Download innovative development strategies and applications for bispecific

Survey
yes no Was this document useful for you?
   Thank you for your participation!

* Your assessment is very important for improving the workof artificial intelligence, which forms the content of this project

Document related concepts

Phagocyte wikipedia , lookup

Blood type wikipedia , lookup

Gluten immunochemistry wikipedia , lookup

Complement system wikipedia , lookup

Immunoprecipitation wikipedia , lookup

Innate immune system wikipedia , lookup

Psychoneuroimmunology wikipedia , lookup

Immune system wikipedia , lookup

Sjögren syndrome wikipedia , lookup

Duffy antigen system wikipedia , lookup

DNA vaccination wikipedia , lookup

Adaptive immune system wikipedia , lookup

Molecular mimicry wikipedia , lookup

Immunomics wikipedia , lookup

Adoptive cell transfer wikipedia , lookup

Autoimmune encephalitis wikipedia , lookup

Immunocontraception wikipedia , lookup

ELISA wikipedia , lookup

Anti-nuclear antibody wikipedia , lookup

Antibody wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Cancer immunotherapy wikipedia , lookup

Immunosuppressive drug wikipedia , lookup

Monoclonal antibody wikipedia , lookup

Transcript
INNOVATIVE DEVELOPMENT
STRATEGIES AND
APPLICATIONS FOR
BISPECIFIC ANTIBODIES
Advanced protein design and engineering techniques
have enabled the production of bispecific antibodies,
unique antibody constructs that have the ability
to bind two targets. This rapidly growing class of
therapeutics is being investigated globally in clinical
trials with several on the market for a wide variety of
cancer types and a diverse spectrum of diseases and
conditions such as rheumatoid arthritis, hemophilia,
postmenopausal bone loss, plaque psoriasis, and
idiopathic pulmonary fibrosis. Specially engineered
antibodies with a variety of formats simultaneously
bind to two different epitopes on the same antigen
or different antigens, increasing selectivity and
effectiveness. [1]
Despite the widespread use of monoclonal antibodies
for passive cancer immunotherapy, patients often
do not respond completely, exhibiting resistance to
therapy or tumor recurrence. This is a result of the
complexity involved in proliferation and survival of
tumor cells, with multiple mutations and multiple and/
or redundant pathways involved, including cross-talk
between pathways. Bispecific antibodies are uniquely
designed to overcome deficiencies in the effectiveness
of monoclonal antibodies. By targeting two different
receptors on the same cell or by targeting two ligands,
the bispecific antibody can block two signaling
pathways involved in cancer cell proliferation and the
inflammatory response. This multi-targeted approach
is highly effective in destroying tumor cells. Bispecific
antibodies can also be engineered to guide immune
effector cells such as Natural Killer (NK) cells or T-cells
to bind tumor cells, leading to their destruction. [1, 2]
Currently in Clinical Development
There are more than 60 different bispecific antibody
formats, developed to match a variety of clinical
applications. There are 30 plus bispecific antibodies in
clinical development as therapeutics for a wide variety
of cancer types, as well as several diseases. Many of
these bispecific antibodies are designed to redirect
T cells to kill tumor cells, while others are designed
to bind two different disease mediators (i.e., soluble
ligands, cell surface receptors, and other proteins).
For several non-oncology applications - rheumatoid
arthritis, idiopathic pulmonary fibrosis, and psoriasis
- the bispecific antibodies function as a blockade of
pro-inflammatory cytokines. [3]
One of the frontrunners, currently on the market for
the treatment of refractory B-cell precursor acute
lymphoblastic leukemia, is Amgen’s Blincyto®
(blinatumomab). Blincyto simultaneously targets the
T-cell surface protein complex CD3 and the tumorassociated antigen CD19, and so it serves as a
mediator to redirect immune effector cells to tumor
cells, in order to facilitate the destruction of the latter.
[1]
Another bispecific antibody approved for therapy
is Fresenius Biotech’s catumaxomab (Removab®),
for the treatment of malignant ascites. Removab
simultaneously targets the epithelial cell adhesion
molecule (EpCAM) and CD3. This effectively brings
into close proximity tumor cells, which express EpCAM,
and T-cells which express CD3. Also, the Fc domain
of Removab interacts with other immune system
effectors such as natural killer cells, macrophages,
and dendritic cells. The close proximity of the tumor
cells with the immune cells promotes activation of
immune cells, and subsequent destruction of cancer
cells. [4]
The two major classes of bispecific antibodies are small
single chain Fv (scFv)-based bispecific antibodies
and the larger immunoglobulin-G (IgG)-like bispecific
antibodies. [2] Each class has unique benefits, but also
drawbacks with development potential. The more than
60 formats that fall within these two larger categories
vary in many ways including their pharmacokinetic
half-life, molecular weight, spatial relationship
