SlideShare ist ein Scribd-Unternehmen logo
1 von 9
Downloaden Sie, um offline zu lesen
E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9

available at www.sciencedirect.com

www.elsevier.com/locate/yexcr

Review

Introduction to current and future protein therapeutics: A
protein engineering perspective
Paul J. Carter⁎
Department of Antibody Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA

A R T I C L E I N F O R M A T I O N

A B S T R A C T

Article Chronology:

Protein therapeutics and its enabling sister discipline, protein engineering, have emerged since the

Received 18 January 2011

early 1980s. The first protein therapeutics were recombinant versions of natural proteins. Proteins

Revised version received

purposefully modified to increase their clinical potential soon followed with enhancements

20 February 2011

derived from protein or glycoengineering, Fc fusion or conjugation to polyethylene glycol.

Accepted 24 February 2011

Antibody-based drugs subsequently arose as the largest and fastest growing class of protein

Available online 1 March 2011

therapeutics. The rationale for developing better protein therapeutics with enhanced efficacy,
greater safety, reduced immunogenicity or improved delivery comes from the convergence of

Keywords:

clinical, scientific, technological and commercial drivers that have identified unmet needs and

Protein therapeutics

provided strategies to address them. Future protein drugs seem likely to be more extensively

Antibody therapeutics

engineered to improve their performance, e.g., antibodies and Fc fusion proteins with enhanced

Fc fusion proteins

effector functions or extended half-life. Two old concepts for improving antibodies, namely

Engineered protein scaffolds

antibody-drug conjugates and bispecific antibodies, have advanced to the cusp of clinical success.
As for newer protein therapeutic platform technologies, several engineered protein scaffolds are in
early clinical development and offer differences and some potential advantages over antibodies.
© 2011 Elsevier Inc. All rights reserved.

Contents
Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .
Brief history of protein therapeutics . . . . . . . . . . . . . . . . . . .
Protein therapeutics enabled by protein engineering . . . . . . . . .
Insulin — the first recombinant human protein therapeutic . . . . .
Beginning of engineered proteins as therapeutics . . . . . . . . . . .
Fc fusion proteins . . . . . . . . . . . . . . . . . . . . . . . . . . .
Protein conjugation to polyethylene glycol . . . . . . . . . . . . . .
The rise of antibody therapeutics . . . . . . . . . . . . . . . . . . .
Rationale for next generation protein therapeutics . . . . . . . . . . . .
Strengths and limitations of antibody and other protein therapeutics
Clinical rationale . . . . . . . . . . . . . . . . . . . . . . . . . . .
Scientific rationale . . . . . . . . . . . . . . . . . . . . . . . . . . .

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

⁎ Fax: +1 650 467 8318.
E-mail address: pjc@gene.com.
Abbreviations: FDA, Food and Drug Administration; PEG, polyethylene glycol; TPO, thrombopoietin
0014-4827/$ – see front matter © 2011 Elsevier Inc. All rights reserved.
doi:10.1016/j.yexcr.2011.02.013

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

.
.
.
.
.
.
.
.
.
.
.
.

1262
1262
1262
1262
1263
1263
1263
1263
1264
1264
1264
1264
1262

E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9

Commercial rationale . . . . . . . . . . . . . . . . . . . .
Platform technologies for next generation protein therapeutics
Next generation antibodies and Fc fusion proteins . . . . .
Engineered protein scaffolds . . . . . . . . . . . . . . . .
Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . .
References . . . . . . . . . . . . . . . . . . . . . . . . . . . .

Introduction
Since the early 1980s proteins have emerged as a major new class
of pharmaceuticals with ~200 marketed products that are mainly
therapeutics with a small number of diagnostics and vaccines [1].
Protein therapeutics can be classified based upon their pharmacologic activity as drugs that: i) replace a protein that is deficient
or abnormal, ii) augment an existing pathway, iii) provide a novel
function or activity, iv) interfere with a molecule or organism, or
iv) deliver a payload such as a radionuclide, cytotoxic drug, or
protein effector [2]. Alternatively, protein therapeutics can be
grouped into molecular types that include: antibody-based drugs,
anticoagulants, blood factors, bone morphogenetic proteins,
engineered protein scaffolds, enzymes, Fc fusion proteins, growth
factors, hormones, interferons, interleukins, and thrombolytics
(Fig. 1) [1,3]. Antibody-based drugs are the largest and fastest
growing class of protein therapeutics with 24 marketed antibody
drugs in the USA [4] and over 240 more in clinical development [5].
This introductory article provides a brief history of protein
therapeutics, offers a rationale for developing better protein drugs,
and identifies some major future opportunities as well as emerging
technologies that may meet them. The 30-year history of protein
drugs is illustrated here with examples that pioneered their class or
have high clinical or commercial significance. The rationale for
creating better protein drugs, including antibodies [6], comes from
the convergence of clinical, scientific, technological and commercial drivers that collectively have identified major unmet needs and
provide strategies to address them. Major future opportunities for
protein therapeutics, are improved efficacy, greater safety, reduced
immunogenicity or improved delivery. A plethora of emerging
technologies for the generation and optimization of protein therapeutics are anticipated to help address these opportunities. Space

Recombinant
proteins

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

.
.
.
.
.
.

1265
1265
1265
1266
1267
1267

Brief history of protein therapeutics
Protein therapeutics enabled by protein engineering
Protein therapeutics and its enabling sister discipline, protein
engineering, have emerged since the early 1980s. Protein engineering by rational design or molecular evolution allows the
systemic dissection of protein structure–function relationships and
the generation of novel proteins with modified activities or entirely
new properties. Indeed, protein engineering has revolutionized
protein therapeutics by providing the tools to customize existing
proteins or to create novel proteins for specific clinical applications.
A brief history of protein therapeutics is provided here from a
protein engineering perspective. The focus is on technologies with
the greatest impact on current protein therapeutics — engineered
versions of natural proteins, Fc fusion proteins, conjugation to the
polymer, polyethylene glycol (PEG), and antibody therapeutics as
the quintessential protein therapeutic platform.

Insulin — the first recombinant human protein therapeutic
The first human protein therapeutic derived from recombinant
DNA technology was human insulin (Humulin®) created at
Genentech [7], developed by Eli Lilly, and approved by the US
Food and Drug Administration (FDA) in 1982. Recombinant insulin

Antibodies
Fab fragment

Conjugate PEG
Engineer protein
Engineer glycosylation

.
.
.
.
.
.

constraints limit examples here to a sampling of technologies of
broad applicability — so called platform technologies. The interested reader is referred to many excellent articles on protein
therapeutics including those in this special issue and others cited
herein.

Anticoagulants
Blood factors
Bone morphogenetic
proteins
Enzymes
Growth factors
Hormones
Interferons
Interleukins
Thrombolytics
Optional
modifications

.
.
.
.
.
.

Fc fusion
proteins
IgG
Receptor
Ligand
Peptide

Fc

Conjugate PEG

Bispecific
Conjugate drug
Conjugate radionuclide

Fig. 1 – Overview of currently marketed protein therapeutics from a protein engineering perspective.
E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9

was used to replace its natural counterpart that can be deficient in
diabetes mellitus. Other recombinant human proteins were soon
developed as therapeutics to replace the natural proteins deficient
in some patients (e.g., growth hormone) or augment existing
pathways (e.g., interferon-α, tissue plasminogen activator, and
erythropoietin) [2].

Beginning of engineered proteins as therapeutics
Beyond recombinant versions of natural proteins, the next major
step in the evolution of protein therapeutics was the redesign of
proteins to increase their clinical potential. Injected insulin fails to
adequately mimic endogenous insulin in two major ways: rapid
onset of action after meals and prolonged maintenance of low level
insulin between meals [8]. Insulin has been engineered, commonly
by one to three amino acid replacements, to create analogs that are
either rapid-acting or long-acting and better mimic different
properties of endogenous insulin [8]. Multiple insulin analogs have
been approved for the treatment of diabetes mellitus including the
rapid-acting analogs, insulin lispro (Humalog®), insulin aspart
(NovoRapid® and Novolog®) and insulin glulisine (Apidra®) and
the long-acting analogs, insulin glargine (Optisulin® and Lantus®)
and insulin detemir (Levemir®) [1,8]. Insulin analogs offer multiple benefits over first generation recombinant insulin drugs that
include improved physiologic profile, greater convenience for
patients, reduced risk of hypoglycemia, and less weight gain in
some cases [9]. The commercial importance of engineered insulins
is illustrated by Lantus®, which was one of the top ten selling
biopharmaceuticals in 2009 [1].
Many protein therapeutics are produced in mammalian cells
and have one or more N- and/or O-linked glycans [10]. Engineering
a protein with additional glycosylation sites can increase the in
vivo serum half-life and thereby target exposure [11]. An early
example of such “glycoengineering” of proteins for half-life
extension is provided by darbepoetin-α (Aranesp®), administered
to anemic patients to stimulate the production of red blood cells in
the bone marrow. The first generation drug, epoetin-α, has 3 Nlinked glycosylation sites and was engineered with 2 additional
glycosylation sites to create darbepoetin-α, a commercially
successful longer-acting second generation drug [12]. Protein
glycosylation can sometimes reduce bioactivity, albeit offset by
longer half-life. Darbepoetin-α has higher specific activity in
patients than epoetin-α, reflecting that the 3-fold increase in
serum half-life that increases drug exposure more than offsets a 5fold reduction in receptor binding affinity [12].

1263

Beyond half-life extension, Fc fusion can provide several
additional benefits such as facilitating expression and secretion
of recombinant protein, enabling facile purification by protein A
chromatography, improving solubility and stability, and enhancing
potency by increasing valency. Optional additional properties
conferred by Fc fusion include binding to Fcγ receptors and/or
complement to support secondary immune functions [13,14]. The
first demonstration of the Fc fusion protein concept was CD4-Fc
[16]. Five Fc fusion protein therapeutics were approved by 2010,
the prototypical and most successful drug of this class being
etanercept (Enbrel®), a TNF receptor 2-Fc fusion protein [1].
Etanercept was the top-selling protein therapeutic in 2009 with
$6.6 billion in world-wide sales in rheumatoid arthritis and other
autoimmune diseases [1]. Romiplostim (Nplate®) is the first
peptide-Fc fusion (“peptibody”) approved as a human therapeutic.
Romiplostim is a thrombopoietin (TPO) receptor agonist approved
as a treatment for thrombocytopenia [17].

Protein conjugation to polyethylene glycol
Protein conjugation to chemicals such as radionuclide chelators,
cytotoxic drugs and PEG can endow proteins with brand new
capabilities to increase their clinical potential.
The most clinically and commercially successful protein
conjugates thus far have been with PEG. Protein conjugation to
PEG – “PEGylation” – increases their hydrodynamic volume,
prevents rapid renal clearance, and thereby increases the serum
half-life, as extensively reviewed [15,18,19]. At least 8 PEGylated
proteins are approved as human therapeutics [18]. Indeed, longacting second generation interferon-α molecules that are PEGylated have largely superseded their first generation non-PEGylated
counterparts [20]. Benefits of PEGylation are that it is possible to
tune the serum-half of proteins by varying the number of attached
PEG molecules, their size and extent of branching [21]. PEGylation
can also reduce the immunogenicity of proteins [18,19], although
PEGylated proteins can still sometimes be immunogenic [22]. A
draw back to PEGylation is that it can impair protein function, a
limitation that is offset by the greatly increased half-life, and
potentially ameliorated by site-specific PEGylation and/or tailoring
the PEG [18,19]. PEG is a non-natural and apparently nonbiodegradable polymer, a molecular attribute that has unclear
clinical significance. PEGylated proteins can induce renal tubular
vacuolation in animals, albeit with unaltered clinical pathology
[23] and unknown impact on long term safety.

The rise of antibody therapeutics
Fc fusion proteins
Another significant advance in protein therapeutics came with the
advent of fusion proteins by joining the genes encoding 2 or more
different proteins. Fusion proteins combine properties of their
component parts. The most clinically and commercially successful
fusion protein therapeutics to date contain the Fc region of
immunoglobulins [1]. Such Fc fusions can endow peptides or
proteins with the IgG-like property of long serum half-life (days to
weeks), by virtue of binding to the salvage receptor, FcRn [13,14]. By
contrast, proteins of ~70 kDa and smaller are typically eliminated
rapidly from circulation by renal filtration and have half-lives of a
few minutes to a few hours, that can in many cases render them
marginal or unsuitable for therapeutic applications [15].

