You are on page 1of 10

mAbs

ISSN: 1942-0862 (Print) 1942-0870 (Online) Journal homepage: http://www.tandfonline.com/loi/kmab20

Monoclonal antibodies directed against human


FcRn and their applications

Gregory J. Christianson, Victor Z. Sun, Shreeram Akilesh, Emanuele


Pesavento, Gabriele Proetzel & Derry C. Roopenian

To cite this article: Gregory J. Christianson, Victor Z. Sun, Shreeram Akilesh, Emanuele
Pesavento, Gabriele Proetzel & Derry C. Roopenian (2012) Monoclonal antibodies directed
against human FcRn and their applications, mAbs, 4:2, 208-216, DOI: 10.4161/mabs.4.2.19397

To link to this article: http://dx.doi.org/10.4161/mabs.4.2.19397

Published online: 01 Mar 2012.

Submit your article to this journal

Article views: 271

View related articles

Full Terms & Conditions of access and use can be found at


http://www.tandfonline.com/action/journalInformation?journalCode=kmab20

Download by: [121.207.210.105] Date: 04 October 2015, At: 20:58


Report

mAbs 4:2, 208-216; March/April 2012; © 2012 Landes Bioscience

Monoclonal antibodies directed


against human FcRn and their applications
Gregory J. Christianson, Victor Z. Sun, Shreeram Akilesh,† Emanuele Pesavento,‡ Gabriele Proetzel and Derry C. Roopenian*
The Jackson Laboratory; Bar Harbor, ME USA; †Current Address: Barnes-Jewish Hospital; St. Louis, MO USA; ‡Current Address: VIB Department of Molecular and Cellular
Interactions; Laboratory for Cellular and Molecular Immunology; Vrije Universiteit; Brussels, Belgium

Key words: FcRn, IgG, monoclonal antibody, albumin, therapy

Abbreviations: MHC, major histocompatibility complex; IgG, immunoglobulin G (h for human, m for mouse); FcRn, Fc receptor

©2013 Landes Bioscience. Do not distribute.


neonatal (h for human, m for mouse); mAb, monoclonal antibody; β2M, beta-2 microglobulin; HSA, human serum albumin;
GFP, enhanced green fluorescent protein; IF, immunofluorescence

The MHC class I-like Fc receptor (FcRn) is an intracellular trafficking Fc receptor that is uniquely responsible for the extended
serum half-life of antibodies of the IgG subclass and their ability to transport across cellular barriers. By performing
Downloaded by [121.207.210.105] at 20:58 04 October 2015

these functions, FcRn affects numerous facets of antibody biology and pathobiology. Its critical role in controlling IgG
pharmacokinetics has been leveraged for the design of therapeutic antibodies and related biologics. FcRn also traffics
serum albumin and is responsible for the enhanced pharmacokinetic properties of albumin-conjugated therapeutics.
The understanding of FcRn and its therapeutic applications has been limited by a paucity of reliable serological reagents
against human FcRn. Here, we describe the properties of a new panel of highly specific monoclonal antibodies (mAbs)
directed against human FcRn with diverse epitope specificities. We show that this antibody panel can be used to study
the tissue expression pattern of human FcRn, to selectively block IgG and serum albumin binding to human FcRn in vitro
and to inhibit FcRn function in vivo. This mAb panel provides a powerful resource for probing the biology of human FcRn
and for the evaluation of therapeutic FcRn blockade strategies.

Introduction and bioavailability of IgG monoclonal antibodies (mAbs) and


IgG-Fc fusion proteins and albumin conjugated protein thera-
Although originally referred to as the neonatal Fc receptor, FcRn peutics.5,17,18 While the biological and therapeutic importance of
influences IgG serum levels and tissue distribution at all stages FcRn’s varied and expanding functional attributes are increas-
of life.1-5 This divergent member of the MHC class I family mol- ingly documented, major gaps remain in the understanding of
ecule is a heterodimer composed of an evolutionally distinct the cell biology of FcRn, its tissue sites of action, and methods to
α-chain in complex with the β2-microglobulin (β2M) light chain exploit this biology for therapeutic purposes.
that is common to most other class I molecules. FcRn is an intra- Of particular relevance is the need for reliable serological
cellular trafficking, integral membrane Fc receptor for IgG. It reagents for detection of human FcRn (hFcRn) and for prob-
resides primarily in the early acidic endosomes where it captures ing its biology in normal and pathological states. This report
endocytosed IgG by binding to the Fc region only at a low pH.6-8 describes the generation, characterization and applications of a
FcRn rescues bound IgG from degradation in the lysosomal com- part of mAbs with varied epitope specificities directed against
partment and transports its ligand to the cell surface for release human (h)FcRn. Our data demonstrate that the mAbs can read-
at neutral extracellular pH. Through this mechanism, FcRn is ily detect hFcRn by flow cytometry and on tissue sections, which
responsible for the long serum half-life of IgG.9 FcRn transports makes them valuable tools for defining the sites of hFcRn expres-
IgG across the tight epithelial barriers of the endothelium and sion. Furthermore, the ability of certain mAbs to differentially
mucosa, thus influencing its bioavailability.7,10 In antigen pre- block hIgG or human serum albumin binding supports a model
senting cells, FcRn controls the presentation of antigens in IgG of distinct binding sites for these ligands to hFcRn.5,16 Finally,
immune complexes to T cells11,12 and IgG-Fc fusion protein vac- treatment of hFcRn transgenic mice with a hIgG blocking mAb
cines.13,14 FcRn also controls serum albumin homeostasis by an provides a proof-of-principle that anti-hFcRn mAbs can be
analogous trafficking mechanism as for IgG.5,15,16 These properties applied therapeutically to specifically control the serum persis-
have proven to be critical for maximizing the pharmacokinetics tence of human IgG in vivo.

*Correspondence to: Derry C. Roopenian; Email: derry.roopenian@jax.org


Submitted: 12/26/11; Revised: 01/17/12; Accepted: 01/18/12
http://dx.doi.org/10.4161/mabs.4.2.19397

208 mAbs Volume 4 Issue 2


Report Report

©2013 Landes Bioscience. Do not distribute.


