM, Kalled SL, Taylor FR. What is IgG4? A review of the biology of a unique immunoglobulin subtype. Curr Opin Rheumatol 2011; 23:119-24; PMID:21124094; http://dx.doi.org/ 10.1097/BOR.0b013e3283412fd4 Davies AM, Jefferis R, Sutton BJ. Crystal structure of deglycosylated human IgG4-Fc. Mol Immunol 2014; 62:46-53; PMID:24956411; http://dx.doi.org/ 10.1016/j.molimm.2014.05.015 Davies AM, Rispens T, Ooijevaar-de Heer P, Gould HJ, Jefferis R, Aalberse RC, Sutton BJ. Structural determinants of unique properties of human IgG4-Fc. J Mol Biol 2014; 426:630-44; PMID:24211234; http://dx.doi.org/10.1016/j.jmb.2013.10.039 Gmyrek GB, Akilesh HM, Graham DB, Fuchs A, Yang L, Miller MJ, Sandoval GJ, Sheehan KC, Schreiber RD, Diamond MS, et al. Loss of DAP12 and FcRgamma drives exaggerated IL-12 production and CD8(C) T cell response by CCR2(C) Mo-DCs. PloS One 2013; 8:e76145; PMID:24155889; http://dx.doi. org/10.1371/journal.pone.0076145 Kaneko Y, Nimmerjahn F, Ravetch JV. Anti-inflammatory activity of immunoglobulin G resulting from Fc sialylation. Science 2006; 313:670-3; PMID: 16888140; http://dx.doi.org/10.1126/science.1129594 Winer J, Jung CK, Shackel I, Williams PM. Development and validation of real-time quantitative reverse transcriptase-polymerase chain reaction for monitoring gene expression in cardiac myocytes in vitro. Anal Biochem 1999; 270:41-9; PMID:10328763; http://dx.doi. org/10.1006/abio.1999.4085

Volume 6 Issue 6

IgG4 can induce an M2-like phenotype in human monocyte-derived macrophages through FcγRI.

Antibodies evoke cellular responses through the binding of their Fc region to Fc receptors, most of which contain immunoreceptor tyrosine-based activa...
369KB Sizes 3 Downloads 4 Views