Quantitative Assays To Accelerate Therapeutic Antibody Development
Whitepaper
Published: April 30, 2024
Credit: iStock
Therapeutic antibodies represent a powerful class of drugs used for the treatment of cancer, immune disease or viral infection.
In the development of new therapeutic antibodies, scientists must choose cell models and assays carefully to optimize functionality, ensuring that an appropriate immune response is activated through their interaction with Fc receptors expressed on the surface of immune cells.
This whitepaper explores the tools available to characterize antibody binding to Fc receptors, including quantitative assays to support optimization of therapeutic products.
Download this whitepaper to learn more about:
- Immune signaling pathways and targets for therapeutic antibodies
- How to assay the effectiveness of antibodies against a range of Fc receptors
- Kits to support the optimization of therapeutic antibodies and Fc receptor research
Quantitative Assays to Measure Therapeutic
Antibody Binding to Fc Receptors
Fc Receptors, expressed on the surface of immune
cells, bind the Fc (fragment crystallizable) portion of
antibodies and play an essential role in modulating our
immune defense system. They also impact the efficacy of
therapeutic antibodies and therefore are of great interest
in drug development [1]. Indeed, the effectiveness of a
therapeutic antibody depends not only on how tightly it
binds to the intended target, but also on how long it is
present in the patient’s blood and how well it engages an
appropriate immune response through its interaction with
Fc receptors.
Therapeutic antibodies represent a powerful class of
drugs used for the treatment of cancer, immune disease,
or viral infection. They include neutralizing antibodies
targeting cytokines or cytokine receptors, as well as
cytotoxic antibodies and antibody-drug conjugates that
bind to cancer-specific targets and directly kill the tumor
cells. Antibodies against immune targets such as anti-PD-1/
PD-L1 or anti-CTLA4 antibodies are designed to enhance
immune responses within the tumor microenvironment.
The half-life of an antibody is controlled by binding to
the neonatal Fc receptor for IgG (FcRn), which regulates
distinct functions in IgG transport and homeostasis. Other
Fc receptors trigger immune responses through antibodydependent cell mediated cytotoxicity (ADCC), a mechanism
in which a natural killer (NK) cell is activated by antibodies
bound to tumor cells, followed by the lysis of the tumor
cell.
These Fc receptors are classified based on the type of
immunoglobulin (Ig) that they recognize: Fcγ receptors
bind to IgG, Fcα receptors bind to IgA, and Fcε receptors
bind to IgE.
The development of new therapeutic antibodies
(which are mostly IgGs) may require optimization of
their interaction with FcRn or with the appropriate FcγR.
Scientists must choose cell models and assays carefully
depending on their intended goal.
The Fcγ receptors function as antibody checkpoints.
They contain multiple extracellular immunoglobulin
domains responsible for binding the Fc region of IgG. The
FcγR family includes FcγRI (CD64), FcγRIIa (CD32a), FcγRIIb
(CD32b), FcγRIIc (CD32c), FcγRIIIa (CD16a), and FcγRIIIb
(CD16b), which differ in IgG affinity due to divergences
in molecular structure [2, 3]. Five of the receptors are
activating and only FcγRIIb is inhibitory (Figure 1).
Activating Fcγ receptors contain two ITAMs
(immunoreceptor tyrosine-based activation motif) in their
cytoplasmic domain, consisting of amino acid sequence
Antibody Checkpoints
Figure 1. The antibody checkpoint family, expressed on the surface of
innate immune cells and B cells, comprises activating and inhibitory
Fcγ receptors. Inspired from [4].
Created with BioRender.com
Introduction to Fc Receptors
1 bpsbioscience.com
YxxL/Ix(6-8)YxxL/I. FcγRI and FcγRIIIa do not contain
an ITAM but signal through another ITAM-containing
membrane-anchored subunit.
Upon activation of the receptor by IgG binding, the
ITAM is phosphorylated on both tyrosine residues by an
intracellular tyrosine kinase of the Src family. It becomes a
docking site for SH2 domain-containing signaling proteins,
initiating the signal transduction cascade necessary to
generate a biological response (Figure 2).