Antibody-based drugs are the largest and fastest growing class of
protein therapeutics with 24 marketed antibody therapeutics in
the USA (28 FDA-approved and 4 later withdrawn from the
market) [4] and at least 240 more in clinical development [5].
Antibody-based drugs contributed $38 billion of $99 billion in
world-wide sales for protein biopharmaceuticals in 2009 [1].
Moreover, 5 of the 10 top-selling protein therapeutics in 2009
were antibodies, namely, infliximab (Remicade®), bevacizumab
(Avastin®), rituximab (Rituxan® and MabThera®), adalimumab
(Humira®) and trastuzumab (Herceptin®) [1].
The rapid growth of antibody therapeutics since the mid-1990s
was made possible by the advent of antibody chimerization
and humanization [24] technologies that largely overcame the
1264

E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9

limitations of rodent monoclonal antibodies as drugs, as well as
the emergence of facile routes to human antibodies using
transgenic mice [25,26] or phage display [25,27] technologies.
Other advances that have contributed to the success of antibodies
as therapeutics include the industrialization of antibody production technologies [28] that can provide sufficient quantities of
antibodies to allow higher doses to be used in patients. The development of methods to express and secrete functional antibody
fragments from E. coli [29,30] has significantly stimulated the field
of antibody engineering and enabled subsequent technologies
such as phage display. The next major advance in the expression of
antibody fragments in E. coli was high titer (gram per liter)
production in a fermentor [31], that facilitated the development of
antibody fragment drugs such as the anti-VEGF Fab, ranibizumab
(Lucentis®). The emergence of antibody as therapeutics likely also
reflects improved choices of antibody and target antigen for
intervention in the pathobiology of disease.
The majority of approved antibody drugs are unmodified
(“naked”) IgG molecules. However, other antibody formats have
been approved for therapy including IgG “armed” with radionuclides or cytotoxic drugs (antibody-drug conjugates) for cancer
treatment, as well as Fab fragments and a PEGylated Fab′ fragment
[1].

Rationale for next generation protein therapeutics
A major quest with protein therapeutics, especially antibodies
[6,32], is the development of even better next generation drugs
with enhanced efficacy, greater safety or improved delivery.
Current antibody therapeutics reveal major strengths that provide
a foundation upon which to build, as well as significant limitations
to overcome that offers new opportunities [6,33]. Some antibody
strengths and many of their limitations apply more broadly to
protein therapeutics and are discussed below as a compass to guide
the development of next generation protein therapeutics. The
impetus to develop better protein therapeutics is also discussed
here as a convergence of clinical, scientific, and commercial forces
that identify unmeet needs (this section), in conjunction with
technological tools that many help address them (next section).

Strengths and limitations of antibody and other protein
therapeutics
One of the greatest strength of antibodies as a therapeutic platform
is the potential ease of generating high affinity and specificity
human antibodies to virtually any target of therapeutic interest,
e.g., using transgenic mice [25,26] or phage display [25,27]
technologies. Moreover, antibodies are readily tailored for therapeutic applications by modulating (enhancing or attenuating)
their existing properties or endowing them with new activities as
discussed in a later section and reviewed more extensively
elsewhere [33,34]. As for other protein therapeutics, antibodies
are commonly well tolerated by patients, but not invariably so.
Antibody drug development benefits from recombinant production methods that are well-established, broadly applicable and
commonly support expression at high titer (multi-gram per liter)
[28]. Widely available knowledge and experience across all facets
of antibody drug development also facilitates the generation of
future antibody therapeutics. An advantage of antibodies over

other drug formats is that the success rate has been somewhat
higher — 17% for humanized antibodies from the first clinical trial
to regulatory approval [35].
Proteins have several significant limitations as therapeutics. For
example, protein therapeutics are very expensive, reflecting high
production costs, that may limit patient access and also clinical
applications [28]. This high cost issue is exacerbated for protein
therapeutics where multi-gram doses are needed for a treatment
course, as is the case for some antibodies. Intracellular delivery of
proteins is possible in the laboratory setting, e.g., by protein
transduction [36]. However, targeted pharmaceutic delivery of
proteins to reach intracellular targets is in its infancy, so protein
therapeutics are currently limited to cell surface or extracellular
targets.
Another shortcoming of proteins therapeutics is that they are
denatured and/or proteolyzed in the gut and are thus not orally
bioavailable. Proteins, including antibodies [37], show very poor
ability to penetrate the specialized endothelial structure of the
neurovasculature known as the blood-brain barrier. The integrity
of the blood-brain barrier can be partially compromised in some
pathological settings such as chronic neurodegenerative diseases
[38]. Nevertheless, proteins with current technologies are poorly
suited to the treatment of chronic neurodegenerative diseases or
tumors that originate or have metastasized to the brain.
Antibodies are relatively large (IgG ~150 kDa) compared to
other proteins and this large size may limit their tissue penetration
and ability to localize to their targets. For example, antibody
localization to tumors in patients is typically very inefficient:
≤0.01% of the injected dose per gram of tumor [39]. Comparison of
mathematical modeling with experimental data, strongly suggests
that molecular size and binding affinity are major determinants of
tumor targeting [40]. Many other factors likely contribute to tumor
penetration including antigen expression level and turnover rate
[41].

Clinical rationale
From a clinical standpoint, first generation protein drugs can
provide substantial patient benefit. However, in many cases there
is room for improvement in efficacy and/or safety. For example,
several anti-cancer antibodies provide an overall survival benefit,
including bevacizumab [42] and trastuzumab [43] in combination
with chemotherapy in metastatic colorectal and breast cancers,
respectively. Nevertheless, there remains plenty of opportunity for
enhancing antibodies for cancer treatment, such as elevating the
overall survival rate of patients, as well as many strategies being
explored to do so [44]. All therapeutic proteins are potentially
immunogenic in patients and occasionally an anti-drug antibody
response can lead to a major safety issue. For example, a truncated
and PEGylated form of TPO (MGDF-PEG) elicited an antibody
response in some patients that neutralized endogenous TPO
leading to severe and persistent anemia, i.e., the opposite of the
desired effect of increasing platelet counts [22]. This issue was
overcome with the peptibody, Romiplostim (see Fc fusion proteins
section) [17].

Scientific rationale
Scientifically, our expanding knowledge suggests drug mechanisms
of action to enhance, and mechanisms of resistance or toxicity to
E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9

overcome. For example, anti-CD3 antibodies illustrate the benefits of
understanding and overcoming mechanisms of drug toxicity and
resistance. In 1986 the anti-CD3 antibody, muromonab-CD3
(OKT3®), became the first antibody approved by the FDA for
human therapy — for the treatment of acute allograft rejection in
renal transplant patients. In 1993 muromonab-CD3 was approved
for the treatment of acute rejection of heart and liver transplants in
patients resistant to steroid treatment. Unfortunately, muromonabCD3 activates T cells resulting in transient cytokine release and acute
and severe flu-like symptoms [45]. Another major limitation of
muromonab-CD3 is that its therapeutic benefit is reduced by
neutralizing antibodies that develop in ~50% of patients [45].
Muromonab-CD3 has since been voluntarily withdrawn from the
US market due to decreased sales. These major limitations of
muromonab-CD3 have been greatly reduced, if not overcome, in
some second generation anti-CD3 antibodies that have advanced to
late stage clinical development [6]. For example, the anti-CD3
antibodies, teplizumab and otelixizumab, combine Fc engineering to
attenuate Fcγ receptor binding with chimerization or humanization
to reduce immunogenicity [34].
Drug development, including for protein therapeutics, seems
likely to gain from advances in the understanding of targets and
their role in the pathobiology of disease. For example, there is a
growing appreciation that targets are not isolated gene products
but integral components of networks of interconnected signaling
pathways [46,47].

Commercial rationale
Clinical and commercial success with protein therapeutics has
bred intense competition between different organizations striving
to develop protein therapeutics to the same antigen and/or
overlapping therapeutic indications. Indeed, this is already
reflected in marketed drugs as illustrated by 5 different protein
therapeutics that block TNF including 3 different IgG (infliximab,
adalimumab and golimumab), a PEGylated Fab′ fragment (certolizumab pegol) and a receptor-Fc fusion (etanercept) [6]. Such
competition is increasingly commonplace as patents dominating
individual targets expire. The generation of high affinity and
potency IgG is often possible, arguably commoditized. This
provides a strong commercial incentive to develop superiorperforming antibody drugs to differentiate products from those of
competitors. The success of innovator protein therapeutics has
brought a rise in biosimilar efforts (i.e., products that are deemed
highly similar to innovator protein therapeutics according to drug
regulatory authorities). Indeed, several biosimilar approval applications have been filed in the European Union with 13 biosimilars
being approved by European drug regulators by early 2009 [48].
For creators of innovator drugs, the impetus to develop improved
next generation protein therapeutics that out perform the
previous generation drugs has increased substantially [48].

Platform technologies for next generation
protein therapeutics
Many protein engineering tools have been developed that allow
one to optimize favorable properties of proteins, attenuate
undesired attributes and create proteins with entirely novel
activities. Space constraints limit the discussion here to next

1265

generation antibody and Fc fusion protein therapeutics and the
emerging technologies of engineered protein scaffolds.

Next generation antibodies and Fc fusion proteins
Many promising strategies are emerging to enhance the existing
properties of antibodies or to endow them with new activities, as
reviewed in detail elsewhere [6,32,33]. Antibodies (IgG) have
multiple binding partners including antigen, Fcγ receptors,
complement and FcRn. Each of these interactions can be tailored
in ways that may improve the clinical properties of antibodies
[32,34,49]. Platform technologies that seem likely to impact the
next generation of antibody therapeutics include old concepts that
are finally coming to fruition, such as antibody-drug conjugates
and bispecific antibodies. Technologies for improving Fc-mediated
functions, such as half-life extension and effector function enhancement, seem likely to benefit the next generation of antibody
therapeutics. Additionally, these technologies appear directly
applicable to the optimization of Fc fusion proteins.
The 50-year old concept of bispecific antibodies [50] came of
age in 2009 with the first regulatory approval of a bispecific
antibody — catumaxomab (Removab®, anti-EpCAM x anti-CD3) in
Europe. Catumaxomab is approved for the treatment of malignant
ascites in patients with EpCAM-positive carcinomas [1,51].
Recruitment of T cells via the anti-CD3 arm of catumaxomab and
other immune effector cells via its Fc region may contribute to
antitumor activity. A tandem bispecific scFv format known as a
“BiTE”, that binds CD3 and a tumor-associated antigen, is a
particularly effective way to retarget T cells to kill tumor cells.
Blinatumomab, a BiTE specific for CD19 and CD3, gave several
complete responses in a phase I clinical trial in non-Hodgkin's
lymphoma [52] and is currently in multiple phase II clinical trials.
Another major area of current interest with bispecific antibodies is
in the simultaneous blockade of 2 disease mediators, which may
lead to greater therapeutic benefit than targeting individual
disease mediators [53]. Until recently, a major bottleneck in the
development of bispecific antibodies as therapeutics was the
challenge of producing these complex molecules in sufficient
quantity and quality for clinical applications. This issue has been
largely overcome by the advent of many alternative formats and
production methods for bispecific antibodies [6].
Antibody-drug conjugates, a concept that dates back to the
1970s, use antibodies to deliver a cytotoxic payload to kill a target
such as tumor cells [54,55]. One antibody-drug conjugate,
gemtuzumab ozogamicin (Mylotarg®), was approved by the FDA
for the treatment of acute myeloid leukemia. Gemtuzumab
ozogamicin was withdrawn from the market in 2010 due to safety
concerns and insufficient patient benefit in post approval clinical
trials. Much progress has been made over the decades in many
different facets of antibody-drug conjugates, including better
choices of target antigen and antibody, and the development of
more potent drugs and linkers of greater stability [54,55]. Pivotal
clinical trials are in progress for brentuximab vedotin (SGN-35,
[56]) in Hodgkin's disease and trastuzumab emtansine (T-DM1,
[57]) in metastatic breast cancer over-expressing HER2/neu
following demonstration of robust antitumor activity and acceptable safety profiles in earlier clinical trials.
Engineering proteins to increase their serum half-life has the
potential benefits of increased exposure leading to greater target
localization and greater efficacy, lower or less frequent dosing and
1266

E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9

lower cost. In the case of antibodies, the serum half-life in mice can
be extended by at least a few fold by engineering the Fc region for
higher affinity binding to the salvage receptor (FcRn), whilst
preserving the pH dependence of binding [58]. Half-life extension
of Fc-engineered antibodies in non-human primates was subsequently demonstrated by several different groups [59]. Antibody
half-life extension can result in increased target exposure and
improved preclinical in vivo efficacy as recently shown for antiEGFR and anti-VEGF antibodies [60]. Engineering antibodies for
pH-dependent interaction with their target antigen has also been
shown to extend antibody half-life in vivo [61]. The first half-life
extended antibody to enter clinical trials was the humanized antiRSV antibody, MEDI-557, although pharmacokinetic data have yet
to be reported.
Another major area of interest for improving antibody performance is effector function enhancement, as reviewed extensively elsewhere [34,62] and discussed briefly below. Antibody
effector functions include ADCC (antibody-dependent cellular
cytotoxicity), ADCP (antibody-dependent cellular phagocytosis)
and complement-dependent cytotoxicity (CDC). Effector functions
may be advantageous where destruction of the target cells is
desired, as is often the case for oncology targets.
Indeed many antibodies approved for oncologic indications
(e.g., rituximab, trastuzumab, alemtuzumab, cetuximab and
ofatumumab) support one or many effector functions that may
contribute to their clinical antitumor activity, with the strongest
evidence being for rituximab [63].
For antibody and Fc fusion protein therapeutics potential
benefits from enhancing effector functions include greater efficacy,
lower or less frequent dosing and decreased cost-of-goods. As for
risks, these include more frequent or severe adverse events. ADCC,
ADCP and CDC have been augmented by Fc protein engineering as
extensively reviewed elsewhere [62]. ADCC has also been
augmented to a similar level by modifying the glycan attached to
the Fc, e.g., afucosylation achieved through cell line engineering
[62]. At least 8 effector function enhanced antibodies have
advanced into early clinical development [34].