between different binding sites, number of antigenbinding sites, valency for each antigen, and ability to
support secondary immune functions. [1] One of the
benefits of the IgG-like bispecific antibodies is that
they retain Fc-mediated effector functions, and the Fc
region enhances purification, stability and solubility.
The larger size of the IgG-like bispecific antibodies
results in longer half-lives, while the smaller scFvbased bispecific antibodies have improved tissue
penetration.[3]
Improvements Needed in Antibody Engineering and
Development
With many companies jockeying for success in clinical
trials, there are some critical hurdles to overcome and
need for innovation and improvement. Throughout
the antibody engineering and development process of
new constructs, manufacturability issues such as low
expression yields and product instability/short halflife have hindered development. [3]
A big challenge is in rapid discovery of potent lead
bispecific antibodies and their targets, which requires
a high-throughput approach. There is also a need
for rapid purification of clinical-grade bispecific
antibodies. Immunogenicity introduces complexities
in drug design and development, and adverse effects
from immunogenicity, mainly caused by a “cytokine
storm,” can stifle clinical trials. [3]
Newer formats with greater potential for developability
may overcome these problems. Here we’ll examine
the most recent strategies in bispecific antibody
engineering.
Optimizing Target/Ligand Interactions
In a powerful new strategy to aid engineering and
selection efforts, Rhodin et al. of Lilly Research
Laboratories developed mathematical modeling
parameters to make predictions about how engineered
antibody properties will affect binding to cell surface
antigens. [5] Their model encompassed several
parameters: antibody affinity, valency, and antigen
density.
Intriguingly, the model predicted that for bispecific
antibodies, the antigen density has a very strong effect
on the binding affinity of the lowest expressing antigen.
Rhodin et al. was able to predict the magnitude of
this ratio based upon the expression
ratio of the two antigens being
targeted. To confirm their predictions,
they validated experimentally using
bispecific antibodies with different valencies on a
variety of cell surface antigens, including EGFR and
MET. The authors developed a conceptual model
through which antigen density has a strong effect on
binding to the less predominant antigen. This occurs
with the initial binding event, in which the antibody is
more likely to encounter the more highly expressed
antigen because of its elevated abundance. After
this initial binding event, the second arm is located
in a constrained position near the cell surface which
allows rapid binding to its target antigen.
Another novel strategy for development and quality
control is monitoring target/ligand binding of bispecific
antibodies through surface plasmon resonance
(SPR). While an enzyme-linked immunosorbent assay
(ELISA) is more commonly used to assess ligand
binding, it does not provide dynamic output, as only
endpoint data can be obtained. In contrast, SPR, which
is label-free, allows users to view the dynamics of
bispecific antibody binding and dissociation events
with two targets. SPR systems such as Biacore (GE
Healthcare) are label free, the readout is continuous,
and data analysis can be done at multiple time points,
or by fitting entire binding curves to determine affinity
and kinetic parameters. SPR can detect even weak
binding interactions. [6]
Half-life Extension Strategies
Small chain (scFv)-based bispecific antibodies
have some advantages over the larger sized IgGlike bispecific antibodies, including enhanced tissue
penetration as well as easier manufacturing. Their
smaller size affords the ability to bind epitopes that
may be sterically inaccessible to IgG-like bispecific
antibodies, and they are less immunogenic due to the
lack of an Fc region. However, the short half-life of
scFv-based bispecific antibodies is a major drawback
compared to that of IgG-like bispecific antibodies. A
short half-life leads to issues in clinical applications
such as fast off-rates, rapid blood clearance, and poor
retention time at target sites, which limits potency and
increases the number of therapeutic doses required.
[3]
One strategy for increasing half-life is to attach flexible,
hydrophilic molecules, such as a single PEG chain(s)
using a site-directed approach. This effectively
increases the hydrodynamic volume, which increases
the half-life. [7]
A more widely adopted technique is to fuse scFvbased bispecific antibodies to human serum albumin
or alternatively, an albumin-binding moiety. This
increases half-life through multiple mechanisms:
by increasing the molecule size and promoting the
recycling of the bispecific antibodies. Another option
for antibody engineers is to fuse an Fc fragment to
scFv-based bispecific antibodies. This also has the
dual effect of increasing the size of the antibody and
promoting recycling. [3]
In another novel approach, human mesenchymal
stromal cells (MSCs) can be genetically modified
to produce and secrete bispecific antibodies