Downloaded by [121.207.210.105] at 20:58 04 October 2015

Figure 1. hFcRn binding and blocking activity of anti-hFcRn antisera. (A) Representation of the construct design lacking the cytoplasmic targeting
domain (left). Confocal images of HeLa cells transiently transfected with the hFcRn-GFP after incubation with hIgG at the indicated pH, fixed and
stained with goat anti-hIgGAF568 (right). (B) Flow cytometric data of 293hFcRn-GFP cells incubated with hIgGAF647 at pH 6.0 (left) or 7.2 (right). (C) Representa-
tive results from evaluation of antisera from mice primed with hFcRn. Binding of the antisera to hFcRn-GFP was detected with goat anti-mouse IgG-PE
at pH 7.2 (left). Functional blockade of hIgG binding to hFcRn was determined by the ability to inhibit the binding of hIgGAF647 at pH 6.0 (right). Values
within scatter plots represent the AF647 mean fluorescence intensity (MFI) of each hFcRn-GFP+-gated cell population. Data are representative of at
least two experiments.

Results rather than through the Fc region. Next, to determine whether


the antisera could inhibit the binding of hIgG, we assessed their
Screening assay validation and antisera generation. We first ability to compete with Alexa Fluor® 647-labeled hIgG (hIg-
established a flow cytometry-based assay suitable for detecting GAF647) for binding to GFP-tagged human FcRn expressed on
antibody activity specific for human and mouse FcRn and for the the surface of 293 cells (293hFcRn-GFP) cells at pH 6.0. The antisera
assessment of the binding and functional blockade of hIgG bind- showed different binding and blocking properties. For example,
ing. An enhanced green fluorescent protein (GFP) was spliced antiserum 26,162 bound hFcRn at pH 7.2 and blocked hIgG AF647
between the signal sequences and the N-terminus of FcRn binding at pH 6.0 (Fig. 1C, top). In contrast, antiserum 26898
constructs with truncated cytoplasmic, endosomal targeting bound, but did not block, hIgGAF647 binding to hFcRn (Fig. 1C,
domains (Fig. 1A). This truncation resulted in robust expression middle). Lastly, some antisera, such as antiserum 26902, showed
of the hFcRn-GFP chimeric proteins on the plasma membrane neither hFcRn binding nor blocking activity (Fig. 1C, bottom).
as first validated by confocal immunofluorescence (IF) imaging Generation of anti-FcRn antibody-producing hybridomas.
of transiently transfected HeLa cells (Fig. 1A) and then by flow Mice with antisera showing hFcRn binding or blocking activity
cytometric analysis of HEK293 cells stably transfected with the were used for hybridoma generation. Supernatants from hybrid-
same construct (Fig. 1B). Epitope tagging with GFP did not omas were assayed for their binding to hFcRn using 293hFcRn-GFP
alter the normal pH-dependent binding of hFcRn to hIgG. The cells in a cellular enzyme-linked immunosorbent assay (ELISA),
hFcRn-GFP fusion protein bound hIgG at acidic pH 6 but not and those showing activity against hFcRn in this assay were sub-
neutral pH (Fig. 1A and B). Similar acidic pH-dependent bind- cloned to ensure stable antibody production. mAbs from these
ing of hIgG was observed for HeLa and 293 cells transfected with hybridomas were purified, conjugated to Alexa Fluor 647, and
the mFcRn-GFP construct (data not shown). tested in the previously described flow cytometry assay for their
To generate anti-hFcRn antisera, we immunized B6 wild-type ability to bind at pH 7.2 to human or mouse FcRn. In total, nine
(WT) and FcRn-/- mice with splenocytes from hFcRn transgenic unique hybridoma clones were established. Only one, DVN24,
mice. Plasma samples from immunized mice were first screened showed appreciable cross-reactivity to mFcRn (Fig. 2A). The
for their ability to bind hFcRn at a neutral pH to ensure that relative binding affinity of each mAb for hFcRn was estimated
the antibodies bound hFcRn by their antigen binding domains by its ability to bind 293hFcRn-GFP cells by quantifying the mean

www.landesbioscience.com mAbs 209


©2013 Landes Bioscience. Do not distribute.
Downloaded by [121.207.210.105] at 20:58 04 October 2015

Figure 2. Anti-hFcRn mAb specificities and relative affinities. 293hFcRn-GFP, 293mFcRn-GFP, or control 293 cells were incubated with AF647-labeled anti-hFcRn
mAbs (A) at a fixed concentration of 200 ng/106 cells to determine specificity or (B) with a range of concentrations to assess relative mAb affinities for
hFcRn. Values within scatter plots represent the AF647 MFIs of each hFcRn-EGFP+-gated cell population. Data are representative of at least two experi-
ments.

Table 1. Competitive binding of anti-hFcRn mAbsa


BLOCK DVN1 ADM11 DVN21 DVN22 DVN23 DVN24 ADM31 ADM32
DVN1 b
100 33 32 17 55 0 85 67
ADM11 47 100 21 8 13 8 58 48
DVN21 0 32 100 10 4 55 57 42
DVN22c 0 34 66 100 103 1 37 7
DVN23 c
0 14 11 96 100 31 38 29
DVN24 0 13 0 0 0 100 54 13
ADM31b 106 7 0 12 0 10 100 100
ADM32b 94 13 1 4 4 4 101 100
293hFcRn-GFP cells were exposed to unlabeled anti-hFcRn mAbs (blocking) followed by a 1,000-fold lower concentration of AF647-labeled anti-hFcRn
a

mAbs in a matrix at pH 7.2. AF647 MFIs of the hFcRn-GFP+-gated cell populations were then determined by flow cytometric analyses. Values represent
% inhibition of labeled mAb binding as described in the Materials and Methods; bADM31, ADM32, DVN1 reciprocal blockade; cDVN22, DVN23 reciprocal
blockade.

fluorescent intensity (MFI) of titered mAb concentrations. Most binding to hFcRn. Taken together, this mAb panel identified at
of the mAbs demonstrated comparably high affinity for hFcRn, least five distinct epitopes on hFcRn.
with the exception of ADM12, which demonstrated a somewhat Confocal IF imaging of hFcRn using the mAbs. We then
lower affinity (Fig. 2B). assessed the ability of the mAbs to stain hFcRn in fresh frozen
To evaluate the epitopes of hFcRn targeted by the mAbs, the tissue sections from hFcRn transgenic mice (on a mFcRn-/- back-
ability of the mAbs to cross-compete for binding to hFcRn on ground) or from patient samples (Fig. 3). ADM31AF647 readily
293hFcRn-GFP cells was determined (Table 1). The binding patterns detected hFcRn in the hFcRn transgenic mouse and human
demonstrated a spectrum of epitopes that were consistent with kidney sections (Fig. 3A and D). Using ADM31AF647, the FcRn
both unique and shared hFcRn binding sites. ADM11, DVN21 signal was strongest in the proximal tubules and the the glom-
and DVN24 appear to have unique epitopes on hFcRn since they erulus (Fig. 3B and E), consistent with the known expression
did not compete with the other mAbs in the competitive binding pattern of FcRn in the kidney.19,20 As a negative control, ADM31
assay. DVN1, ADM31 and ADM32 were able to compete with failed to stain kidney sections from B6 mice, confirming its bind-
each other, which suggests that they bind to overlapping epitopes. ing specificity for hFcRn (Fig. 3C). Other tissue types, includ-
DVN22 and DVN23 were each capable of blocking the other’s ing heart, brain, liver, muscle and spleen, similarly demonstrated
patterns of hFcRn staining consistent with previous reports (data