Engineered protein scaffolds
The success of antibodies as protein therapeutics has inspired the
development of so-called “engineered protein scaffolds” as alternative platform technologies for the development of novel next
generation protein therapeutics [64–66]. Engineered protein scaffolds provide binding sites that can be tailored to specifically
recognize desired target molecules, in an analogous way that
antibodies can be developed to recognize their target antigens.
Here the basic features of engineered protein scaffolds are described,
together with their strengths, limitations and some possible future
directions.
Protein scaffolds chosen for engineering have known 3dimensional structures that together represent diverse protein
folds [64–66]. Protein scaffolds are commonly selected with the
following attributes: small, monomeric, high solubility and
stability, and that lack disulfide bonds and glycosylation sites.
Protein scaffolds are typically readily expressed in microbial hosts.
Sequence diversity is introduced into the protein scaffold, e.g., by
randomizing surface accessible residues, to create a so-called
library. Binders to a target of therapeutic interest are then identified by selection using technologies such as phage, yeast or

ribosome display. Increased binding affinity – affinity maturation –
is often needed and readily accomplished by further library
building and selection.
Over 50 different engineered protein scaffolds have been
described with a few advancing into clinical development [64–
66] including: Adnectins® from type III fibronectin domains,
Affibodies® from the synthetic Z domain of staphylococcal protein
A, Anticalins® from lipocalins, Avimers® from LDLR-A modules,
DARPins® from ankyrin repeat proteins, and engineered Kunitz
domains. Engineered protein scaffolds also include single-domain
antibodies from human (dAbs®) and camelids (Nanobodies®).
One engineered protein scaffold protein has been approved for
therapy, namely, ecallantide (Kalbitor®), a potent and selective
inhibitor of plasma kallikrein based upon a Kunitz domain [67].
Many potential advantages of engineered protein scaffolds over
antibodies have been proposed [64–66], albeit with limited
supporting evidence to date in the context of developing protein
therapeutics. For example, the smaller size and higher stability of
some engineered protein scaffolds may allow for alternative routes
of administration such as pulmonary delivery. The smaller size of
engineered protein scaffolds over antibodies favors more efficient
tissue penetration, but this can be more than offset by much faster
clearance. Engineered protein scaffolds may be more readily
expressed in microbial hosts reflecting their high stability and
solubility, small size, simple structure (single polypeptide), and
that they commonly lack disulfide bonds and/or glycosylation
sites. The small and modular architecture of engineered protein
scaffolds seems particularly well suited to the construction of
multivalent and/or multispecific protein therapeutics (Fig. 2).
Engineered proteins are commonly associated with independent
intellectual property that is outside the scope of antibody patents.
Several of these proposed advantages of engineered protein
scaffolds have been achieved with antibodies by engineering for
improved stability or expression, or by exploiting the modular
architecture of antibodies to create different formats including
ones that are smaller and simpler than IgG [6,32,33].
Engineered protein scaffolds lack Fc-mediated properties of
antibodies such as long serum half-life and effector functions. This
does not appear to be a major limitation, as many different
strategies have been developed to extend the half-life of proteins
as comprehensively reviewed [15]. For example, single-domain
antibodies binding to the long-circulating protein, serum albumin,
have been identified and fused to small proteins, including other
single-domain antibodies to extend their half-life [68,69]. As for
effector functions, these are desired in some clinical settings but
not others. Fc fusion offers a simple way to endow engineered
protein scaffolds with effector functions if needed. Engineered
protein scaffolds, like antibodies, offer significant opportunity for
sequence diversity but individual protein scaffolds appear to
provide less opportunity for structural diversity than do antibodies. The functional impact of lower structural diversity on the
ease of being able to generate binding proteins to specific sites on
targets of interests remains to be determined.
All protein therapeutics, including engineered protein scaffolds,
are potentially immunogenic in patients and may elicit an antidrug antibody response that may neutralize or otherwise interfere
with the drug's activity. This issue can be more significant for
chronic therapy involving repeated drug administration. Clinical
trials are ultimately needed to understand the extent to which
individual engineered protein scaffold drugs are immunogenic in
1267

E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9

Engineered scaffold
proteins

Representative
assemblies

Target
valencies

Target
specificities

Long
half-life

Target A binder

1

1

No

Target B binder

1

1

Yes

Target C binder

1, 1

2

Yes

Half-life extender

2, 1

2

Yes

1, 2

2

Yes

2, 2

2

Yes

1,1,1

3

Yes

Fig. 2 – Modular assembly of engineered protein scaffolds into multivalent and multispecific fusion proteins. Engineered scaffold
proteins binding to different targets or different epitopes on the same target are identified by selection from libraries. The modular
nature of engineered protein scaffolds allows them to be readily assembled into multimers with specific valency for each target
[64–66]. However, steric considerations may prevent all modules from being able to bind their respective targets simultaneously.
Serum half-life extension is commonly required for engineered scaffold proteins to obtain sufficient exposure to support
therapeutic applications. Many different half-life extension strategies for small proteins have been developed [15], including for
engineered protein scaffolds (shown), e.g., binding to serum albumin. Linkers between modules may be included but are omitted
for simplicity in this figure.

patients and whether or not this limits their clinical utility. Human
proteins are often selected for engineered protein scaffolds, which
may help reduce the risk of immunogenicity in patients for therapeutic applications.

Conclusions
Proteins are now well established as a clinically and commercially
important class of therapeutics. Experience with protein therapeutics has provided an understanding of their strengths and
limitations and ways in which they might be improved. Clinical
and commercial success with protein therapeutics provides strong
motivation to develop better protein drugs, whereas new tools,
especially platform technologies provide the likely means to do so.
The next generation of protein therapeutics will likely include
some of the many enhanced antibodies in early clinical development, optimized Fc fusion proteins, as well as new formats such as
engineered protein scaffolds.

REFERENCES

[1] G. Walsh, Biopharmaceutical benchmarks 2010, Nat. Biotechnol.
28 (2010) 917–924.
[2] B. Leader, Q.J. Baca, D.E. Golan, Protein therapeutics: a summary
and pharmacological classification, Nat. Rev. Drug Discov. 7
(2008) 21–39.
[3] N.C. Nicolaides, P.M. Sass, L. Grasso, Advances in targeted
therapeutic agents, Expert Opin. Drug Discov. 5 (2010)
1123–1140.
[4] J.M. Reichert, Metrics for antibody therapeutics development,
mAbs 2 (2010) 695–700.
[5] J.M. Reichert, Antibodies to watch in 2010, mAbs 2 (2010) 28–45.

[6] A.C. Chan, P.J. Carter, Therapeutic antibodies for autoimmunity
and inflammation, Nat. Rev. Immunol. 10 (2010) 301–316.
[7] D.V. Goeddel, D.G. Kleid, F. Bolivar, H.L. Heyneker, D.G. Yansura, R.
Crea, T. Hirose, A. Kraszewski, K. Itakura, A.D. Riggs, Expression in
Escherichia coli of chemically synthesized genes for human
insulin, Proc. Natl Acad. Sci. USA 76 (1979) 106–110.
[8] R. Vigneri, S. Squatrito, L. Sciacca, Insulin and its analogs: actions
via insulin and IGF receptors, Acta Diabetol. 47 (2010) 271–278.
[9] J.S. Freeman, Insulin analog therapy: improving the match with
physiologic insulin secretion, J. Am. Osteopath. Assoc. 109 (2009)
26–36.
[10] R.J. Solá, K. Griebenow, Glycosylation of therapeutic proteins: an
effective strategy to optimize efficacy, BioDrugs 24 (2010) 9–21.
[11] S. Elliott, T. Lorenzini, S. Asher, K. Aoki, D. Brankow, L. Buck, L.
Busse, D. Chang, J. Fuller, J. Grant, N. Hernday, M. Hokum, S. Hu, A.
Knudten, N. Levin, R. Komorowski, F. Martin, R. Navarro, T.
Osslund, G. Rogers, N. Rogers, G. Trail, J. Egrie, Enhancement of
therapeutic protein in vivo activities through glycoengineering,
Nat. Biotechnol. 21 (2003) 414–421.
[12] Z. Kiss, S. Elliott, K. Jedynasty, V. Tesar, J. Szegedi, Discovery and
basic pharmacology of erythropoiesis-stimulating agents (ESAs),
including the hyperglycosylated ESA, darbepoetin alfa: an update
of the rationale and clinical impact, Eur. J. Clin. Pharmacol. 66
(2010) 331–340.
[13] C. Huang, Receptor-Fc fusion therapeutics, traps, and
MIMETIBODY technology, Curr. Opin. Biotechnol. 20 (2009)
692–699.
[14] J.A. Jazayeri, G.J. Carroll, Fc-based cytokines: prospects for
engineering superior therapeutics, BioDrugs 22 (2008) 11–26.
[15] R.E. Kontermann, Strategies to extend plasma half-lives of
recombinant antibodies, BioDrugs 23 (2009) 93–109.
[16] D.J. Capon, S.M. Chamow, J. Mordenti, S.A. Marsters, T. Gregory, H.
Mitsuya, R.A. Byrn, C. Lucas, F.M. Wurm, J.E. Groopman, S. Broder,
D.H. Smith, Designing CD4 immunoadhesins for AIDS therapy,
Nature 337 (1989) 525–531.
[17] D.B. Cines, U. Yasothan, P. Kirkpatrick, Romiplostim, Nat. Rev.
Drug Discov. 7 (2008) 887–888.
[18] S. Jevševar, M. Kunstelj, V.G. Porekar, PEGylation of therapeutic
proteins, Biotechnol. J. 5 (2010) 113–128.
1268