Inhibitory FcγRIIb, expressed mostly in B cells, provides
a negative feedback loop that controls B cell stimulation.
The receptor carries an intracellular ITIM sequence
(immunoreceptor tyrosine-based inhibitory motif), which is
a single I/VXXYXXL motif. Once phosphorylated, it engages
SH2-containing tyrosine phosphatases such as SHP1, SHP2,
or SHIP, which antagonize the phosphotyrosine signals [5, 6].
FcγRs participate in various biological functions
depending on IgG specificity, cellular expression, and
signaling. Receptors present on NK cells bind to antibodies
that are attached to infected cells or invading pathogens to
promote their lysis. FcγRs on phagocytes bind antibodies
attached to invading bacteria to trigger phagocytosis of the
bacterium, while FcγRs on eosinophils cause degranulation.
Therapeutic antibodies take advantage of antibodydependent cell-mediated cytotoxicity (ADCC) activated
by FcγRIIIa. Cell-based studies performed during the
optimization of a therapeutic antibody will be facilitated
by the engineering of target-expressing cells, the design
of cell-based assays using reporter target or effector cells,
or the design of co-culture assays. These research tools are
especially useful in the field of immuno-therapeutics.
The typical ADCC process involves the activation of NK
cells, which express mostly FcγRIIIa (CD16a), and the release
of cytotoxins that attack the target cells. Human FcγRIIIa
exhibits a dimorphism at residue 158, in which variant
Val-158 encodes a higher affinity receptor than variant
Phe-158. FcγRIIIa potentiates the efficacy of therapeutic
antibodies used to treat solid tumors and represents a
direct therapeutic target in hematopoietic cancers.
Data shown in Figure 3 illustrate an effective ADCC
bioassay using Jurkat cells that overexpress FcγRIIIa-F158.
In the example shown here, a conditional luciferase
reporter gene under the control of NFAT (Nuclear Factor
of Activated T cells) response elements was introduced
in the Jurkat cells to allow quantification of NFAT
stimulation. The target cells were incubated with an
antibody of interest. Upon addition of the Jurkat cells and
binding of FcγRIIIa to the antibody, the NFAT signaling
pathway was activated, resulting in a dose-dependent
Figure 2: Activating and inhibitory signaling of FcγRIIa (blue)
and FcγRIIb (red), respectively.
Created with BioRender.com
Activator of ADCC, FcγRIIIa
2
increase in luciferase activity. Thus, the ADCC efficacy of
different antibodies can be compared directly.
FcγRIIb (CD32b) operates as a negative regulator of B
Cell Receptor (BCR)-induced activation of B cells [7]. The
two isoforms FcγRIIb1 and FcγRIIb2, arising from mRNA
splicing, differ in expression and function. The presence of
exon C1 sequence in FcγRIIb1, which is highly expressed at
the surface of B cells, tethers the receptor at the membrane
and dramatically increases its half-life at the cell surface.
The absence of exon C1 in FcγRIIb 2, expressed in myeloid
cells, triggers rapid internalization of the receptor upon
ligand binding. FcγRIIb induces the phagocytosis of
aggregated immunoglobulins and may function as a
“sink” for the removal of IgG immune complexes. Thus,
the biological function of FcγRIIb is to tame antibodydependent responses and to clear the circulation of spent
immune complexes. Defects in FcγRIIb1 signaling lead
to overt inflammation and are involved in autoimmune
diseases.
Antibody checkpoint inhibitor FcγRIIb
Figure 3. Top left: illustration of the ADCC bioassay principle. Top right: flow cytometry analysis of FcγRIIIa-F158 overexpression in the
ADCC Bioassay Effector Jurkat Cell Line (F variant; BPS Bioscience #60540). Bottom left: ADCC response to anti-HER2 antibody drug
Trastuzumab in the presence of HER2-expressing SKBR-3 breast cancer cells. Bottom right: ADCC response to increasing doses of
Trastuzumab, in a co-culture of SKBR-3 and FcγRIIIa-F158/NFAT luciferase reporter Jurkat cells (EC50 = 28.1 ng/ml).