210 mAbs Volume 4 Issue 2


©2013 Landes Bioscience. Do not distribute.
Downloaded by [121.207.210.105] at 20:58 04 October 2015

Figure 3. Immunofluorescent detection of hFcRn (red channel) in kidney tissue sections from (A) hFcRn transgenic mice, (B) hFcRn transgenic mice
costained for proximal convoluted tubule (blue channel) and podocytes (green channel), (C) wild-type B6 mice costained for proximal convoluted
tubule and podocytes, (D) human tissue, (E) human tissue costained for proximal convoluted tubule and podocytes, and (F) human tissue showing
only proximal convoluted tubule and podocytes (GM, Glomerulus, PCT, proximal convoluted tubules).

Figure 4. hFcRn blocking activity. 293hFcRn-GFP cells were incubated with a range of concentrations of DVN1 (○), ADM11 (●), DVN21 (▲), DVN22 (□),
DVN23 (■), DVN24 (◇), ADM31 (◆) or ADM32 (━) at pH 6, then stained for functional binding with (A) 20 μg/mL hIgGAF647, or (B) 200 μg/mL HSAbiotin
followed by streptavidin-PE. Nonfunctional binding was indicated by the AF647 or PE MFIs of cells stained with labeled ligands at pH 7.2 (X). (C) Scatter
plots of 293hFcRn-GFP cells that were untreated at pH 6.0 (upper left) or at pH 7.2 (upper right), exposed to 200 ng/mL of DVN24 (lower left) or ADM11 (low-
er right) at pH 6.0, and then stained for functional binding of hIgGAF647 at the corresponding pH. (D) Scatter plots of 293hFcRn-GFP cells that were untreated
at pH 6.0 (upper left) or at pH 7.5 (upper right), exposed to 200 ng/mL of ADM31 (lower left) or ADM11 (lower right), and then stained for functional
binding with HSA-biotin and counterstained with streptavidin-PE at the corresponding pH. AF647 or PE MFI values of the hFcRn-GFP+-gated cell popu-
lations are indicated. Data are representative of three independent experiments.

not shown). The other mAbs in the part displayed similar tis- Inhibition of hIgG and HSA binding in vitro. The potential
sue staining profiles for hFcRn, with the exception of DNV24, of each mAb clone to inhibit the pH-dependent binding of hIgG
which cross-reacted with mFcRn (data not shown). These results or human serum albumin (HSA) to hFcRn was analyzed using an
demonstrate the utility of the mAbs for defining the cellular and in vitro binding competition assay. In this assay, 293hFcRn-GFP cells
tissue distribution of human FcRn. were treated with increasing concentrations of each mAb clone

www.landesbioscience.com mAbs 211


before labeled hIgG or HSA was added. Under these
competitive conditions, the bound hIgG or HSA was
quantified by flow cytometry (Fig. 4). Both DVN24
and DVN21 potently blocked hIgG binding to hFcRn,
with the maximum effect at 3 μg/mL and 10 μg/mL,
respectively (Fig. 4A; representative scatter plots results
for DVN24, B). In contrast, DVN23, ADM31 and
ADM32 most efficiently blocked HSA binding, leav-
ing IgG binding intact (Fig. 4C; representative scat-
ter plots results for ADM31, D). These results showed
that certain mAbs block the pH-dependent binding
of hIgG and albumin selectively. Thus, the different

©2013 Landes Bioscience. Do not distribute.


epitopes on hFcRn recognized by our mAb part are
consistent with published results that support spatially
distinct IgG-Fc and HSA binding sites on FcRn.5,15,16,21
Treatment with anti-hFcRn mAbs in vivo. We
next investigated whether the newly generated mAbs
that selectively blocked hIgG binding to hFcRn
Downloaded by [121.207.210.105] at 20:58 04 October 2015