continuously throughout the lifetime of the patient. [8]
MSCs have a plethora of beneficial properties which
make them uniquely adapted for this purpose. For one,
they accumulate next to tumors, including metastatic
tumors. They have low levels of immunogenicity.
Other features of MSCs are easy transduction by viral
vectors, in vitro expansion, and long life in vivo. [8, 9]
A Wide Spectrum of Upcoming Applications for
Bispecific Antibodies
Beyond the current applications for bispecific
antibodies discussed here, there are several other
exciting applications now in preclinical development.
One unique application for bispecific antibodies
is in delivery of therapeutic antibodies across the
blood-brain barrier for neurological conditions, as
monospecific antibodies are unable to cross this
barrier.[10] Another unique application for bispecific
antibodies is in the delivery of drug, nanoparticle or
radiolabel payloads to tumor sites. The bispecific
antibody serves to concentrate these payloads at the
tumor site, significantly enhancing serum retention
time. [3]
Bispecific antibodies are being developed for use in
diagnostic assays, functioning as a cross-linker to
bind a reporter molecule and antigen simultaneously.
As an example, a monospecific capture antibody
can be attached to a solid surface, which is used to
capture a target antigen in serum. As a next step, a
reporter molecule and bispecific antibody are added,
which bind the bound antigen, and thus identification
can be made. These bispecific antibody-based
immunoassays are in development for diagnosis
of patients with various infectious diseases: SARS,
hepatitis B, tuberculosis, as well as E. coli infections.
[11]
Another application involves tackling the rising
threat of antibiotic resistant bacteria through protein
engineering. A new bispecific antibody was engineered
to be effective against Pseudomonas aeruginosa
infections, which causes pneumonia and mortality
in cystic fibrosis patients, and is resistant to single
epitope antibiotics. This novel bispecific antibody
binds with one arm to an extracellular polysaccharide
that is important for immune evasion and biofilm
formation and with the other arm, to a virulence factor.
It’s been proven effective in animal studies, and is now
in clinical studies.[12, 13]
Bispecific antibodies have made a rapid entry into the
antibody market, with major applications in oncology
and a variety of diseases/disorders. The focus in
oncology has been in either blocking multiple and
redundant signaling pathways involved in oncogenesis
or redirecting immune effector cells to be in close
proximity to tumor cells. In non-oncology applications,
a major developmental effort has gone into blocking
pro-inflammatory cytokines. Improvements to the
antibody design, engineering and manufacturing
process are needed and are being addressed. A
plethora of exciting applications are underway in this
fast-moving arena.
References
1. S
piess, C., Q. Zhai, and P.J. Carter, Alternative molecular
formats and therapeutic applications for bispecific antibodies.
Mol Immunol, 2015. 67(2 Pt A): p. 95-106.
2.Spasevska I, D.M., Klein C, Dumontet C, Advances in Bispecific
Antibodies Engineering: Novel Concepts for Immunotherapies.
J Blood Disord Transfus 2015. 6(243).
3. F
an, G., et al., Bispecific antibodies and their applications.
Journal of Hematology & Oncology, 2015. 8(1): p. 1-14.
4. F
resenius Biotech. Removab (catumaxomab) Product
Monograph-Removab in Malignant Ascites 2009; Available
from: http://removab.com/tl_files/media/PDFs/110408_
Removab_Produktmono_engl_V4.pdf.
5. R
hoden, J.J., G.L. Dyas, and V.J. Wroblewski, A Modeling and
Experimental Investigation of the Effects of Antigen Density,
Binding Affinity, and Antigen Expression Ratio on Bispecific
Antibody Binding to Cell Surface Targets. J Biol Chem, 2016.
6. K
arllson, R., Applications of Surface Plasmon Resonance
for Detection of Bispecific Antibody Activity. Biopharm
International, 2015. 28(10): p. 38-45.
7. Kontermann, R.E., Strategies for extended serum half-life of
protein therapeutics. Curr Opin Biotechnol, 2011. 22(6): p. 868-76.
8. A
liperta, R., et al., Bispecific antibody releasing-mesenchymal
stromal cell machinery for retargeting T cells towards acute
myeloid leukemia blasts. Blood Cancer Journal, 2015. 5: p.
e348.
9. L
e Blanc, K., et al., HLA expression and immunologic
propertiesof differentiated and undifferentiated mesenchymal
stem cells. Experimental Hematology, 2003. 31(10): p. 890896.
10. C
ouch, J.A., et al., Addressing Safety Liabilities of TfR
Bispecific Antibodies That Cross the Blood-Brain Barrier.
Science Translational Medicine, 2013. 5(183): p. 183ra57183ra57.
11. B
yrne, H., et al., A tale of two specificities: bispecific
antibodies for therapeutic and diagnostic applications.
Trends Biotechnol, 2013. 31(11): p. 621-32.
12. K
ingwell, K., Infectious diseases: Two-hit antibody tackles
bacteria. Nat Rev Drug Discov, 2015. 14(1): p. 15-15.
13. D
iGiandomenico, A., et al., A multifunctional bispecific
antibody protects against Pseudomonas aeruginosa. Science
Translational Medicine, 2014. 6(262): p. 262ra155-262ra155.