Illustration created with Biorender.com.
3 bpsbioscience.com
FcγRIIb is an important therapeutic target for the
treatment of B-cell malignancies. Therefore, FcγRIIbexpressing cells can be useful to identify and characterize
anti-FcγRIIb antibodies, bi-specific T cell engagers,
antibody-drug conjugates, or anti-FcγRIIb CAR (Chimeric
Antigen Receptor) cells.
On the other hand, FcγRIIb contributes to the
effectiveness of immunotherapy by cross-linking antibodies
directed at T cell stimulatory checkpoints such as 4-1BB,
OX40, and CD40. Co-culture assays have been designed to
characterize the agonist activity of checkpoint antibodies
using FcγRIIb-mediated crosslinking.
As shown in Figure 4, FcγRIIb CHO (Chinese Hamster
Ovary) cells placed in co-culture with CD137/NF-κB
reporter cells validated the activating efficacy of an antiCD137 antibody.
In another experiment, a TCR activator (TCRa)
was expressed together with FcγRIIb in CHO cells
(BPS Bioscience #78436). This cell line can be
used in a co-culture assay to screen for regulators
of antibody-mediated signaling and to identify
or characterize agonists of FcγRIIb receptor-mediated
crosslinking of checkpoint targets. As shown in Figure
5, FcγRIIb amplified the effect of an anti-PD-1 antibody,
as indicated by PD-1-mediated inhibition of TCR activity
observed in the presence of TCRa/FcγRIIb CHO cells, which
was not observed in the presence of control TRCa CHO cells.
Figure 5. Left: illustration of the co-culture assay. Right: a co-culture assay was performed using the PD-1/NFAT Reporter Jurkat Cell
Line (BPS Bioscience #60535) with either the TCRa/FcγRIIb CHO Cell Line (BPS Bioscience #78436) or the TCRa CHO Cell line (BPS
Bioscience #60539), in the presence of increasing concentrations of anti-PD-1 antibody (BPS Bioscience #101178).
Illustration created with Biorender.com
Figure 4. Dose response of anti-CD137 antibody in CD137/NFκB-reporter HEK293 cells (BPS Bioscience #79289) co-cultured
with FcγRIIb CHO cells (BPS Bioscience #79511). Cross-linking
of the anti-CD137 antibody by FcγRIIb expressed at the surface
of CHO cells potentiated the activation of NF-κB in CD137-
expressing HEK293 cells (in red). Control CHO cells are shown
in green.
4
Neonatal Fc receptor for IgG (FcRn) is a heterodimeric
protein similar in structure to MHC class I [8]. It consists of
the Fc Gamma Receptor and Transporter, encoded by the
FCGRT gene, associated with beta-2-microglobulin. FcRn
binds to the Fc region of monomeric IgG and transports
the IgG from mother to fetus through the placenta. This
receptor contributes to an effective humoral immunity
by protecting the IgGs from degradation in the lysosome
and recycling them, thereby extending their half-life in
circulation. This can be exploited through the engineering
of therapeutic antibodies to increase their binding to FcRn,
thereby improving their half-life. Evusheld, a cocktail of
mutated antibodies with extended half-lives, has been
used to treat COVID-19, whereas first-in-class drug Enbrel
contains an Fc domain fused to therapeutic protein TNFα
to increase the drug’s half-life.
Conversely, FcRn itself is a candidate target for
autoimmune disease therapy since disrupting the FcRn/
IgG interaction is expected to increase IgG clearance,
including autoantibodies. The first FDA-approved drug
targeting FcRn (efgartigimod), an Fc fragment decoy,
provided proof-of-concept and is now used to treat the
autoimmune disease myasthenia gravis.
Interactions of antibody drugs with Fc receptors are
important factors in therapeutics. We have developed a
suite of proteins, assay kits, lentiviruses, and engineered cell
lines that enable quantitative Fc receptor activation, ADCC
measurement, FcRn binding, and more. BPS Bioscience will
continue to accelerate research by developing innovative
tools to support optimization of therapeutic antibodies
and Fc receptor research.