E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9

[19] F.M. Veronese, A. Mero, The impact of PEGylation on biological
therapies, BioDrugs 22 (2008) 315–329.
[20] G.B. Kresse, Biosimilars — science, status, and strategic
perspective, Eur. J. Pharm. Biopharm. 72 (2009) 479–486.
[21] A.P. Chapman, P. Antoniw, M. Spitali, S. West, S. Stephens, D.J.
King, Therapeutic antibody fragments with prolonged in vivo
half-lives, Nat. Biotechnol. 17 (1999) 780–783.
[22] J. Li, C. Yang, Y. Xia, A. Bertino, J. Glaspy, M. Roberts, D.J. Kuter,
Thrombocytopenia caused by the development of antibodies to
thrombopoietin, Blood 98 (2001) 3241–3248.
[23] A. Bendele, J. Seely, C. Richey, G. Sennello, G. Shopp, Short
communication: renal tubular vacuolation in animals treated
with polyethylene-glycol-conjugated proteins, Toxicol. Sci. 42
(1998) 152–157.
[24] J.C. Almagro, J. Fransson, Humanization of antibodies, Front.
Biosci. 13 (2008) 1619–1633.
[25] N. Lonberg, Fully human antibodies from transgenic mouse and
phage display platforms, Curr. Opin. Immunol. 20 (2008)
450–459.
[26] N. Lonberg, Human monoclonal antibodies from transgenic mice,
Handb. Exp. Pharmacol. (2008) 69–97.
[27] H. Thie, T. Meyer, T. Schirrmann, M. Hust, S. Dübel, Phage display
derived therapeutic antibodies, Curr. Pharm. Biotechnol. 9 (2008)
439–446.
[28] B. Kelley, Industralization of mAb production technology. The
bioprocess industry at a crossroads, mAbs 1 (2009) 443–452.
[29] M. Better, C.P. Chang, R.R. Robinson, A.H. Horwitz, Escherichia coli
secretion of an active chimeric antibody fragment, Science 240
(1988) 1041–1043.
[30] A. Skerra, A. Plückthun, Assembly of a functional immunoglobulin
Fv fragment in Escherichia coli, Science 240 (1988) 1038–1041.
[31] P. Carter, R.F. Kelley, M.L. Rodrigues, B. Snedecor, M. Covarrubias, M.D.
Velligan, W.L. Wong, A.M. Rowland, C.E. Kotts, M.E. Carver, M. Yang,
J.H. Bourell, H.M. Shepard, D. Henner, High level Escherichia coli
expression and production of a bivalent humanized antibody
fragment, Biotechnology 10 (1992) 163–167.
[32] A. Beck, T. Wurch, C. Bailly, N. Corvaia, Strategies and challenges
for the next generation of therapeutic antibodies, Nat. Rev.
Immunol. 10 (2010) 345–352.
[33] P. Chames, M. Van Regenmortel, E. Weiss, D. Baty, Therapeutic
antibodies: successes, limitations and hopes for the future, Br. J.
Pharmacol. 157 (2009) 220–233.
[34] A.C. Chan, P.J. Carter, Therapeutic antibodies for autoimmunity
and inflammation, Nat. Rev. Immunol. 10 (2010) 301–316.
[35] J.M. Reichert, Monoclonal antibodies as innovative therapeutics,
Curr. Pharm. Biotechnol. 9 (2008) 423–430.
[36] A.L. Marschall, A. Frenzel, T. Schirrmann, M. Schüngel, S. Dübel,
Targeting antibodies to the cytoplasm, mAbs 3 (2011) 3–16.
[37] L. Lampson, Monoclonal antibodies in neuro-oncology: getting
past the blood-brain barrier, mAbs 3 (2011) 156–163.
[38] B.V. Zlokovic, The blood-brain barrier in health and chronic
neurodegenerative disorders, Neuron 57 (2008) 178–201.
[39] H.-H. Sedalacek, G. Seemann, D. Hoffmann, J. Czech, P. Lorenz, C.
Kolar, K. Bosslet, Antibodies as Carriers of Cytotoxicity, Karger,
Munich, Germany, 1992.
[40] M.M. Schmidt, K.D. Wittrup, A modeling analysis of the effects of
molecular size and binding affinity on tumor targeting, Mol.
Cancer Ther. 8 (2009) 2861–2871.
[41] M.E. Ackerman, D. Pawlowski, K.D. Wittrup, Effect of antigen
turnover rate and expression level on antibody penetration into
tumor spheroids, Mol. Cancer Ther. 7 (2008) 2233–2240.
[42] H. Hurwitz, L. Fehrenbacher, W. Novotny, T. Cartwright, J.
Hainsworth, W. Heim, J. Berlin, A. Baron, S. Griffing, E. Holmgren,
N. Ferrara, G. Fyfe, B. Rogers, R. Ross, F. Kabbinavar, Bevacizumab
plus irinotecan, fluorouracil, and leucovorin for metastatic
colorectal cancer, N. Engl. J. Med. 350 (2004) 2335–2342.
[43] D.J. Slamon, B. Leyland-Jones, S. Shak, H. Fuchs, V. Paton, A.
Bajamonde, T. Fleming, W. Eiermann, J. Wolter, M. Pegram, J.
Baselga, L. Norton, Use of chemotherapy plus a monoclonal

[44]

[45]

[46]
[47]

[48]
[49]
[50]
[51]
[52]

[53]
[54]

[55]
[56]

[57]

[58]

[59]
[60]

[61]

[62]

[63]

[64]

[65]

antibody against HER2 for metastatic breast cancer that
overexpresses HER2, N. Engl. J. Med. 344 (2001) 783–792.
L.M. Weiner, R. Surana, S. Wang, Monoclonal antibodies: versatile
platforms for cancer immunotherapy, Nat. Rev. Immunol. 10
(2010) 317–327.
D.J. Norman, L. Chatenoud, D. Cohen, M. Goldman, C.F. Shield III,
Consensus statement regarding OKT3-induced cytokine-release
syndrome and human antimouse antibodies, Transplant. Proc. 25
(1993) 89–92.
D.K. Arrell, A. Terzic, Network systems biology for drug discovery,
Clin. Pharmacol. Ther. 88 (2010) 120–125.
A.L. Barabási, N. Gulbahce, J. Loscalzo, Network medicine: a
network-based approach to human disease, Nat. Rev. Genet. 12
(2011) 56–68.
G.B. Kresse, Biosimilars — science, status, and strategic
perspective, Eur. J. Pharm. Biopharm. 72 (2009) 479–486.
P.J. Carter, Potent antibody therapeutics by design, Nat. Rev.
Immunol. 6 (2006) 343–357.
A. Nisonoff, W.J. Mandy, Quantitative estimation of the
hybridization of rabbit antibodies, Nature 194 (1962) 355–359.
R. Linke, A. Klein, D. Seimetz, Catumaxomab: clinical
development and future directions, mAbs 2 (2010) 129–136.
R. Bargou, E. Leo, G. Zugmaier, M. Klinger, M. Goebeler, S.
Knop, R. Noppeney, A. Viardot, G. Hess, M. Schuler, H. Einsele, C.
Brandl, A. Wolf, P. Kirchinger, P. Klappers,
M. Schmidt, G. Riethmüller, C. Reinhardt, P.A. Baeuerle,
P. Kufer, Tumor regression in cancer patients by very low
doses of a T cell-engaging antibody, Science 321 (2008) 974–977.
J.S. Marvin, Z. Zhu, Recombinant approaches to IgG-like bispecific
antibodies, Acta Pharmacol. Sin. 26 (2005) 649–658.
S.C. Alley, N.M. Okeley, P.D. Senter, Antibody-drug conjugates:
targeted drug delivery for cancer, Curr. Opin. Chem. Biol. 14
(2010) 529–537.
P.J. Carter, P.D. Senter, Antibody-drug conjugates for cancer
therapy, Cancer J. 14 (2008) 154–169.
A. Younes, N.L. Bartlett, J.P. Leonard, D.A. Kennedy, C.M. Lynch, E.L.
Sievers, A. Forero-Torres, Brentuximab vedotin (SGN-35) for
relapsed CD30-positive lymphomas, N. Engl. J. Med. 363 (2010)
1812–1821.
I. Niculescu-Duvaz, Trastuzumab emtansine, an antibody-drug
conjugate for the treatment of HER2+ metastatic breast cancer,
Curr. Opin. Mol. Ther. 12 (2010) 350–360.
V. Ghetie, S. Popov, J. Borvak, C. Radu, D. Matesoi, C. Medesan, R.J.
Ober, E.S. Ward, Increasing the serum persistence of an IgG fragment
by random mutagenesis, Nat. Biotechnol. 15 (1997) 637–640.
L.G. Presta, Molecular engineering and design of therapeutic
antibodies, Curr. Opin. Immunol. 20 (2008) 460–470.
J. Zalevsky, A.K. Chamberlain, H.M. Horton, S. Karki, I.W.L. Leung,
T.J. Sproule, G.A. Lazar, D.C. Roopenian, J.R. Desjarlais, Enhanced
antibody half-life improves in vivo activity, Nat. Biotechnol. 28
(2010) 157–159.
T. Igawa, S. Ishii, T. Tachibana, A. Maeda, Y. Higuchi, S. Shimaoka, C.
Moriyama, T. Watanabe, R. Takubo, Y. Doi, T. Wakabayashi, A.
Hayasaka, S. Kadono, T. Miyazaki, K. Haraya, Y. Sekimori, T. Kojima,
Y. Nabuchi, Y. Aso, Y. Kawabe, K. Hattori, Antibody recycling by
engineered pH-dependent antigen binding improves the duration
of antigen neutralization, Nat. Biotechnol. 28 (2010) 1203–1207.
T. Kubota, R. Niwa, M. Satoh, S. Akinaga, K. Shitara, N. Hanai,
Engineered therapeutic antibodies with improved effector
functions, Cancer Sci. 100 (2009) 1566–1572.
G. Cartron, H. Watier, J. Golay, P. Solal-Celigny, From the bench to
the bedside: ways to improve rituximab efficacy, Blood 104
(2004) 2635–2642.
H.K. Binz, P. Amstutz, A. Plückthun, Engineering novel binding
proteins from nonimmunoglobulin domains, Nat. Biotechnol. 23
(2005) 1257–1268.
M. Gebauer, A. Skerra, Engineered protein scaffolds as
next-generation antibody therapeutics, Curr. Opin. Chem. Biol. 13
(2009) 245–255.
E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9

[66] S.D. Nuttall, R.B. Walsh, Display scaffolds: protein engineering
for novel therapeutics, Curr. Opin. Pharmacol. 8 (2008)
609–615.
[67] B. Zuraw, U. Yasothan, P. Kirkpatrick, Ecallantide, Nat. Rev. Drug
Discov. 9 (2010) 189–190.
[68] A. Walker, G. Dunlevy, D. Rycroft, P. Topley, L.J. Holt, T. Herbert, M.
Davies, F. Cook, S. Holmes, L. Jespers, C. Herring, Anti-serum

1269

albumin domain antibodies in the development of highly potent,
efficacious and long-acting interferon, Protein Eng. Des. Sel. 23
(2010) 271–278.
[69] L.J. Holt, A. Basran, K. Jones, J. Chorlton, L.S. Jespers, N.D. Brewis, I.M.
Tomlinson, Anti-serum albumin domain antibodies for extending
the half-lives of short lived drugs, Protein Eng. Des. Sel. 21 (2008)
283–288.

Weitere ähnliche Inhalte

Andere mochten auch

Protein Engeneering
Protein EngeneeringProtein Engeneering
Protein EngeneeringAhreum
 
Protein engineering
Protein engineeringProtein engineering
Protein engineeringSiti Julaiha
 
Protein Engineering
Protein EngineeringProtein Engineering
Protein Engineeringnmicaelo
 
Protein engineering
Protein engineeringProtein engineering
Protein engineeringBen Mair
 
Protein engineering
Protein engineeringProtein engineering
Protein engineeringbansalaman80
 
Protein engineering saurav
Protein engineering sauravProtein engineering saurav
Protein engineering sauravSaurav Das
 

Andere mochten auch (6)

Protein Engeneering
Protein EngeneeringProtein Engeneering
Protein Engeneering
 
Protein engineering
Protein engineeringProtein engineering
Protein engineering
 
Protein Engineering
Protein EngineeringProtein Engineering
Protein Engineering
 
Protein engineering
Protein engineeringProtein engineering
Protein engineering
 
Protein engineering
Protein engineeringProtein engineering
Protein engineering
 
Protein engineering saurav
Protein engineering sauravProtein engineering saurav
Protein engineering saurav
 

Ähnlich wie Introduction to current and future protein therapeutics - a protein engineering perspective

Potential role of bioactive peptides in prevention and treatment of chronic d...
Potential role of bioactive peptides in prevention and treatment of chronic d...Potential role of bioactive peptides in prevention and treatment of chronic d...
Potential role of bioactive peptides in prevention and treatment of chronic d...NxFxProducerDJ
 
Summary of Targeted Protein Degradation in Clinical Trials.pdf
Summary of Targeted Protein Degradation in Clinical Trials.pdfSummary of Targeted Protein Degradation in Clinical Trials.pdf
Summary of Targeted Protein Degradation in Clinical Trials.pdfDoriaFang
 
Recombinant therapeutic proteins market
Recombinant therapeutic proteins marketRecombinant therapeutic proteins market
Recombinant therapeutic proteins marketKuicK Research
 
Pegylation & biosimilars global scenario
Pegylation & biosimilars   global scenarioPegylation & biosimilars   global scenario
Pegylation & biosimilars global scenarioMalay Singh
 
Biosimilar & Patents - Challenges for Research & Analysis
Biosimilar & Patents - Challenges for Research & AnalysisBiosimilar & Patents - Challenges for Research & Analysis
Biosimilar & Patents - Challenges for Research & AnalysisIntellectPeritus Services
 
Download Recombinant therapeutic proteins market & pipeline analysis
Download Recombinant therapeutic proteins market & pipeline analysisDownload Recombinant therapeutic proteins market & pipeline analysis
Download Recombinant therapeutic proteins market & pipeline analysisKuicK Research
 
Protein and peptide delivery system
Protein and peptide delivery systemProtein and peptide delivery system
Protein and peptide delivery systemYamini Shah
 
Cell Engineering and Molecular Pharming in Biopharmaceuticals.pptx
Cell Engineering and Molecular Pharming in Biopharmaceuticals.pptxCell Engineering and Molecular Pharming in Biopharmaceuticals.pptx
Cell Engineering and Molecular Pharming in Biopharmaceuticals.pptxAngela Abraham
 
Microbial production of recombinant pharmaceuticals
Microbial production of recombinant pharmaceuticalsMicrobial production of recombinant pharmaceuticals
Microbial production of recombinant pharmaceuticalsAmjad Afridi
 
CH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdf
CH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdfCH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdf
CH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdfmubarak bilisumaTG
 
PEGS China 2015 Final Agenda
PEGS China 2015 Final AgendaPEGS China 2015 Final Agenda
PEGS China 2015 Final AgendaNicole Proulx
 
Peptides assignment , m.alyami
Peptides assignment , m.alyamiPeptides assignment , m.alyami
Peptides assignment , m.alyamiMohammad Hussain
 
Genetic engineering and pharmaceutical production in microorganisms
Genetic engineering and  pharmaceutical production in microorganismsGenetic engineering and  pharmaceutical production in microorganisms
Genetic engineering and pharmaceutical production in microorganismsNawfal Aldujaily
 
Science As A Business
Science As A BusinessScience As A Business
Science As A BusinessChris Waller
 
9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf
9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf
9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdfDoriaFang
 
Therapeutic proteins
Therapeutic proteinsTherapeutic proteins
Therapeutic proteinsRafa Zubair
 

Ähnlich wie Introduction to current and future protein therapeutics - a protein engineering perspective (20)

Potential role of bioactive peptides in prevention and treatment of chronic d...
Potential role of bioactive peptides in prevention and treatment of chronic d...Potential role of bioactive peptides in prevention and treatment of chronic d...
Potential role of bioactive peptides in prevention and treatment of chronic d...
 
