Sie sind auf Seite 1von 8

http://www.invivogen.

com/review-engineered-pfuse-chig
Engineered Fc Regions Review- 2011
Engineering Fc regions for altered properties

The Fc region of an antibody mediates its serum half-life and
effector functions, such as complement-dependent cytotoxicity
(CDC), antibody-dependent cellular cytotoxicity (ADCC) and
antibody-dependent cell phagocytosis (ADCP).
Engineering the Fc region of a therapeutic monoclonal antibody or
Fc fusion protein allows the generation of molecules that are better
suited to the pharmacology activity required of them [1].
Engineered Fc regions for increased half-life
One approach to improve the efficacy of a therapeutic antibody is
to increase its serum persistence, thereby allowing higher
circulating levels, less frequent administration and reduced doses.

The half-life of an IgG depends on its pH-dependent binding to the
neonatal receptor FcRn (=Fc receptor=Brambell receptor)*.
* In humans, it is found in the placenta to help facilitate transport of mother's IgG to the
growing fetus and it has also been shown to play a role in monitoring IgG turnover. FcRn
binds IgG at acidic pH of (<6.5) but not at neutral or higher pH. Therefore, FcRn can bind IgG
from the intestinal lumen (the inside of the gut) at a slightly acidic pH and ensure efficient
unidirectional transport to the basolateral side (inside the body) where the pH is neutral to
basic (pH 7.07.5).
This receptor also plays a role in adult salvage of IgG through its occurrence in the pathway
of endocytosis in endothelial cells . Fc receptors in the acidic endosomes bind to IgG
internalized through pinocytosis, recycling it to the cell surface, releasing it at the basic pH of
blood, thereby preventing it from undergoing lysosomal degradation. This mechanism may
provide an explanation for the greater half -life of IgG in the blood compared to other
isotypes.









FcRn, which is expressed on the surface of endothelial cells, binds
the IgG in a pH-dependent manner and protects it from
degradation.

Some antibodies that selectively bind the FcRn at pH 6.0, but not
pH 7.4, exhibit a higher half-life in a variety of animal models.

Several mutations located at the interface between the CH2 and
CH3 domains, such as T250Q/M428L [2] and
M252Y/S254T/T256E + H433K/N434F [3], have been shown to
increase the binding affinity to FcRn and the half-life of IgG1 in
vivo.

However, there is not always a direct relationship between
increased FcRn binding and improved half-life [4].

It has been shown that conjugation of some drugs to the
Fc domain of IgG significantly increases their half-life
There are several drugs on the market that have the Fc
portion fused to the effector protein in order to increase its
halflife.
They are: Amevive (alefacept), Arcalyst (rilonacept),
Enbrel (etanercept), Nplate (romiplostim), Orencia
(abatacept) and Nulojix (belatacept).





Amevive
(alefacept)
A genetically engineered
immunosuppressive drug.
Alefacept is a fusion protein: it
combines part of an antibody
with a protein that blocks the
growth of some types of T
cells.
Used to control
inflammation in
moderate to severe
psoriasis with
plaque formation,
where it interferes
with lymphocyte
activation.
[1]
It is
also being studied
in the treatment of
cutaneous T-cell
lymphoma and T-
cell non-Hodgkin
lymphoma.