Conclusion
FcRn
Figure 6. Left: direct binding of a decoy corresponding to an Fc region engineered for high affinity binding to FcRn (EC50=1.35 nM).
The decoy was tested using purified, biotinylated human FcRn (BPS Bioscience #71283). Right: inhibition of FcRn binding to IgG1 by
FcRn Blocker (BPS Bioscience #101468) using the Fc:FcRn Inhibitor Screening Colorimetric Assay Kit (BPS Bioscience #78501).
5 bpsbioscience.com
Product Product type Cat number
ADCC Bioassay Effector Cell F variant Jurkat Cell Line Cell Line 60540
ADCC Bioassay Effector Cell V variant Jurkat Cell Line Cell Line 60541
ADCC Bioassay Effector Cell (Mouse) Jurkat Cell Line Cell Line 79733
ADCP Bioassay Effector Cell FcγRIIa (H variant) NFAT Reporter Jurkat Cell Line 71273
FcγRIIb CHO Recombinant Cell Line Cell Line 79511
FcγRIIIa (CD16a) CHO Cell Line Cell Line 78332
FcγRIIIb (CD16b) CHO Cell Line Cell Line 78333
FcRL5 HEK293 Cell Line Cell Line 78374
FcRL5 CHO Cell Line Cell Line 78375
TCR Activator/FcγRIIb CHO Cell Line Cell Line 78436
Fc (IgG1):FcRn Inhibitor Screening Colorimetric Assay Kit Assay Kit 78501
FcγRIIa (Human) CRISPR/Cas9 Lentivirus (Integrating) Lentivirus 78207
FcγRIIa (Human) CRISPR/Cas9 Lentivirus (Non-Integrating) Lentivirus 78199
FcγRIIIa (CD16a) Lentivirus Lentivirus 79876
FcγRIIb (CD32b) Lentivirus Lentivirus 79877
1. Bournazos S, Ravetch JV. Fcγ Receptor Function and
the Design of Vaccination Strategies. Immunity, 47(2):
224-233 (2017); PMID: 28813656.
2. Indik ZK, Park JG, Hunter S, Schreiber AD. The
molecular dissection of Fc gamma receptor mediated
phagocytosis. Blood, 86(12): 4389-4399 (1995); PMID:
8541526.
3. Nimmerjahn F, Ravetch JV. Divergent immunoglobulin
g subclass activity through selective Fc receptor
binding. Science, 310(5753): 1510-1512 (2005); PMID:
16322460.
4. Teige I, Mårtensson L, Frendéus BL. Targeting
the Antibody Checkpoints to Enhance Cancer
Immunotherapy - Focus on FcγRIIB. Front Immunol. 10:
481 (2019); PMID: 30930905.
5. Scharenberg AM, Kinet JP. The emerging field of
receptor-mediated inhibitory signaling: SHP or SHIP?
Cell, 87(6): 961-964 (1996); PMID: 8978600.
6. Cambier JC. Inhibitory receptors abound? Proc
Natl Acad Sci USA. 94(12): 5993-5995 (1997); PMID:
9177155.
7. Anania JC, Chenoweth AM, Wines BD, Hogarth PM.
The Human FcγRII (CD32) Family of Leukocyte FcR
in Health and Disease. Front Immunol. 10: 464 (2019);
PMID: 30941127.
8. Zhu X, Meng G, Dickinson BL, Li X, Mizoguchi E, Miao
L, Wang Y, Robert C, Wu B, Smith PD, Lencer WI,
Blumberg RS. MHC class I-related neonatal Fc receptor
for IgG is functionally expressed in monocytes,
intestinal macrophages, and dendritic cells. J Immunol.
166(5): 3266-3276 (2001); PMID: 11207281.
References
6
Brought to you by
Access This Whitepaper for FREE Now!
Information you provide will be shared with the sponsors for this content. Technology Networks or its sponsors may contact you to offer you content or products based on your interest in this topic. You may opt-out at any time.