could accelerate the clearance of IgG antibodies


in vivo. For these studies, we focused primarily on
DVN24 because it was the most effective inhibitor
of hIgG binding to hFcRn in vitro (Fig. 4A). Tracer
amounts of hIgG, mIgG1 and mIgA were injected
into hFcRn transgenic mice to establish β-phase Figure 5. Blockade of IgG by anti-hFcRn mAbs in vivo. (A) B6 Tg276 mice were co-
injected ip with 100 μg each of hIgG, mIgG1 (1B7.11) and mIgA (2F11-10) as tracers.
clearance baselines before the mice were treated with The mice were then injected with 1,000 μg (◇), 300 μg (◆), 100 μg (△), 30 μg (▲)
varying doses of DVN24 or an isotype-matched mAb or 0 μg (□) of DVN24 or with 1,000 μg (X) of a control IgG2a mAb into groups of 3
(Fig. 5A and B). Mice treated with DVN24 showed mice on days 2, 3 and 4 d after tracer injection. (B) B6 Tg276 mice (n = 3) were co-
2.6- to 8.2-fold reduced concentrations of hIgG in injected ip with 100 μg hIgG and 1 mg of HSA tracers followed by injections with
serum compared with mice treated with a similar 1 mg doses of ADM32 (-), DVN24 (◇) or isotype matched mIgG2a (X). (C) B6 Tg276
mice (n = 4–5) were injected with 100 μg hIgG tracer and then treated on days 5–7
amount of isotype control mAb, with the maximal or 10–12 with 1 mg of DVN24 (◇) or mIgG2a (X). Significant differences (A–C) are
effect observed at 1 mg dosages. We also determined indicated as *p < 0.05 and **p < 0.01 comparing 1,000 μg injections of DVN24 with
whether the therapeutic administration of DVN24 the control isotype matched mIgG2a. (D) B6 WT mice (n = 8) were injected ip with
affected the serum persistence of HSA. DVN24 failed 200 μg of mIgG1 (1B7.11) as a tracer, and then injected ip on days 4–6 with 1 mg
to promote the clearance of tracer HSA while promot- doses of DVN24 (◇), ADM31 (◆) or vehicle only (X). Tracer plasma concentrations
were determined by ELISA and plotted either as percent remaining as compared
ing the clearance of hIgG in the same mice (Fig. 5B). with the first time point plasma concentrations or as plasma concentrations ±SD.
These results are consistent with the in vitro block- Significant differences (D) are indicated as **p < 0.01 comparing 1,000 μg injections
ade studies described above (Fig. 4), and support the of DVN24 with the vehicle only control.
binding specificity of DVN24 to a site on hFcRn that
specifically interacts with hIgG and results in the accel-
eration of hIgG clearance in vivo without affecting the half-life primarily by the need for reliable serological reagents to probe
of HSA. the biology of hFcRn. Immunization of mice with cells from
Since DVN24 cross-reacts with mFcRn (Fig. 2A), we hFcRn-transgenic mice resulted in a panel of nine independently
addressed whether administration of this mAb could influence derived, stable hybridoma clones that secrete mAbs specific for
the serum persistence of mIgG in mice. Treatment of B6 WT native heavy chain determinants of hFcRn. The applications of
mice with DVN24 resulted in an approximate 2.8-fold reduction the antibody part are summarized in Table 2. We show here that
of tracer mIgG1 compared with treatment with the HSA-blocking these mAbs have promise as incisive reagents capable of unlock-
mAb ADM31, which had a negligible effect (Fig. 5D). While the ing a deeper understanding of the tissue and cellular expression
treatment was not as efficient as that found in the hFcRn studies, patterns of hFcRn, which may facilitate a better understanding of
the results indicate that the mAbs show both species and epitope FcRn biology and lead to development of improved therapeutics.
specificity in this in vivo system. Whether the mAb panel may be applied to the study of FcRn
in other mammalian species, including primates, remains to be
Discussion determined.
The major function of FcRn is to bind IgG and serum albu-
The overall goal of this study was to develop and characterize a min in endosomal compartments and rescue these ligands by
diverse panel of mAbs with specificity for hFcRn and to explore returning them intact to the cell surface. This trafficking is
their applications in vitro and in vivo. This effort was motivated highly dependent on pH because FcRn requires an acidic pH

212 mAbs Volume 4 Issue 2


Table 2. Summary of anti-hFcRn mAb characteristics
mAb Specificity Shared Blockade in vitro
Name Isotype hFcRn mFcRn pH a Bindingb hIgG HSA mIgG FACSc IFc WBc Straind
DVN1 IgG1 + - 7–8 31, 32 - - - + + - WT
ADM11 IgG2b + - 6–8 - - - - + + - WT
ADM12 IgG2b + - 6–8 NDe - - - + + - WT
DVN21 IgG1 + - 6–8 - + - - + + - FcRn-/-
DVN22 IgG1 + - 7–8 23 - - - + + - FcRn-/-
DVN23 IgG2b + - 6–8 22 - - - + + - FcRn-/-
DVN24 IgG2a + + 6–8 - + - + + + - FcRn-/-

©2013 Landes Bioscience. Do not distribute.


ADM31 IgG2b + - 6–8 1, 32 - + - + + - FcRn-/-
ADM32 IgG2b + - 6–8 1, 31 - + - + + - FcRn-/-
a
mAbs binding hFcRn on 293 hFcRn-GFP
cells; Reciprocally compete for binding hFcRn; Detection by cytometric analysis and immunofluorescence (IF), but
b c

fail by western blot (WB); dMouse strains used as responders to generate mAbs; eNot determined.

(~6) for either ligand to bind with high affinity. While bind- FcRn can be used to treat disease. The palliative effects of high
Downloaded by [121.207.210.105] at 20:58 04 October 2015

ing of both IgG and HSA are pH-dependent, their binding to dose administration of IgG in a number of autoimmune disorders
FcRn appear to occur at geographically distinct sites.5,15,16,21 FcRn can be explained at least partially by the ability of this treatment
binding to the Fc region of IgG is mediated by protonation of to saturate FcRn-mediated protection of IgG, resulting in rapid
histidine residues on the Fc fragment that then bind to acidic clearance of pathogenic IgG autoantibodies.27-29 Blockade strate-
residues on the a1 and a2 domains of FcRn.22,23 Less is known gies specifically directed at FcRn include the use of IgG mAbs
about the FcRn-albumin interaction surface, but recent stud- genetically engineered in the Fc region for high affinity binding
ies support a similar histidine-dependent mechanism in which to FcRn (referred to as AbDegs),30-32 and synthetic peptides that
protonated histidine residue (H166) on FcRn engages acidic Fc bind the Fc engagement site of FcRn.33
residues.15,21 As summarized in Table 2, the binding patterns of Blockade using mAbs specifically directed against FcRn is an
the mAb part observed in competition experiments in vitro iden- obvious alternative therapeutic approach to promote the clear-
tified both unique and overlapping epitopes on hFcRn. These ance of potentially pathogenic IgG. Proof of principle for mAb
studies, in combination with the ability of certain anti-hFcRn blockade of FcRn has been demonstrated previously in a rat
mAbs to inhibit the pH-dependent binding of hFcRn to hIgG model of experimentally-induced autoimmune myasthenia gra-
(DVN21 and DVN24) with others inhibiting the binding of vis in which treatment of the FcRn heavy chain-specific mAb
HSA (DVN23, ADM31 and ADM32), suggest that the mAb 1G3 lead to a transient reduction of serum IgG concentrations
epitope footprints are discrete and spatially distinct. Direct bio- as well as the severity of disease.34 However, similar studies were
chemical measurements using methods including surface plas- not conducted for hFcRn. This approach is therapeutically rele-
mon resonance could be used more to exactly define the mAb vant because hFcRn has more stringent binding requirements for
affinities and epitope specificities. The anti-FcRn mAb panel IgG than rodent FcRn. Therefore, a ‘humanized’ FcRn system
can therefore serve as a novel and useful tool for probing interac- is needed to test antibodies and therapies that may be applied to
tions of hFcRn with its ligands and their trafficking patterns. For patients. To this end, we utilized hFcRn-transgenic mice to test
example, two mAbs, DVN1 and DVN22, require a neutral pH the feasibility of hFcRn blockade by epitope-specific antibodies.
to bind hFcRn. At this neutral pH, one (DVN1) cross-competes We first screened each anti-hFcRn mAb that we had generated
with ADM31 and ADM32. The fact that ADM31 and ADM32 for its ability to block IgG binding to hFcRn in vitro. IgG bind-
are able to bind hFcRn and block HSA binding at an acidic pH ing blockade was not a consistent feature in that of the seven anti-
while DVN1 binds this site only at a neutral pH may be of use for hFcRn mAbs tested that bound hFcRn at an acidic (endosomal)
probing pH-sensitive HSA/FcRn interactions along the recycling pH, only DVN21 and DVN24 effectively blocked hIgG binding
pathway using ultramicroscopic/ultrastructural techniques. in vitro (Table 2), and only DVN24 demonstrated therapeutic
Autoantibodies with IgG isotypes contribute to a wide range efficacy by accelerating the clearance of hIgG in vivo (Fig. 5;
of autoimmune disorders, including Goodpasture’s syndome, negative data for DVN21 are not shown). The lack of therapeu-
myasthenia gravis, idiopathic thrombocytopenic purpura (ITP), tic effect with the other anti-hFcRn mAbs, which bind hFcRn
lupus and rheumatoid arthritis. Through its ability to rescue IgG avidly and, therefore, presumably remain bound to hFcRn but
from destruction in the lysosomal compartment, FcRn is respon- do not impede the endosomal trafficking of its normal molecu-
sible for the maintenance of high concentrations of IgG in cir- lar cargo, can be interpreted as support for the durability of the
culation. FcRn-deficient mice are more resistant to autoimmune FcRn recycling pathway.
diseases caused by pathogenic IgG autoantibodies because they DVN24 is the only mAb identified to have a therapeutic affect
are unable to maintain the high concentrations of serum IgG.24-26 on hIgG clearance in the hFcRn Tg model. Since the Fc frag-
These studies lead to the concept that the therapeutic blockade of ment of mIgG minimally binds hFcRn,35 this therapeutic effect