Arcalyst
(rilonacept
)
An interleukin 1 inhibitor.
Rilonacept is a dimeric fusion
protein consisting of the
ligand-binding domains of the
extracellular portions of the
human interleukin-1 receptor
component (IL-1R1) and IL-1
receptor accessory protein (IL-
1RAcP) linked in-line to the
fragment-crystallizable portion
(Fc region) of human IgG1 that
binds and neutralizes IL-1
"Orphan Drug" is
used for the
treatment of
cryopyrin-
associated
periodic
syndromes (CAPS),
including familial
cold
autoinflammatory
syndrome,
Muckle-Wells
syndrome and
neonatal onset
multisystem
inflammatory
disease
Enbrel
(etanercep
t)
Treats autoimmune diseases
by interfering with tumor
necrosis factor (TNF; a soluble
rheumatoid,
juvenile
rheumatoid and
inflammatory cytokine) by
acting as a TNF inhibitor.
A fusion protein produced by
recombinant DNA.
It fuses the TNF receptor to
the constant end of the IgG1
antibody.
First, the developers isolated
the DNA sequence that codes
the human gene for soluble
TNF receptor 2, which is a
receptor that binds to tumor
necrosis factor-alpha.
Second, they isolated the DNA
sequence that codes the
human gene for the Fc end of
immunoglobulin G1 (IgG1).
Third, they linked the DNA for
TNF receptor 2 to the DNA for
IgG1 Fc.
Finally, they expressed the
linked DNA to produce a
protein that links the protein
for TNF receptor 2 to the
protein for IgG1 Fc.
psoriatic arthritis,
plaque psoriasis
and ankylosing
spondylitis.


Engineered Fc regions for altered effector
function
Depending on the therapeutic antibody or Fc fusion
protein application, it may be desired to either reduce or increase
the effector function. For antibodies that target cell-surface
molecules, especially those on immune cells, abrogating effector
functions is required. Conversely, for antibodies intended for
oncology use, increasing effector functions may improve the
therapeutic activity.
The four human IgG isotypes bind the activating Fc receptors
(FcRI, FcRIIa, FcRIIIa), the inhibitory FcRIIb receptor, and the
first component of complement (C1q) with different affinities,
yielding very different effector functions [5].

Binding of IgG to the FcRs or C1q depends on residues
located in the hinge region and the CH2 domain. Two regions of
the CH2 domain are critical for FcRs and C1q binding, and have
unique sequences in IgG2 and IgG4. Substitutions into human
IgG1 of IgG2 residues at positions 233-236 and IgG4 residues at
positions 327, 330 and 331 were shown to greatly reduce ADCC
and CDC [6, 7]. Furthermore, Idusogie et al. demonstrated that
alanine substitution at different positions, including K322,
significantly reduced complement activation [8]. Similarly,
mutations in the CH2 domain of murine IgG2A were shown to
reduce the binding to FcRI, and C1q [9].
Numerous mutations have been made in the CH2 domain of
human IgG1 and their effect on ADCC and CDC tested in vitro [7-
10]. Notably, alanine substitution at position 333 was reported to
increase both ADCC and CDC [7, 9.] Lazar et al. described a triple
mutant (S239D/I332E/A330L) with a higher affinity for FcRIIIa and
a lower affinity for FcRIIb resulting in enhanced ADCC [11]. The
same mutations were used to generate an antibody with increased
ADCC [12]. Richards et al. studied a slightly different triple mutant
(S239D/I332E/G236A) with improved FcRIIIa affinity and
FcRIIa/FcRIIb ratio that mediates enhanced phagocytosis of
target cells by macrophages [13].
Due to their lack of effector functions, IgG4 antibodies represent
the preferred IgG subclass for receptor blocking without cell
depletion. IgG4 molecules can exchange half-molecules in a
dynamic process termed Fab-arm exchange. This phenomenon
can occur between therapeutic antibodies and endogenous IgG4.

The S228P mutation has been shown to prevent this
recombination process allowing the design of less unpredictable
therapeutic IgG4 antibodies [14].