Summary of Targeted Protein Degradation in Clinical Trials.pdf
Summary of Targeted Protein Degradation in Clinical Trials.pdfSummary of Targeted Protein Degradation in Clinical Trials.pdf
Summary of Targeted Protein Degradation in Clinical Trials.pdf
 
Pavithra- ABT therapeuic proteins
Pavithra- ABT therapeuic proteinsPavithra- ABT therapeuic proteins
Pavithra- ABT therapeuic proteins
 
Recombinant therapeutic proteins market
Recombinant therapeutic proteins marketRecombinant therapeutic proteins market
Recombinant therapeutic proteins market
 
Pegylation & biosimilars global scenario
Pegylation & biosimilars   global scenarioPegylation & biosimilars   global scenario
Pegylation & biosimilars global scenario
 
Biosimilar & Patents - Challenges for Research & Analysis
Biosimilar & Patents - Challenges for Research & AnalysisBiosimilar & Patents - Challenges for Research & Analysis
Biosimilar & Patents - Challenges for Research & Analysis
 
Download Recombinant therapeutic proteins market & pipeline analysis
Download Recombinant therapeutic proteins market & pipeline analysisDownload Recombinant therapeutic proteins market & pipeline analysis
Download Recombinant therapeutic proteins market & pipeline analysis
 
Protein and peptide delivery system
Protein and peptide delivery systemProtein and peptide delivery system
Protein and peptide delivery system
 
9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche
9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche
9. Dr. Thomas Schreitmueller - F. Hoffmann-La Roche
 
Cell Engineering and Molecular Pharming in Biopharmaceuticals.pptx
Cell Engineering and Molecular Pharming in Biopharmaceuticals.pptxCell Engineering and Molecular Pharming in Biopharmaceuticals.pptx
Cell Engineering and Molecular Pharming in Biopharmaceuticals.pptx
 
Microbial production of recombinant pharmaceuticals
Microbial production of recombinant pharmaceuticalsMicrobial production of recombinant pharmaceuticals
Microbial production of recombinant pharmaceuticals
 
CH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdf
CH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdfCH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdf
CH£1&2.-PHARM Biotech (Biot 3112).2020 LECT pptx.pdf
 
PEGS China 2015 Final Agenda
PEGS China 2015 Final AgendaPEGS China 2015 Final Agenda
PEGS China 2015 Final Agenda
 
Biopharmaceuticals an overview
Biopharmaceuticals   an overviewBiopharmaceuticals   an overview
Biopharmaceuticals an overview
 
Peptides assignment , m.alyami
Peptides assignment , m.alyamiPeptides assignment , m.alyami
Peptides assignment , m.alyami
 
Genetic engineering and pharmaceutical production in microorganisms
Genetic engineering and  pharmaceutical production in microorganismsGenetic engineering and  pharmaceutical production in microorganisms
Genetic engineering and pharmaceutical production in microorganisms
 
Science As A Business
Science As A BusinessScience As A Business
Science As A Business
 
9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf
9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf
9 Types of Drug Conjugates Overview ADC、RDC、ISAC、SMDC、AOC.pdf
 
Precision medicine.ppt
Precision medicine.pptPrecision medicine.ppt
Precision medicine.ppt
 
Therapeutic proteins
Therapeutic proteinsTherapeutic proteins
Therapeutic proteins
 

Mehr von National Institute of Biologics

Defining your-target-product-profile in-vitro-diagnostic-products
Defining your-target-product-profile in-vitro-diagnostic-productsDefining your-target-product-profile in-vitro-diagnostic-products
Defining your-target-product-profile in-vitro-diagnostic-productsNational Institute of Biologics
 
Accelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapiesAccelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapiesNational Institute of Biologics
 
Canonical structures for the hypervariable regions of immunoglobulins
Canonical structures for the hypervariable regions of immunoglobulinsCanonical structures for the hypervariable regions of immunoglobulins
Canonical structures for the hypervariable regions of immunoglobulinsNational Institute of Biologics
 
Development trends for human monoclonal antibody therapeutics
Development trends for human monoclonal antibody therapeuticsDevelopment trends for human monoclonal antibody therapeutics
Development trends for human monoclonal antibody therapeuticsNational Institute of Biologics
 
Therapeutic fc fusion proteins and peptides as successful alternatives to ant...
Therapeutic fc fusion proteins and peptides as successful alternatives to ant...Therapeutic fc fusion proteins and peptides as successful alternatives to ant...
Therapeutic fc fusion proteins and peptides as successful alternatives to ant...National Institute of Biologics
 
Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...
Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...
Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...National Institute of Biologics
 
Therapeutic antibodies for autoimmunity and inflammation
Therapeutic antibodies for autoimmunity and inflammationTherapeutic antibodies for autoimmunity and inflammation
Therapeutic antibodies for autoimmunity and inflammationNational Institute of Biologics
 
Pharmaceutical monoclonal antibodies production - guidelines to cell engine...
Pharmaceutical monoclonal antibodies   production - guidelines to cell engine...Pharmaceutical monoclonal antibodies   production - guidelines to cell engine...
Pharmaceutical monoclonal antibodies production - guidelines to cell engine...National Institute of Biologics
 
Intended use of reference products & who international standards or reference...
Intended use of reference products & who international standards or reference...Intended use of reference products & who international standards or reference...
Intended use of reference products & who international standards or reference...National Institute of Biologics
 
Evaluation of similar biotherapeutic products (SBP's) scientific principles ...
Evaluation of similar biotherapeutic products (SBP's)   scientific principles ...Evaluation of similar biotherapeutic products (SBP's)   scientific principles ...
Evaluation of similar biotherapeutic products (SBP's) scientific principles ...National Institute of Biologics
 

Mehr von National Institute of Biologics (20)

Waters protein therapeutics application proctocols
Waters protein therapeutics application proctocolsWaters protein therapeutics application proctocols
Waters protein therapeutics application proctocols
 
Potential aggregation prone regions in biotherapeutics
Potential aggregation prone regions in biotherapeuticsPotential aggregation prone regions in biotherapeutics
Potential aggregation prone regions in biotherapeutics
 
How the biologics landscape is evolving
How the biologics landscape is evolvingHow the biologics landscape is evolving
How the biologics landscape is evolving
 
Evaluation of antibody drugs quality safety
Evaluation of antibody drugs quality safetyEvaluation of antibody drugs quality safety
Evaluation of antibody drugs quality safety
 
Approved m abs_feb_2015
Approved m abs_feb_2015Approved m abs_feb_2015
Approved m abs_feb_2015
 
Translating next generation sequencing to practice
Translating next generation sequencing to practiceTranslating next generation sequencing to practice
Translating next generation sequencing to practice
 
From biomarkers to diagnostics –the road to success
From biomarkers to diagnostics –the road to successFrom biomarkers to diagnostics –the road to success
From biomarkers to diagnostics –the road to success
 
Defining your-target-product-profile in-vitro-diagnostic-products
Defining your-target-product-profile in-vitro-diagnostic-productsDefining your-target-product-profile in-vitro-diagnostic-products
Defining your-target-product-profile in-vitro-diagnostic-products
 
Accelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapiesAccelerating development and approval of targeted cancer therapies
Accelerating development and approval of targeted cancer therapies
 
Canonical structures for the hypervariable regions of immunoglobulins
Canonical structures for the hypervariable regions of immunoglobulinsCanonical structures for the hypervariable regions of immunoglobulins
Canonical structures for the hypervariable regions of immunoglobulins
 
Canonical correlation
Canonical correlationCanonical correlation
Canonical correlation
 
Development trends for human monoclonal antibody therapeutics
Development trends for human monoclonal antibody therapeuticsDevelopment trends for human monoclonal antibody therapeutics
Development trends for human monoclonal antibody therapeutics
 
Therapeutic fc fusion proteins and peptides as successful alternatives to ant...
Therapeutic fc fusion proteins and peptides as successful alternatives to ant...Therapeutic fc fusion proteins and peptides as successful alternatives to ant...
Therapeutic fc fusion proteins and peptides as successful alternatives to ant...
 
Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...
Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...
Fc fusion proteins and fc rn - structural insights for longer-lasting and mor...
 
Therapeutic antibodies for autoimmunity and inflammation
Therapeutic antibodies for autoimmunity and inflammationTherapeutic antibodies for autoimmunity and inflammation
Therapeutic antibodies for autoimmunity and inflammation
 
Pharmaceutical monoclonal antibodies production - guidelines to cell engine...
Pharmaceutical monoclonal antibodies   production - guidelines to cell engine...Pharmaceutical monoclonal antibodies   production - guidelines to cell engine...
Pharmaceutical monoclonal antibodies production - guidelines to cell engine...
 
Intended use of reference products & who international standards or reference...
Intended use of reference products & who international standards or reference...Intended use of reference products & who international standards or reference...
Intended use of reference products & who international standards or reference...
 
How dissimilarly similar are biosimilars
How dissimilarly similar are biosimilarsHow dissimilarly similar are biosimilars
How dissimilarly similar are biosimilars
 
Evaluation of similar biotherapeutic products (SBP's) scientific principles ...
Evaluation of similar biotherapeutic products (SBP's)   scientific principles ...Evaluation of similar biotherapeutic products (SBP's)   scientific principles ...
Evaluation of similar biotherapeutic products (SBP's) scientific principles ...
 
Biosimilars - global scenario and challenges
Biosimilars  - global scenario and challengesBiosimilars  - global scenario and challenges
Biosimilars - global scenario and challenges
 

Kürzlich hochgeladen

Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...
Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...
Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...narwatsonia7
 
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr
9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr
9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi NcrDelhi Call Girls
 
Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...
Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...
Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...Nehru place Escorts
 
Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...
Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...
Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...rajnisinghkjn
 
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...narwatsonia7
 
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service MumbaiLow Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbaisonalikaur4
 
Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...
Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...
Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...narwatsonia7
 
Radiation Dosimetry Parameters and Isodose Curves.pptx
Radiation Dosimetry Parameters and Isodose Curves.pptxRadiation Dosimetry Parameters and Isodose Curves.pptx
Radiation Dosimetry Parameters and Isodose Curves.pptxDr. Dheeraj Kumar
 
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaCall Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaPooja Gupta
 
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...narwatsonia7
 
epilepsy and status epilepticus for undergraduate.pptx
epilepsy and status epilepticus  for undergraduate.pptxepilepsy and status epilepticus  for undergraduate.pptx
epilepsy and status epilepticus for undergraduate.pptxMohamed Rizk Khodair
 
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking ModelsMumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Modelssonalikaur4
 
Pharmaceutical Marketting: Unit-5, Pricing
Pharmaceutical Marketting: Unit-5, PricingPharmaceutical Marketting: Unit-5, Pricing
Pharmaceutical Marketting: Unit-5, PricingArunagarwal328757
 
Report Back from SGO: What’s New in Uterine Cancer?.pptx
Report Back from SGO: What’s New in Uterine Cancer?.pptxReport Back from SGO: What’s New in Uterine Cancer?.pptx
Report Back from SGO: What’s New in Uterine Cancer?.pptxbkling
 
Case Report Peripartum Cardiomyopathy.pptx
Case Report Peripartum Cardiomyopathy.pptxCase Report Peripartum Cardiomyopathy.pptx
Case Report Peripartum Cardiomyopathy.pptxNiranjan Chavan
 
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Availablenarwatsonia7
 
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️saminamagar
 
call girls in Connaught Place DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...
call girls in Connaught Place  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...call girls in Connaught Place  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...
call girls in Connaught Place DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...saminamagar
 

Kürzlich hochgeladen (20)

Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...
Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...
Housewife Call Girls Hsr Layout - Call 7001305949 Rs-3500 with A/C Room Cash ...
 
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Hsr Layout Just Call 7001305949 Top Class Call Girl Service Available
 
9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr
9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr
9873777170 Full Enjoy @24/7 Call Girls In North Avenue Delhi Ncr
 
Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...
Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...
Call Girls Service in Virugambakkam - 7001305949 | 24x7 Service Available Nea...
 
Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...
Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...
Dwarka Sector 6 Call Girls ( 9873940964 ) Book Hot And Sexy Girls In A Few Cl...
 
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
Housewife Call Girls Bangalore - Call 7001305949 Rs-3500 with A/C Room Cash o...
 
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service MumbaiLow Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
Low Rate Call Girls Mumbai Suman 9910780858 Independent Escort Service Mumbai
 
Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...
Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...
Russian Call Girls Gunjur Mugalur Road : 7001305949 High Profile Model Escort...
 
Radiation Dosimetry Parameters and Isodose Curves.pptx
Radiation Dosimetry Parameters and Isodose Curves.pptxRadiation Dosimetry Parameters and Isodose Curves.pptx
Radiation Dosimetry Parameters and Isodose Curves.pptx
 
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
Call Girls Jayanagar Just Call 7001305949 Top Class Call Girl Service Available
 
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service NoidaCall Girls Service Noida Maya 9711199012 Independent Escort Service Noida
Call Girls Service Noida Maya 9711199012 Independent Escort Service Noida
 
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
Call Girls Electronic City Just Call 7001305949 Top Class Call Girl Service A...
 
epilepsy and status epilepticus for undergraduate.pptx
epilepsy and status epilepticus  for undergraduate.pptxepilepsy and status epilepticus  for undergraduate.pptx
epilepsy and status epilepticus for undergraduate.pptx
 
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking ModelsMumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
Mumbai Call Girls Service 9910780858 Real Russian Girls Looking Models
 
Pharmaceutical Marketting: Unit-5, Pricing
Pharmaceutical Marketting: Unit-5, PricingPharmaceutical Marketting: Unit-5, Pricing
Pharmaceutical Marketting: Unit-5, Pricing
 
Report Back from SGO: What’s New in Uterine Cancer?.pptx
Report Back from SGO: What’s New in Uterine Cancer?.pptxReport Back from SGO: What’s New in Uterine Cancer?.pptx
Report Back from SGO: What’s New in Uterine Cancer?.pptx
 
Case Report Peripartum Cardiomyopathy.pptx
Case Report Peripartum Cardiomyopathy.pptxCase Report Peripartum Cardiomyopathy.pptx
Case Report Peripartum Cardiomyopathy.pptx
 
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service AvailableCall Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
Call Girls ITPL Just Call 7001305949 Top Class Call Girl Service Available
 
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️call girls in munirka  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
call girls in munirka DELHI 🔝 >༒9540349809 🔝 genuine Escort Service 🔝✔️✔️
 
call girls in Connaught Place DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...
call girls in Connaught Place  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...call girls in Connaught Place  DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...
call girls in Connaught Place DELHI 🔝 >༒9540349809 🔝 genuine Escort Service ...
 

Introduction to current and future protein therapeutics - a protein engineering perspective