www.landesbioscience.com mAbs 213


is facilitated solely by its antigen specificity for FcRn. However, into which we had already inserted 118 bp of sequence includ-
the effect proved to be transient and required multiple dosages ing the sequence encoding the 23 amino acid hFcRn signal
of >50/mg/kg to achieve an appreciable therapeutic effect. This sequence downstream of the CMV-IE promoter, and upstream
transience is very likely the result of the short serum persistence and in-frame with GFP to produce an N-terminal GFP-tagged
of native DVN24, which virtually disappears from the circu- tailless mFcRn construct. All PCR-amplified inserts were bi-
lation within hours after administration (results not shown). directionally sequence-verified across the cloning sites.
Whether the therapeutic efficiency of DVN24 will be enhanced Cell lines. Purified hFcRn-GFP and mFcRn-GFP plasmids
by the replacement of the mouse Fc with the human IgG-Fc, thus were transfected into HeLa or HEK293 cells using Lipofectamine
enabling it to capitalize on hFcRn’s normal recycling function, Plus (Invitrogen, 15338-100) following the manufacturer’s proto-
remains to be determined. Finally, it is of interest that DVN24 col. Stably transfected HEK293 cells were selected using 400 μg/
was also the only mAb identified that cross-reacts with mFcRn. mL G418 (Sigma-Aldrich, G5013) in 10% FBS-supplemented
Administration of this mAb accelerated the clearance of mIgG1 DMEM. Since the constructs lacked the endosomal targeting

©2013 Landes Bioscience. Do not distribute.


in B6 WT mice, albeit less efficiently than its effect on hIgG domain, the resulting 293hFcRn-GFP and 293mFcRn-GFP cells expressed
clearance in hFcRn Tg mice. DVN24, therefore, has the dual FcRn at high levels on their plasma membrane, as verified by
potential for investigating the effects of the therapeutic blockade immunofluorescence imaging and FACS analysis after staining
of FcRn in humanized and standard mouse models. with anti-FcRn mAbs.
Antisera, hybridoma generation and screening. To generate
Materials and Methods anti-hFcRn antibody responses, B6 WT or FcRn null mice were
Downloaded by [121.207.210.105] at 20:58 04 October 2015

immunized multiple times via intraperitoneal (i.p.) injection


Mice. C57BL6/J (B6) mice. FcRn knockout mice on the with spleen cells from Tg276 mice in and complete or incomplete
C57BL6/J background (designated B6.129X1-Fcgrttm1Dcr /DcrJ) Freund’s adjuvant. Plasma samples collected from boosted mice
carrying a null allele of the α-chain of FcRn have been previously were assayed by flow cytometry for anti-FcRn binding and the
described (FcRn-/-).9,36 Mice carrying hFcRn transgene (desig- functional blockade of IgG binding. To assess hFcRn binding,
nated B6.Cg-Fcgrttm1Dcr Tg(CAG-FCGRT)276Dcr/DcrJ) carry 293hFcRn-GFP cells were incubated with antisera, stained with goat
the null allele of mFcRn and the hFcRn cDNA transgene under anti-mouse IgG-R-phycoeythrin (Southern Biotech, 1031-09),
the control of the CAG promoter and are referred to as Tg276 or and then analyzed using a FACSCalibur (BD Biosciences, San
hFcRn transgenic mice.31,36 All mice were maintained under spe- Jose, CA USA). The ability to block hFcRn function was assessed
cific pathogen-free conditions and the procedures were approved by adding antisera to 293hFcRn-EGFP cells in FACS buffer (PBS with
by the Jackson Laboratory Animal Care and Use Committee. 1% BSA and 0.05% NaN3), pH 6 or pH 7.2, followed by the
Generation of hFcRn and mFcRn constructs for trans- addition of hIgGAF647, and data were similarly acquired using a
fection. To produce the hFcRn construct, the 5' non-coding FACSCalibur.
sequence (118 bp) including the sequence encoding the 23 Spleen cells from immunized mice with high binding or block-
AA hFcRn signal sequence was PCR-amplified from hFcRn ing activity were fused with SP2/0-Ag14 myeloma cells accord-
cDNA (kindly provided by Clark Anderson, Ohio State ing to standard protocols. Selection for hybridomas was provided
University) using forward CCC CCC CCG CTA GCG AAG by refeeding cells on days 2, 3, 4, 5, 7, 9 and 11 with DMEM-20
CCC CTC CTC GGC GTC CTG GT (NheI site underlined) supplemented with HAT and 100 U/mL IL-6. A cellular ELISA
and reverse CCC CCC CCA CCG GTC CGC CCA GGC was used to screen the hybridomas for hFcRn binding activity
TCC CAG GAA GGA GAA A (AgeI site underlined) prim- by incubating culture supernatant with 293hFcRn-GFP cells, stain-
ers. Extra cytosine nucleotides were included at the 5' ends of ing with goat anti-mouse IgG-alkaline phosphatase (Southern
primers to increase the efficiency of restriction endonuclease Biotech, 1031-04), and measured by adding p-nitrophenyl phos-
activity. This PCR product was digested with NheI and AgeI, phate (AMRESCO, 0617) at 1 mg/mL in substrate buffer (50
and inserted downstream of the CMV-IE promoter, between mM sodium bicarbonate, 5 mM MgCl2, pH 8.6) followed by the
the NheI and AgeI restriction sites upstream and in-frame with addition of 1% sodium dodecyl sulfate (Sigma-Aldrich, L4390)
the EGFP coding sequence of the pEGFP-Cl vector (Clontech, to solubilize cells prior to OD405 determination on a SpectraMAX
6084-1) to produce an N-terminal EGFP-tagged hFcRn con- 190 plate reader (Molecular Devices, Sunnyvale, CA). Productive
struct lacking the C-terminal endosomal targeting domain. hybridomas were expanded from anti-hFcRn positive wells, and
The mFcRn construct was generated from RNA isolated from cloned 3 times by limiting dilution to verify stable productive
ten day old C57BL/6J neonatal proximal small intestine and secretion of the resulting subclones. All mAb supernatants were
PCR amplified using forward (CCC CCC CCC TCG AGG protein G (GE Healthcare, 17-0404-01) purified and labeled
GTC AGA GAC CCG CCC CCA, XhoI site underlined) and with Alexa Fluor 674 (Invitrogen, A-20173). Specificity and rela-
reverse (CCC CCC CCG AAT TCG CGC ATC CTG CCC tive affinity were measured by staining 293hFcRn-GFP, 293mFcRn-GFP
CAC AA, EcoRI site underlined) primers. As described above, and 293 cells with AF647-labeled anti-hFcRn mAbs at a fixed
cytosine nucleotides were added to the 5' ends of the primers concentration of 200 ng/106 cells to determine specificity, or
to improve restriction endonuclease cleavage of the PCR prod- with a range of indicated concentrations to determine affinity.
uct. A 944 bp product was digested with XhoI and EcoRI, and Data were acquired using a FACSCalibur and relative affinities
cloned into the corresponding sites of the pEGFP-C1 vector for hFcRn were represented as ratios of the Alexa Fluor 647 MFI