Features of Engineered Fc
Engineered
Fc
Isotype Species Mutations
FcR/C1q
Binding
Effector
Function
Refs
hIgG1e1-Fc IgG1 Human T250Q/M428L
Increased
binding to FcRn
Increased
half-life
2
hIgG1e2-Fc IgG1 Human
1M252Y/S254T/T256E +
H433K/N434F
Increased
binding to FcRn
Increased
half-life
3
hIgG1e3-Fc IgG1 Human
E233P/L234V/L235A/?G236
+ A327G/A330S/P331S
Reduced
binding to
FcRI
Reduced
ADCC and
CDC
6,7
hIgG1e4-Fc IgG1 Human E333A
Increased
binding to
FcRIIIa
Increased
ADCC and
CDC
8,10
hIgG1e5-Fc IgG1 Human S239D/A330L/I332E
Increased
binding to
FcRIIIa
Increased
ADCC
11,12
hIgG1e6-Fc IgG1 Human P257I/Q311
Increased
binding to FcRn
Unchanged
half-life
4
hIgG1e7-Fc IgG1 Human K326W/E333S
Increased
binding to C1q
Increased
CDC
9
hIgG1e9-Fc IgG1 Human S239D/I332E/G236A
Increased
FcRIIa/FcRIIb
ratio
Increased
macrophage
phagocytosis
13
hIgG2e1-Fc IgG1 Human K322A
Reduced
binding to C1q
Reduced
CDC
8
hIgG4e1-Fc IgG4 Human S228P -
Reduced
Fab-arm
exchange
14
mIgG2Ae1-
Fc
IgG2a Mouse
L235E +
E318A/K320A/K322A
Reduced
binding to
FcRI and C1q
Reduced
ADCC and
CDC
9

1. Strohl WR., 2009. Optimization of Fc-mediated effector functions of monoclonal antibodies. Curr Opin
Biotechnol. 20(6):685-91. Review.
2. Hinton PR. et al., 2004. Engineered human IgG antibodies with longer serum half-lives in primates. J
Biol Chem. 279(8):6213-6.
3. Vaccaro C. et al., 2005. Engineering the Fc region of immunoglobulin G to modulate in vivo antibody
levels. Nat Biotechnol. 23(10):1283-8.
4. Datta-Mannan A. et al., 2007. Humanized IgG1 Variants with Differential Binding Properties to the
Neonatal Fc Receptor : Relationship to Pharmacokinetics in Mice and Primates. Drug Metab. Dispos.
35: 86 - 94.
5. Bruhns P. et al., 2009. Specificity and affinity of human Fcgamma receptors and their polymorphic
variants for human IgG subclasses. Blood. 113(16):3716-25.
6. Armour KL. et al., 1999. Recombinant human IgG molecules lacking Fcgamma receptor I binding and
monocyte triggering activities. Eur J Immunol. 29(8):2613-24.
7. Shields RL. et al., 2001. High resolution mapping of the binding site on human IgG1 for Fc gamma
RI, Fc gamma RII, Fc gamma RIII, and FcRn and design of IgG1 variants with improved binding to the
Fc gamma R. J Biol Chem. 276(9):6591-604.
8. Idusogie EE. et al., 2000. Mapping of the C1q binding site on rituxan, a chimeric antibody with a
human IgG1 Fc. J Immunol. 164(8):4178-84.
9. Steurer W. et al., 1995. Ex vivo coating of islet cell allografts with murine CTLA4/Fc promotes graft
tolerance. J Immunol. 155(3):1165- 74.
10. Idusogie EE. et al., 2001. Engineered antibodies with increased activity to recruit complement. J
Immunol. 166(4):2571-5.
11. Lazar GA. et al., 2006. Engineered antibody Fc variants with enhanced effector function. PNAS
103(11): 40054010.
12. Ryan MC. et al., 2007. Antibody targeting of B-cell maturation antigen on malignant plasma cells.
Mol. Cancer Ther., 6: 3009 - 3018.
13. Richards JO,. et al., 2008. Optimization of antibody binding to FcgammaRIIa enhances macrophage
phagocytosis of tumor cells. Mol Cancer Ther. 7(8):2517-27.
14. Labrijn AF. et al., 2009. Therapeutic IgG4 antibodies engage in Fab-arm exchange with endogenous
human IgG4 in vivo. Nat Biotechnol. 27(8):767-71.

Das könnte Ihnen auch gefallen