  • 1. E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9 available at www.sciencedirect.com www.elsevier.com/locate/yexcr Review Introduction to current and future protein therapeutics: A protein engineering perspective Paul J. Carter⁎ Department of Antibody Engineering, Genentech, Inc., 1 DNA Way, South San Francisco, CA 94080-4990, USA A R T I C L E I N F O R M A T I O N A B S T R A C T Article Chronology: Protein therapeutics and its enabling sister discipline, protein engineering, have emerged since the Received 18 January 2011 early 1980s. The first protein therapeutics were recombinant versions of natural proteins. Proteins Revised version received purposefully modified to increase their clinical potential soon followed with enhancements 20 February 2011 derived from protein or glycoengineering, Fc fusion or conjugation to polyethylene glycol. Accepted 24 February 2011 Antibody-based drugs subsequently arose as the largest and fastest growing class of protein Available online 1 March 2011 therapeutics. The rationale for developing better protein therapeutics with enhanced efficacy, greater safety, reduced immunogenicity or improved delivery comes from the convergence of Keywords: clinical, scientific, technological and commercial drivers that have identified unmet needs and Protein therapeutics provided strategies to address them. Future protein drugs seem likely to be more extensively Antibody therapeutics engineered to improve their performance, e.g., antibodies and Fc fusion proteins with enhanced Fc fusion proteins effector functions or extended half-life. Two old concepts for improving antibodies, namely Engineered protein scaffolds antibody-drug conjugates and bispecific antibodies, have advanced to the cusp of clinical success. As for newer protein therapeutic platform technologies, several engineered protein scaffolds are in early clinical development and offer differences and some potential advantages over antibodies. © 2011 Elsevier Inc. All rights reserved. Contents Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Brief history of protein therapeutics . . . . . . . . . . . . . . . . . . . Protein therapeutics enabled by protein engineering . . . . . . . . . Insulin — the first recombinant human protein therapeutic . . . . . Beginning of engineered proteins as therapeutics . . . . . . . . . . . Fc fusion proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . Protein conjugation to polyethylene glycol . . . . . . . . . . . . . . The rise of antibody therapeutics . . . . . . . . . . . . . . . . . . . Rationale for next generation protein therapeutics . . . . . . . . . . . . Strengths and limitations of antibody and other protein therapeutics Clinical rationale . . . . . . . . . . . . . . . . . . . . . . . . . . . Scientific rationale . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ⁎ Fax: +1 650 467 8318. E-mail address: pjc@gene.com. Abbreviations: FDA, Food and Drug Administration; PEG, polyethylene glycol; TPO, thrombopoietin 0014-4827/$ – see front matter © 2011 Elsevier Inc. All rights reserved. doi:10.1016/j.yexcr.2011.02.013 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1262 1262 1262 1262 1263 1263 1263 1263 1264 1264 1264 1264
  • 2. 1262 E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9 Commercial rationale . . . . . . . . . . . . . . . . . . . . Platform technologies for next generation protein therapeutics Next generation antibodies and Fc fusion proteins . . . . . Engineered protein scaffolds . . . . . . . . . . . . . . . . Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . Introduction Since the early 1980s proteins have emerged as a major new class of pharmaceuticals with ~200 marketed products that are mainly therapeutics with a small number of diagnostics and vaccines [1]. Protein therapeutics can be classified based upon their pharmacologic activity as drugs that: i) replace a protein that is deficient or abnormal, ii) augment an existing pathway, iii) provide a novel function or activity, iv) interfere with a molecule or organism, or iv) deliver a payload such as a radionuclide, cytotoxic drug, or protein effector [2]. Alternatively, protein therapeutics can be grouped into molecular types that include: antibody-based drugs, anticoagulants, blood factors, bone morphogenetic proteins, engineered protein scaffolds, enzymes, Fc fusion proteins, growth factors, hormones, interferons, interleukins, and thrombolytics (Fig. 1) [1,3]. Antibody-based drugs are the largest and fastest growing class of protein therapeutics with 24 marketed antibody drugs in the USA [4] and over 240 more in clinical development [5]. This introductory article provides a brief history of protein therapeutics, offers a rationale for developing better protein drugs, and identifies some major future opportunities as well as emerging technologies that may meet them. The 30-year history of protein drugs is illustrated here with examples that pioneered their class or have high clinical or commercial significance. The rationale for creating better protein drugs, including antibodies [6], comes from the convergence of clinical, scientific, technological and commercial drivers that collectively have identified major unmet needs and provide strategies to address them. Major future opportunities for protein therapeutics, are improved efficacy, greater safety, reduced immunogenicity or improved delivery. A plethora of emerging technologies for the generation and optimization of protein therapeutics are anticipated to help address these opportunities. Space Recombinant proteins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1265 1265 1265 1266 1267 1267 Brief history of protein therapeutics Protein therapeutics enabled by protein engineering Protein therapeutics and its enabling sister discipline, protein engineering, have emerged since the early 1980s. Protein engineering by rational design or molecular evolution allows the systemic dissection of protein structure–function relationships and the generation of novel proteins with modified activities or entirely new properties. Indeed, protein engineering has revolutionized protein therapeutics by providing the tools to customize existing proteins or to create novel proteins for specific clinical applications. A brief history of protein therapeutics is provided here from a protein engineering perspective. The focus is on technologies with the greatest impact on current protein therapeutics — engineered versions of natural proteins, Fc fusion proteins, conjugation to the polymer, polyethylene glycol (PEG), and antibody therapeutics as the quintessential protein therapeutic platform. Insulin — the first recombinant human protein therapeutic The first human protein therapeutic derived from recombinant DNA technology was human insulin (Humulin®) created at Genentech [7], developed by Eli Lilly, and approved by the US Food and Drug Administration (FDA) in 1982. Recombinant insulin Antibodies Fab fragment Conjugate PEG Engineer protein Engineer glycosylation . . . . . . constraints limit examples here to a sampling of technologies of broad applicability — so called platform technologies. The interested reader is referred to many excellent articles on protein therapeutics including those in this special issue and others cited herein. Anticoagulants Blood factors Bone morphogenetic proteins Enzymes Growth factors Hormones Interferons Interleukins Thrombolytics Optional modifications . . . . . . Fc fusion proteins IgG Receptor Ligand Peptide Fc Conjugate PEG Bispecific Conjugate drug Conjugate radionuclide Fig. 1 – Overview of currently marketed protein therapeutics from a protein engineering perspective.
  • 3. E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9 was used to replace its natural counterpart that can be deficient in diabetes mellitus. Other recombinant human proteins were soon developed as therapeutics to replace the natural proteins deficient in some patients (e.g., growth hormone) or augment existing pathways (e.g., interferon-α, tissue plasminogen activator, and erythropoietin) [2]. Beginning of engineered proteins as therapeutics Beyond recombinant versions of natural proteins, the next major step in the evolution of protein therapeutics was the redesign of proteins to increase their clinical potential. Injected insulin fails to adequately mimic endogenous insulin in two major ways: rapid onset of action after meals and prolonged maintenance of low level insulin between meals [8]. Insulin has been engineered, commonly by one to three amino acid replacements, to create analogs that are either rapid-acting or long-acting and better mimic different properties of endogenous insulin [8]. Multiple insulin analogs have been approved for the treatment of diabetes mellitus including the rapid-acting analogs, insulin lispro (Humalog®), insulin aspart (NovoRapid® and Novolog®) and insulin glulisine (Apidra®) and the long-acting analogs, insulin glargine (Optisulin® and Lantus®) and insulin detemir (Levemir®) [1,8]. Insulin analogs offer multiple benefits over first generation recombinant insulin drugs that include improved physiologic profile, greater convenience for patients, reduced risk of hypoglycemia, and less weight gain in some cases [9]. The commercial importance of engineered insulins is illustrated by Lantus®, which was one of the top ten selling biopharmaceuticals in 2009 [1]. Many protein therapeutics are produced in mammalian cells and have one or more N- and/or O-linked glycans [10]. Engineering a protein with additional glycosylation sites can increase the in vivo serum half-life and thereby target exposure [11]. An early example of such “glycoengineering” of proteins for half-life extension is provided by darbepoetin-α (Aranesp®), administered to anemic patients to stimulate the production of red blood cells in the bone marrow. The first generation drug, epoetin-α, has 3 Nlinked glycosylation sites and was engineered with 2 additional glycosylation sites to create darbepoetin-α, a commercially successful longer-acting second generation drug [12]. Protein glycosylation can sometimes reduce bioactivity, albeit offset by longer half-life. Darbepoetin-α has higher specific activity in patients than epoetin-α, reflecting that the 3-fold increase in serum half-life that increases drug exposure more than offsets a 5fold reduction in receptor binding affinity [12]. 1263 Beyond half-life extension, Fc fusion can provide several additional benefits such as facilitating expression and secretion of recombinant protein, enabling facile purification by protein A chromatography, improving solubility and stability, and enhancing potency by increasing valency. Optional additional properties conferred by Fc fusion include binding to Fcγ receptors and/or complement to support secondary immune functions [13,14]. The first demonstration of the Fc fusion protein concept was CD4-Fc [16]. Five Fc fusion protein therapeutics were approved by 2010, the prototypical and most successful drug of this class being etanercept (Enbrel®), a TNF receptor 2-Fc fusion protein [1]. Etanercept was the top-selling protein therapeutic in 2009 with $6.6 billion in world-wide sales in rheumatoid arthritis and other autoimmune diseases [1]. Romiplostim (Nplate®) is the first peptide-Fc fusion (“peptibody”) approved as a human therapeutic. Romiplostim is a thrombopoietin (TPO) receptor agonist approved as a treatment for thrombocytopenia [17]. Protein conjugation to polyethylene glycol Protein conjugation to chemicals such as radionuclide chelators, cytotoxic drugs and PEG can endow proteins with brand new capabilities to increase their clinical potential. The most clinically and commercially successful protein conjugates thus far have been with PEG. Protein conjugation to PEG – “PEGylation” – increases their hydrodynamic volume, prevents rapid renal clearance, and thereby increases the serum half-life, as extensively reviewed [15,18,19]. At least 8 PEGylated proteins are approved as human therapeutics [18]. Indeed, longacting second generation interferon-α molecules that are PEGylated have largely superseded their first generation non-PEGylated counterparts [20]. Benefits of PEGylation are that it is possible to tune the serum-half of proteins by varying the number of attached PEG molecules, their size and extent of branching [21]. PEGylation can also reduce the immunogenicity of proteins [18,19], although PEGylated proteins can still sometimes be immunogenic [22]. A draw back to PEGylation is that it can impair protein function, a limitation that is offset by the greatly increased half-life, and potentially ameliorated by site-specific PEGylation and/or tailoring the PEG [18,19]. PEG is a non-natural and apparently nonbiodegradable polymer, a molecular attribute that has unclear clinical significance. PEGylated proteins can induce renal tubular vacuolation in animals, albeit with unaltered clinical pathology [23] and unknown impact on long term safety. The rise of antibody therapeutics Fc fusion proteins Another significant advance in protein therapeutics came with the advent of fusion proteins by joining the genes encoding 2 or more different proteins. Fusion proteins combine properties of their component parts. The most clinically and commercially successful fusion protein therapeutics to date contain the Fc region of immunoglobulins [1]. Such Fc fusions can endow peptides or proteins with the IgG-like property of long serum half-life (days to weeks), by virtue of binding to the salvage receptor, FcRn [13,14]. By contrast, proteins of ~70 kDa and smaller are typically eliminated rapidly from circulation by renal filtration and have half-lives of a few minutes to a few hours, that can in many cases render them marginal or unsuitable for therapeutic applications [15]. Antibody-based drugs are the largest and fastest growing class of protein therapeutics with 24 marketed antibody therapeutics in the USA (28 FDA-approved and 4 later withdrawn from the market) [4] and at least 240 more in clinical development [5]. Antibody-based drugs contributed $38 billion of $99 billion in world-wide sales for protein biopharmaceuticals in 2009 [1]. Moreover, 5 of the 10 top-selling protein therapeutics in 2009 were antibodies, namely, infliximab (Remicade®), bevacizumab (Avastin®), rituximab (Rituxan® and MabThera®), adalimumab (Humira®) and trastuzumab (Herceptin®) [1]. The rapid growth of antibody therapeutics since the mid-1990s was made possible by the advent of antibody chimerization and humanization [24] technologies that largely overcame the
  • 4. 1264 E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9 limitations of rodent monoclonal antibodies as drugs, as well as the emergence of facile routes to human antibodies using transgenic mice [25,26] or phage display [25,27] technologies. Other advances that have contributed to the success of antibodies as therapeutics include the industrialization of antibody production technologies [28] that can provide sufficient quantities of antibodies to allow higher doses to be used in patients. The development of methods to express and secrete functional antibody fragments from E. coli [29,30] has significantly stimulated the field of antibody engineering and enabled subsequent technologies such as phage display. The next major advance in the expression of antibody fragments in E. coli was high titer (gram per liter) production in a fermentor [31], that facilitated the development of antibody fragment drugs such as the anti-VEGF Fab, ranibizumab (Lucentis®). The emergence of antibody as therapeutics likely also reflects improved choices of antibody and target antigen for intervention in the pathobiology of disease. The majority of approved antibody drugs are unmodified (“naked”) IgG molecules. However, other antibody formats have been approved for therapy including IgG “armed” with radionuclides or cytotoxic drugs (antibody-drug conjugates) for cancer treatment, as well as Fab fragments and a PEGylated Fab′ fragment [1]. Rationale for next generation protein therapeutics A major quest with protein therapeutics, especially antibodies [6,32], is the development of even better next generation drugs with enhanced efficacy, greater safety or improved delivery. Current antibody therapeutics reveal major strengths that provide a foundation upon which to build, as well as significant limitations to overcome that offers new opportunities [6,33]. Some antibody strengths and many of their limitations apply more broadly to protein therapeutics and are discussed below as a compass to guide the development of next generation protein therapeutics. The impetus to develop better protein therapeutics is also discussed here as a convergence of clinical, scientific, and commercial forces that identify unmeet needs (this section), in conjunction with technological tools that many help address them (next section). Strengths and limitations of antibody and other protein therapeutics One of the greatest strength of antibodies as a therapeutic platform is the potential ease of generating high affinity and specificity human antibodies to virtually any target of therapeutic interest, e.g., using transgenic mice [25,26] or phage display [25,27] technologies. Moreover, antibodies are readily tailored for therapeutic applications by modulating (enhancing or attenuating) their existing properties or endowing them with new activities as discussed in a later section and reviewed more extensively elsewhere [33,34]. As for other protein therapeutics, antibodies are commonly well tolerated by patients, but not invariably so. Antibody drug development benefits from recombinant production methods that are well-established, broadly applicable and commonly support expression at high titer (multi-gram per liter) [28]. Widely available knowledge and experience across all facets of antibody drug development also facilitates the generation of future antibody therapeutics. An advantage of antibodies over other drug formats is that the success rate has been somewhat higher — 17% for humanized antibodies from the first clinical trial to regulatory approval [35]. Proteins have several significant limitations as therapeutics. For example, protein therapeutics are very expensive, reflecting high production costs, that may limit patient access and also clinical applications [28]. This high cost issue is exacerbated for protein therapeutics where multi-gram doses are needed for a treatment course, as is the case for some antibodies. Intracellular delivery of proteins is possible in the laboratory setting, e.g., by protein transduction [36]. However, targeted pharmaceutic delivery of proteins to reach intracellular targets is in its infancy, so protein therapeutics are currently limited to cell surface or extracellular targets. Another shortcoming of proteins therapeutics is that they are denatured and/or proteolyzed in the gut and are thus not orally bioavailable. Proteins, including antibodies [37], show very poor ability to penetrate the specialized endothelial structure of the neurovasculature known as the blood-brain barrier. The integrity of the blood-brain barrier can be partially compromised in some pathological settings such as chronic neurodegenerative diseases [38]. Nevertheless, proteins with current technologies are poorly suited to the treatment of chronic neurodegenerative diseases or tumors that originate or have metastasized to the brain. Antibodies are relatively large (IgG ~150 kDa) compared to other proteins and this large size may limit their tissue penetration and ability to localize to their targets. For example, antibody localization to tumors in patients is typically very inefficient: ≤0.01% of the injected dose per gram of tumor [39]. Comparison of mathematical modeling with experimental data, strongly suggests that molecular size and binding affinity are major determinants of tumor targeting [40]. Many other factors likely contribute to tumor penetration including antigen expression level and turnover rate [41]. Clinical rationale From a clinical standpoint, first generation protein drugs can provide substantial patient benefit. However, in many cases there is room for improvement in efficacy and/or safety. For example, several anti-cancer antibodies provide an overall survival benefit, including bevacizumab [42] and trastuzumab [43] in combination with chemotherapy in metastatic colorectal and breast cancers, respectively. Nevertheless, there remains plenty of opportunity for enhancing antibodies for cancer treatment, such as elevating the overall survival rate of patients, as well as many strategies being explored to do so [44]. All therapeutic proteins are potentially immunogenic in patients and occasionally an anti-drug antibody response can lead to a major safety issue. For example, a truncated and PEGylated form of TPO (MGDF-PEG) elicited an antibody response in some patients that neutralized endogenous TPO leading to severe and persistent anemia, i.e., the opposite of the desired effect of increasing platelet counts [22]. This issue was overcome with the peptibody, Romiplostim (see Fc fusion proteins section) [17]. Scientific rationale Scientifically, our expanding knowledge suggests drug mechanisms of action to enhance, and mechanisms of resistance or toxicity to