214 mAbs Volume 4 Issue 2


from each mAb concentration to the MFI yielded from its high- For the IF studies shown, hFcRn was detected using Alexa
est concentration. Fluor 647 labeled mAbs. Mouse kidney tissues were counter-
Competitive binding assay. Anti-hFcRn mAbs were screened stained with rat anti-mouse podocalyxin (clone 192703) (R&D
for their ability to compete for binding to hFcRn on ice. In matrix Systems Inc., MAB1556) coupled with a goat anti-rat FITC
fashion, unlabeled mAbs (blocking) were individually added (Invitrogen, 629511) and biotin Lotus Tetragonolobus Lectin
to 106 293hFcRn-EGFP cells at a final concentration of 500 μg/mL (LTL) (Vector Laboratories, B-1325) coupled with streptavidin
for 30 min, and cells were washed with 4 mL FACS buffer, pH DyLite 488 (Thermo Scientific, 21832) to highlight podocytes
7.2. The same set of mAbs labeled with AF647 were then added and proximal convoluted tubules. IF labeling was performed
individually to antibody treated 293hFcRn-GFP cells at a final con- using standard protocols and imaged with a Leica SP5 confo-
centration of 500 ng/mL, incubated for 30 min, and cells were cal microscope within 24 h of mounting. Human tissues were
washed with 4 mL FACS buffer, pH 7.2. Events were acquired stained in a similar fashion.
on a FACSCalibur and the percent inhibition of labeled antibody mAb blockade of FcRn in vivo. hFcRn Tg276 mice carry-

©2013 Landes Bioscience. Do not distribute.


binding was calculated from the Alexa Fluor 647 MFI of each ing one copy of the hFcRn or B6 WT mice were first injected
hFcRn-GFP+ -gated cell population as follows: % Inhibition = i.p. with 100 μg of tracer mIgG1 (1B7-11, anti-TNP, American
(1 - ((MFIEXP - MFISELF)/MFINONE)) x 100, where MFIEXP is the Type Culture Collection, TIB-191), mIgA (2F11-15, anti-TNP,
MFI of a given labeled mAb in the presence of a competing unla- American Type Culture Collection, TIB-194), human IgG
beled blocking mAb, MFISELF is the MFI of a given labeled mAb (Gammagard, Baxter, 1501375) or 1 mg HSA (Sigma-Aldrich,
in the presence of the same unlabeled blocking mAb, MFINONE is A-7284) as tracers. ELISA of serial blood samples was used to
Downloaded by [121.207.210.105] at 20:58 04 October 2015

the MFI of a given labeled mAb in the absence of an unlabeled assess the plasma concentrations of each tracer. The tracer anti-
blocking mAb. bodies were captured with TNP-bovine IgG (EMD Chemicals,
Antibody blockade of hIgG and HSA binding in vitro. 324101), mouse anti-human IgG Fc (Southern Biotech, 9040-
Antisera and mAbs were screened for the ability to block hFcRn 01), or rabbit anti-HSA (United States Biological, A1327-46),
function on ice by adding either 2 μL of heteroantisera, or respectively. Goat anti-mouse IgG1-AP (Southern Biotech, 1070-
defined amounts of purified mAbs, to 106 293hFcRn-GFP cells in 04), rat anti-mouse IgA-AP (Southern Biotech, 1165-04), mouse
25 μL FACS buffer, pH 6 or 7.2 for 30 min, then washed with anti-human kappa-AP (Southern Biotech, 9230-04) or goat
corresponding pH FACS buffer. The cells were incubated for 60 anti-HSA-AP (Bethy Laboratories, A80-229AP), respectively,
min with hIgG-Alexa Fluor 647 (1 μg) or 10 μg human serum were used for detection. The ELISA plates were washed 3 times
albumin-biotin (HSA-biotin), and washed with corresponding with 250 μL ELISA wash (PBS, 0.05% Tween 20, 0.05% NaN3)
pH FACS buffer. For assessment of HSA-biotin binding, cells between each step, and both the block and all dilutions, were
were incubated for 30 min with streptavidin-phycoerythrin made in ELISA wash with 1% BSA. The ELISA was developed
(Southern Biotech, 7100-09) followed by washing with cor- using p-nitrophenyl phosphate substrate added at 1 mg/mL in
responding pH FACS buffer. HSA binding FACS buffer was substrate buffer prior to OD405 measurement on a SpectraMAX
albumin-free. MFI was determined by acquiring events on a 190 plate reader.
FACSCalibur.
Immunofluorescence and confocal microscopy. The expres- Disclosure of Potential Conflicts of Interest
sion pattern of hFcRn in various tissues was determined using D.C.R and S.A. have applied for US and European patents that
immunofluorescence (IF) techniques. Organs from age and include some of the mAbs described.
sex-matched mice were collected and immediately embedded
with OCT and frozen using Precision Cryo-embedding System Acknowledgments
(IHC world, Woodstock, MD). Sectioning of these tissues was This work was supported by the grants from the Alliance for
performed with a Leica CM 1950 Cryostat (Buffalo Grove, IL). Lupus Research and the National Institutes of Health (NIH RO1
Seven to 10 μm sections were applied to Fisherbrand Superfrost® DK56597).
Plus slides and stored at -20°C. Quality control of the prepa-
rations was evaluated by H&E staining of consecutive sections. Authors’ Contributions
Normal human kidney tissue was harvested from nephrectomy G.J.C., V.Z.S., S.A., E.P. and S.P. designed and performed exper-
specimens and snap frozen in OCT medium and sectioned iments and analyzed data. D.C.R. designed experiments and
as described previously. Use of human tissue specimens was analyzed data. G.J.C., V.Z.S., S.A., G.P. and D.C.R. contributed
approved by the Washington University Institutional Review to the writing of the manuscript.
Board (IRB #201107288).

www.landesbioscience.com mAbs 215


13. Ye L, Zeng R, Bai Y, Roopenian DC, Zhu X. 25. Ghetie V, Hubbard JG, Kim JK, Tsen MF, Lee Y, Ward
References
Efficient mucosal vaccination mediated by the neo- ES. Abnormally short serum half-lives of IgG in beta
1. Ghetie V, Ward ES. Multiple roles for the major natal Fc receptor. Nat Biotechnol 2011; 29:158- 2-microglobulin-deficient mice. Eur J Immunol 1996;
histocompatibility complex class I-related recep- 63; PMID:21240266; http://dx.doi.org/10.1038/ 26:690-6; PMID:8605939; http://dx.doi.org/10.1002/
tor FcRn. Annu Rev Immunol 2000; 18:739-66; nbt.1742. eji.1830260327.
PMID:10837074; http://dx.doi.org/10.1146/annurev. 14. Lu L, Palaniyandi S, Zeng R, Bai Y, Liu X, Wang Y, 26. Israel EJ, Wilsker DF, Hayes KC, Schoenfeld D,
immunol.18.1.739. et al. A neonatal Fc receptor-targeted mucosal vac- Simister NE. Increased clearance of IgG in mice that
2. Roopenian DC, Akilesh S. FcRn: the neonatal Fc cine strategy effectively induces HIV-1 antigen-specific lack beta 2-microglobulin: possible protective role of
receptor comes of age. Nat Rev Immunol 2007; 7:715- immunity to genital infection. J Virol 2011; 85:10542- FcRn. Immunology 1996; 89:573-8; PMID:9014824;
25; PMID:17703228; http://dx.doi.org/10.1038/ 53; PMID:21849464; http://dx.doi.org/10.1128/ http://dx.doi.org/10.1046/j.1365-2567.1996.d01-
nri2155. JVI.05441-11. 775.x.
3. Ward ES, Ober RJ. Chapter 4: Multitasking by 15. Andersen JT, Dee Qian J, Sandlie I. The conserved 27. Jin F, Balthasar JP. Mechanisms of intravenous immuno-
exploitation of intracellular transport functions the histidine 166 residue of the human neonatal Fc globulin action in immune thrombocytopenic purpura.
many faces of FcRn. Adv Immunol 2009; 103:77-115; receptor heavy chain is critical for the pH-dependent Hum Immunol 2005; 66:403-10; PMID:15866704;
PMID:19755184; http://dx.doi.org/10.1016/S0065- binding to albumin. Eur J Immunol 2006; 36:3044- http://dx.doi.org/10.1016/j.humimm.2005.01.029.
2776(09)03004-1. 51; PMID:17048273; http://dx.doi.org/10.1002/ 28. Akilesh S, Petkova S, Sproule TJ, Shaffer DJ,
4. Kuo TT, Baker K, Yoshida M, Qiao SW, Aveson VG, eji.200636556.

©2013 Landes Bioscience. Do not distribute.


Christianson GJ, Roopenian D. The MHC class
Lencer WI, et al. Neonatal Fc receptor: from immu- 16. Anderson CL, Chaudhury C, Kim J, Bronson CL, I-like Fc receptor promotes humorally mediated auto-
nity to therapeutics. J Clin Immunol 2010; 30:777-89; Wani MA, Mohanty S. Perspective—FcRn transports immune disease. J Clin Invest 2004; 113:1328-33;
PMID:20886282; http://dx.doi.org/10.1007/s10875- albumin: relevance to immunology and medicine. PMID:15124024.
010-9468-4. Trends Immunol 2006; 27:343-8; PMID:16731041; 29. Li N, Zhao M, Hilario-Vargas J, Prisayanh P, Warren S,
5. Andersen JT, Sandlie I. The versatile MHC class http://dx.doi.org/10.1016/j.it.2006.05.004. Diaz LA, et al. Complete FcRn dependence for intrave-
I-related FcRn protects IgG and albumin from deg- 17. Neumann E, Frei E, Funk D, Becker MD, Schrenk nous Ig therapy in autoimmune skin blistering diseases.
radation: implications for development of new diag- HH, Müller-Ladner U, et al. Native albumin for J Clin Invest 2005; 115:3440-50; PMID:16284651;
nostics and therapeutics. Drug Metab Pharmacokinet targeted drug delivery. Expert Opin Drug Deliv 2010; http://dx.doi.org/10.1172/JCI24394.
2009; 24:318-32; PMID:19745559; http://dx.doi.
Downloaded by [121.207.210.105] at 20:58 04 October 2015

7:915-25; PMID:20586704; http://dx.doi.org/10.151 30. Vaccaro C, Zhou J, Ober RJ, Ward ES. Engineering
org/10.2133/dmpk.24.318. 7/17425247.2010.498474. the Fc region of immunoglobulin G to modulate in
6. Tesar DB, Björkman PJ. An intracellular traffic 18. Andersen JT, Pehrson R, Tolmachev V, Daba MB, vivo antibody levels. Nat Biotechnol 2005; 23:1283-8;
jam: Fc receptor-mediated transport of immuno- Abrahmsén L, Ekblad C. Extending half-life by indi- PMID:16186811; http://dx.doi.org/10.1038/nbt1143.
globulin G. Curr Opin Struct Biol 2010; 20:226- rect targeting of the neonatal Fc receptor (FcRn) using 31. Petkova SB, Akilesh S, Sproule TJ, Christianson GJ,
33; PMID:20171874; http://dx.doi.org/10.1016/j. a minimal albumin binding domain. J Biol Chem Al Khabbaz H, Brown AC, et al. Enhanced half-life
sbi.2010.01.010. 2011; 286:5234-41; PMID:21138843; http://dx.doi. of genetically engineered human IgG1 antibodies in
7. He W, Ladinsky MS, Huey-Tubman KE, Jensen org/10.1074/jbc.M110.164848. a humanized FcRn mouse model: potential applica-
GJ, McIntosh JR, Björkman PJ. FcRn-mediated 19. Akilesh S, Huber TB, Wu H, Wang G, Hartleben B, tion in humorally mediated autoimmune disease. Int
antibody transport across epithelial cells revealed Kopp JB, et al. Podocytes use FcRn to clear IgG from Immunol 2006; 18:1759-69; PMID:17077181; http://
by electron tomography. Nature 2008; 455:542- the glomerular basement membrane. Proc Natl Acad dx.doi.org/10.1093/intimm/dxl110.
6; PMID:18818657; http://dx.doi.org/10.1038/ Sci USA 2008; 105:967-72; PMID:18198272; http:// 32. Patel DA, Puig-Canto A, Challa DK, Perez Montoyo H,
nature07255. dx.doi.org/10.1073/pnas.0711515105. Ober RJ, Ward ES. Neonatal Fc receptor blockade by
8. Gan Z, Ram S, Vaccaro C, Ober RJ, Ward ES. 20. Haymann JP, Levraud JP, Bouet S, Kappes V, Hagège Fc engineering ameliorates arthritis in a murine model.
Analyses of the recycling receptor, FcRn, in live cells J, Nguyen G, et al. Characterization and localization of J Immunol 2011; 187:1015-22; PMID:21690327;
reveal novel pathways for lysosomal delivery. Traffic the neonatal Fc receptor in adult human kidney. J Am http://dx.doi.org/10.4049/jimmunol.1003780.
2009; 10:600-14; PMID:19192244; http://dx.doi. Soc Nephrol 2000; 11:632-9; PMID:10752522. 33. Mezo AR, Sridhar V, Badger J, Sakorafas P, Nienaber
org/10.1111/j.1600-0854.2009.00887.x. 21. Andersen JT, Dalhus B, Cameron J, Daba MB, V. X-ray crystal structures of monomeric and dimeric
9. Roopenian DC, Christianson GJ, Sproule TJ, Brown Plumridge A, Evans L, et al. Structure-based mutagen- peptide inhibitors in complex with the human neonatal
AC, Akilesh S, Jung N, et al. The MHC class I-like IgG esis reveals the albumin-binding site of the neonatal Fc Fc receptor, FcRn. J Biol Chem 2010; 285:27694-
receptor controls perinatal IgG transport, IgG homeo- receptor. Nat Commun 2012; 3:610; PMID:22215085; 701; PMID:20592032; http://dx.doi.org/10.1074/jbc.
stasis and fate of IgG-Fc-coupled drugs. J Immunol http://dx.doi.org/10.1038/ncomms1607. M110.120667.
2003; 170:3528-33; PMID:12646614. 22. Burmeister WP, Huber AH, Bjorkman PJ. Crystal 34. Liu L, Garcia AM, Santoro H, Zhang Y, McDonnell K,
10. Baker K, Qiao SW, Kuo T, Kobayashi K, Yoshida M, structure of the complex of rat neonatal Fc receptor Dumont J, et al. Amelioration of experimental autoim-
Lencer WI, et al. Immune and non-immune functions with Fc. Nature 1994; 372:379-83; PMID:7969498; mune myasthenia gravis in rats by neonatal FcR block-
of the (not so) neonatal Fc receptor, FcRn. Semin http://dx.doi.org/10.1038/372379a0. ade. J Immunol 2007; 178:5390-8; PMID:17404325.
Immunopathol 2009; 31:223-36; PMID:19495758; 23. Martin WL, West AP Jr, Gan L, Bjorkman PJ. 35. Ober RJ, Radu CG, Ghetie V, Ward ES. Differences
http://dx.doi.org/10.1007/s00281-009-0160-9. Crystal structure at 2.8 A of an FcRn/heterodimeric Fc in promiscuity for antibody-FcRn interactions across
11. Baker K, Qiao SW, Kuo TT, Aveson VG, Platzer B, complex: mechanism of pH-dependent binding. Mol species: implications for therapeutic antibodies. Int
Andersen JT, et al. Neonatal Fc receptor for IgG (FcRn) Cell 2001; 7:867-77; PMID:11336709; http://dx.doi. Immunol 2001; 13:1551-9; PMID:11717196; http://
regulates cross-presentation of IgG immune complexes org/10.1016/S1097-2765(01)00230-1. dx.doi.org/10.1093/intimm/13.12.1551.
by CD8-CD11b+ dendritic cells. Proc Natl Acad Sci 24. Christianson GJ, Blankenburg RL, Duffy TM, Panka 36. Roopenian DC, Christianson GJ, Sproule TJ. Human
USA 2011; 108:9927-32; PMID:21628593; http:// D, Roths JB, Marshak-Rothstein A, et al. beta2- FcRn transgenic mice for pharmacokinetic evalua-
dx.doi.org/10.1073/pnas.1019037108. microglobulin dependence of the lupus-like autoim- tion of therapeutic antibodies. Methods Mol Biol
12. Liu X, Lu L, Yang Z, Palaniyandi S, Zeng R, Gao mune syndrome of MRL-lpr mice. J Immunol 1996; 2010; 602:93-104; PMID:20012394; http://dx.doi.
LY, et al. The neonatal FcR-mediated presentation 156:4932-9; PMID:8648144. org/10.1007/978-1-60761-058-8_6.
of immune-complexed antigen is associated with
endosomal and phagosomal pH and antigen stabil-
ity in macrophages and dendritic cells. J Immunol
2011; 186:4674-86; PMID:21402891; http://dx.doi.
org/10.4049/jimmunol.1003584.

216 mAbs Volume 4 Issue 2

You might also like