  • 5. E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9 overcome. For example, anti-CD3 antibodies illustrate the benefits of understanding and overcoming mechanisms of drug toxicity and resistance. In 1986 the anti-CD3 antibody, muromonab-CD3 (OKT3®), became the first antibody approved by the FDA for human therapy — for the treatment of acute allograft rejection in renal transplant patients. In 1993 muromonab-CD3 was approved for the treatment of acute rejection of heart and liver transplants in patients resistant to steroid treatment. Unfortunately, muromonabCD3 activates T cells resulting in transient cytokine release and acute and severe flu-like symptoms [45]. Another major limitation of muromonab-CD3 is that its therapeutic benefit is reduced by neutralizing antibodies that develop in ~50% of patients [45]. Muromonab-CD3 has since been voluntarily withdrawn from the US market due to decreased sales. These major limitations of muromonab-CD3 have been greatly reduced, if not overcome, in some second generation anti-CD3 antibodies that have advanced to late stage clinical development [6]. For example, the anti-CD3 antibodies, teplizumab and otelixizumab, combine Fc engineering to attenuate Fcγ receptor binding with chimerization or humanization to reduce immunogenicity [34]. Drug development, including for protein therapeutics, seems likely to gain from advances in the understanding of targets and their role in the pathobiology of disease. For example, there is a growing appreciation that targets are not isolated gene products but integral components of networks of interconnected signaling pathways [46,47]. Commercial rationale Clinical and commercial success with protein therapeutics has bred intense competition between different organizations striving to develop protein therapeutics to the same antigen and/or overlapping therapeutic indications. Indeed, this is already reflected in marketed drugs as illustrated by 5 different protein therapeutics that block TNF including 3 different IgG (infliximab, adalimumab and golimumab), a PEGylated Fab′ fragment (certolizumab pegol) and a receptor-Fc fusion (etanercept) [6]. Such competition is increasingly commonplace as patents dominating individual targets expire. The generation of high affinity and potency IgG is often possible, arguably commoditized. This provides a strong commercial incentive to develop superiorperforming antibody drugs to differentiate products from those of competitors. The success of innovator protein therapeutics has brought a rise in biosimilar efforts (i.e., products that are deemed highly similar to innovator protein therapeutics according to drug regulatory authorities). Indeed, several biosimilar approval applications have been filed in the European Union with 13 biosimilars being approved by European drug regulators by early 2009 [48]. For creators of innovator drugs, the impetus to develop improved next generation protein therapeutics that out perform the previous generation drugs has increased substantially [48]. Platform technologies for next generation protein therapeutics Many protein engineering tools have been developed that allow one to optimize favorable properties of proteins, attenuate undesired attributes and create proteins with entirely novel activities. Space constraints limit the discussion here to next 1265 generation antibody and Fc fusion protein therapeutics and the emerging technologies of engineered protein scaffolds. Next generation antibodies and Fc fusion proteins Many promising strategies are emerging to enhance the existing properties of antibodies or to endow them with new activities, as reviewed in detail elsewhere [6,32,33]. Antibodies (IgG) have multiple binding partners including antigen, Fcγ receptors, complement and FcRn. Each of these interactions can be tailored in ways that may improve the clinical properties of antibodies [32,34,49]. Platform technologies that seem likely to impact the next generation of antibody therapeutics include old concepts that are finally coming to fruition, such as antibody-drug conjugates and bispecific antibodies. Technologies for improving Fc-mediated functions, such as half-life extension and effector function enhancement, seem likely to benefit the next generation of antibody therapeutics. Additionally, these technologies appear directly applicable to the optimization of Fc fusion proteins. The 50-year old concept of bispecific antibodies [50] came of age in 2009 with the first regulatory approval of a bispecific antibody — catumaxomab (Removab®, anti-EpCAM x anti-CD3) in Europe. Catumaxomab is approved for the treatment of malignant ascites in patients with EpCAM-positive carcinomas [1,51]. Recruitment of T cells via the anti-CD3 arm of catumaxomab and other immune effector cells via its Fc region may contribute to antitumor activity. A tandem bispecific scFv format known as a “BiTE”, that binds CD3 and a tumor-associated antigen, is a particularly effective way to retarget T cells to kill tumor cells. Blinatumomab, a BiTE specific for CD19 and CD3, gave several complete responses in a phase I clinical trial in non-Hodgkin's lymphoma [52] and is currently in multiple phase II clinical trials. Another major area of current interest with bispecific antibodies is in the simultaneous blockade of 2 disease mediators, which may lead to greater therapeutic benefit than targeting individual disease mediators [53]. Until recently, a major bottleneck in the development of bispecific antibodies as therapeutics was the challenge of producing these complex molecules in sufficient quantity and quality for clinical applications. This issue has been largely overcome by the advent of many alternative formats and production methods for bispecific antibodies [6]. Antibody-drug conjugates, a concept that dates back to the 1970s, use antibodies to deliver a cytotoxic payload to kill a target such as tumor cells [54,55]. One antibody-drug conjugate, gemtuzumab ozogamicin (Mylotarg®), was approved by the FDA for the treatment of acute myeloid leukemia. Gemtuzumab ozogamicin was withdrawn from the market in 2010 due to safety concerns and insufficient patient benefit in post approval clinical trials. Much progress has been made over the decades in many different facets of antibody-drug conjugates, including better choices of target antigen and antibody, and the development of more potent drugs and linkers of greater stability [54,55]. Pivotal clinical trials are in progress for brentuximab vedotin (SGN-35, [56]) in Hodgkin's disease and trastuzumab emtansine (T-DM1, [57]) in metastatic breast cancer over-expressing HER2/neu following demonstration of robust antitumor activity and acceptable safety profiles in earlier clinical trials. Engineering proteins to increase their serum half-life has the potential benefits of increased exposure leading to greater target localization and greater efficacy, lower or less frequent dosing and
  • 6. 1266 E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9 lower cost. In the case of antibodies, the serum half-life in mice can be extended by at least a few fold by engineering the Fc region for higher affinity binding to the salvage receptor (FcRn), whilst preserving the pH dependence of binding [58]. Half-life extension of Fc-engineered antibodies in non-human primates was subsequently demonstrated by several different groups [59]. Antibody half-life extension can result in increased target exposure and improved preclinical in vivo efficacy as recently shown for antiEGFR and anti-VEGF antibodies [60]. Engineering antibodies for pH-dependent interaction with their target antigen has also been shown to extend antibody half-life in vivo [61]. The first half-life extended antibody to enter clinical trials was the humanized antiRSV antibody, MEDI-557, although pharmacokinetic data have yet to be reported. Another major area of interest for improving antibody performance is effector function enhancement, as reviewed extensively elsewhere [34,62] and discussed briefly below. Antibody effector functions include ADCC (antibody-dependent cellular cytotoxicity), ADCP (antibody-dependent cellular phagocytosis) and complement-dependent cytotoxicity (CDC). Effector functions may be advantageous where destruction of the target cells is desired, as is often the case for oncology targets. Indeed many antibodies approved for oncologic indications (e.g., rituximab, trastuzumab, alemtuzumab, cetuximab and ofatumumab) support one or many effector functions that may contribute to their clinical antitumor activity, with the strongest evidence being for rituximab [63]. For antibody and Fc fusion protein therapeutics potential benefits from enhancing effector functions include greater efficacy, lower or less frequent dosing and decreased cost-of-goods. As for risks, these include more frequent or severe adverse events. ADCC, ADCP and CDC have been augmented by Fc protein engineering as extensively reviewed elsewhere [62]. ADCC has also been augmented to a similar level by modifying the glycan attached to the Fc, e.g., afucosylation achieved through cell line engineering [62]. At least 8 effector function enhanced antibodies have advanced into early clinical development [34]. Engineered protein scaffolds The success of antibodies as protein therapeutics has inspired the development of so-called “engineered protein scaffolds” as alternative platform technologies for the development of novel next generation protein therapeutics [64–66]. Engineered protein scaffolds provide binding sites that can be tailored to specifically recognize desired target molecules, in an analogous way that antibodies can be developed to recognize their target antigens. Here the basic features of engineered protein scaffolds are described, together with their strengths, limitations and some possible future directions. Protein scaffolds chosen for engineering have known 3dimensional structures that together represent diverse protein folds [64–66]. Protein scaffolds are commonly selected with the following attributes: small, monomeric, high solubility and stability, and that lack disulfide bonds and glycosylation sites. Protein scaffolds are typically readily expressed in microbial hosts. Sequence diversity is introduced into the protein scaffold, e.g., by randomizing surface accessible residues, to create a so-called library. Binders to a target of therapeutic interest are then identified by selection using technologies such as phage, yeast or ribosome display. Increased binding affinity – affinity maturation – is often needed and readily accomplished by further library building and selection. Over 50 different engineered protein scaffolds have been described with a few advancing into clinical development [64– 66] including: Adnectins® from type III fibronectin domains, Affibodies® from the synthetic Z domain of staphylococcal protein A, Anticalins® from lipocalins, Avimers® from LDLR-A modules, DARPins® from ankyrin repeat proteins, and engineered Kunitz domains. Engineered protein scaffolds also include single-domain antibodies from human (dAbs®) and camelids (Nanobodies®). One engineered protein scaffold protein has been approved for therapy, namely, ecallantide (Kalbitor®), a potent and selective inhibitor of plasma kallikrein based upon a Kunitz domain [67]. Many potential advantages of engineered protein scaffolds over antibodies have been proposed [64–66], albeit with limited supporting evidence to date in the context of developing protein therapeutics. For example, the smaller size and higher stability of some engineered protein scaffolds may allow for alternative routes of administration such as pulmonary delivery. The smaller size of engineered protein scaffolds over antibodies favors more efficient tissue penetration, but this can be more than offset by much faster clearance. Engineered protein scaffolds may be more readily expressed in microbial hosts reflecting their high stability and solubility, small size, simple structure (single polypeptide), and that they commonly lack disulfide bonds and/or glycosylation sites. The small and modular architecture of engineered protein scaffolds seems particularly well suited to the construction of multivalent and/or multispecific protein therapeutics (Fig. 2). Engineered proteins are commonly associated with independent intellectual property that is outside the scope of antibody patents. Several of these proposed advantages of engineered protein scaffolds have been achieved with antibodies by engineering for improved stability or expression, or by exploiting the modular architecture of antibodies to create different formats including ones that are smaller and simpler than IgG [6,32,33]. Engineered protein scaffolds lack Fc-mediated properties of antibodies such as long serum half-life and effector functions. This does not appear to be a major limitation, as many different strategies have been developed to extend the half-life of proteins as comprehensively reviewed [15]. For example, single-domain antibodies binding to the long-circulating protein, serum albumin, have been identified and fused to small proteins, including other single-domain antibodies to extend their half-life [68,69]. As for effector functions, these are desired in some clinical settings but not others. Fc fusion offers a simple way to endow engineered protein scaffolds with effector functions if needed. Engineered protein scaffolds, like antibodies, offer significant opportunity for sequence diversity but individual protein scaffolds appear to provide less opportunity for structural diversity than do antibodies. The functional impact of lower structural diversity on the ease of being able to generate binding proteins to specific sites on targets of interests remains to be determined. All protein therapeutics, including engineered protein scaffolds, are potentially immunogenic in patients and may elicit an antidrug antibody response that may neutralize or otherwise interfere with the drug's activity. This issue can be more significant for chronic therapy involving repeated drug administration. Clinical trials are ultimately needed to understand the extent to which individual engineered protein scaffold drugs are immunogenic in
  • 7. 1267 E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9 Engineered scaffold proteins Representative assemblies Target valencies Target specificities Long half-life Target A binder 1 1 No Target B binder 1 1 Yes Target C binder 1, 1 2 Yes Half-life extender 2, 1 2 Yes 1, 2 2 Yes 2, 2 2 Yes 1,1,1 3 Yes Fig. 2 – Modular assembly of engineered protein scaffolds into multivalent and multispecific fusion proteins. Engineered scaffold proteins binding to different targets or different epitopes on the same target are identified by selection from libraries. The modular nature of engineered protein scaffolds allows them to be readily assembled into multimers with specific valency for each target [64–66]. However, steric considerations may prevent all modules from being able to bind their respective targets simultaneously. Serum half-life extension is commonly required for engineered scaffold proteins to obtain sufficient exposure to support therapeutic applications. Many different half-life extension strategies for small proteins have been developed [15], including for engineered protein scaffolds (shown), e.g., binding to serum albumin. Linkers between modules may be included but are omitted for simplicity in this figure. patients and whether or not this limits their clinical utility. Human proteins are often selected for engineered protein scaffolds, which may help reduce the risk of immunogenicity in patients for therapeutic applications. Conclusions Proteins are now well established as a clinically and commercially important class of therapeutics. Experience with protein therapeutics has provided an understanding of their strengths and limitations and ways in which they might be improved. Clinical and commercial success with protein therapeutics provides strong motivation to develop better protein drugs, whereas new tools, especially platform technologies provide the likely means to do so. The next generation of protein therapeutics will likely include some of the many enhanced antibodies in early clinical development, optimized Fc fusion proteins, as well as new formats such as engineered protein scaffolds. REFERENCES [1] G. Walsh, Biopharmaceutical benchmarks 2010, Nat. Biotechnol. 28 (2010) 917–924. [2] B. Leader, Q.J. Baca, D.E. Golan, Protein therapeutics: a summary and pharmacological classification, Nat. Rev. Drug Discov. 7 (2008) 21–39. [3] N.C. Nicolaides, P.M. Sass, L. Grasso, Advances in targeted therapeutic agents, Expert Opin. Drug Discov. 5 (2010) 1123–1140. [4] J.M. Reichert, Metrics for antibody therapeutics development, mAbs 2 (2010) 695–700. [5] J.M. Reichert, Antibodies to watch in 2010, mAbs 2 (2010) 28–45. [6] A.C. Chan, P.J. Carter, Therapeutic antibodies for autoimmunity and inflammation, Nat. Rev. Immunol. 10 (2010) 301–316. [7] D.V. Goeddel, D.G. Kleid, F. Bolivar, H.L. Heyneker, D.G. Yansura, R. Crea, T. Hirose, A. Kraszewski, K. Itakura, A.D. Riggs, Expression in Escherichia coli of chemically synthesized genes for human insulin, Proc. Natl Acad. Sci. USA 76 (1979) 106–110. [8] R. Vigneri, S. Squatrito, L. Sciacca, Insulin and its analogs: actions via insulin and IGF receptors, Acta Diabetol. 47 (2010) 271–278. [9] J.S. Freeman, Insulin analog therapy: improving the match with physiologic insulin secretion, J. Am. Osteopath. Assoc. 109 (2009) 26–36. [10] R.J. Solá, K. Griebenow, Glycosylation of therapeutic proteins: an effective strategy to optimize efficacy, BioDrugs 24 (2010) 9–21. [11] S. Elliott, T. Lorenzini, S. Asher, K. Aoki, D. Brankow, L. Buck, L. Busse, D. Chang, J. Fuller, J. Grant, N. Hernday, M. Hokum, S. Hu, A. Knudten, N. Levin, R. Komorowski, F. Martin, R. Navarro, T. Osslund, G. Rogers, N. Rogers, G. Trail, J. Egrie, Enhancement of therapeutic protein in vivo activities through glycoengineering, Nat. Biotechnol. 21 (2003) 414–421. [12] Z. Kiss, S. Elliott, K. Jedynasty, V. Tesar, J. Szegedi, Discovery and basic pharmacology of erythropoiesis-stimulating agents (ESAs), including the hyperglycosylated ESA, darbepoetin alfa: an update of the rationale and clinical impact, Eur. J. Clin. Pharmacol. 66 (2010) 331–340. [13] C. Huang, Receptor-Fc fusion therapeutics, traps, and MIMETIBODY technology, Curr. Opin. Biotechnol. 20 (2009) 692–699. [14] J.A. Jazayeri, G.J. Carroll, Fc-based cytokines: prospects for engineering superior therapeutics, BioDrugs 22 (2008) 11–26. [15] R.E. Kontermann, Strategies to extend plasma half-lives of recombinant antibodies, BioDrugs 23 (2009) 93–109. [16] D.J. Capon, S.M. Chamow, J. Mordenti, S.A. Marsters, T. Gregory, H. Mitsuya, R.A. Byrn, C. Lucas, F.M. Wurm, J.E. Groopman, S. Broder, D.H. Smith, Designing CD4 immunoadhesins for AIDS therapy, Nature 337 (1989) 525–531. [17] D.B. Cines, U. Yasothan, P. Kirkpatrick, Romiplostim, Nat. Rev. Drug Discov. 7 (2008) 887–888. [18] S. Jevševar, M. Kunstelj, V.G. Porekar, PEGylation of therapeutic proteins, Biotechnol. J. 5 (2010) 113–128.
  • 8. 1268 E XP E RI ME N T AL C E L L R E SE A RC H 31 7 ( 20 1 1) 1 2 61 – 1 26 9 [19] F.M. Veronese, A. Mero, The impact of PEGylation on biological therapies, BioDrugs 22 (2008) 315–329. [20] G.B. Kresse, Biosimilars — science, status, and strategic perspective, Eur. J. Pharm. Biopharm. 72 (2009) 479–486. [21] A.P. Chapman, P. Antoniw, M. Spitali, S. West, S. Stephens, D.J. King, Therapeutic antibody fragments with prolonged in vivo half-lives, Nat. Biotechnol. 17 (1999) 780–783. [22] J. Li, C. Yang, Y. Xia, A. Bertino, J. Glaspy, M. Roberts, D.J. Kuter, Thrombocytopenia caused by the development of antibodies to thrombopoietin, Blood 98 (2001) 3241–3248. [23] A. Bendele, J. Seely, C. Richey, G. Sennello, G. Shopp, Short communication: renal tubular vacuolation in animals treated with polyethylene-glycol-conjugated proteins, Toxicol. Sci. 42 (1998) 152–157. [24] J.C. Almagro, J. Fransson, Humanization of antibodies, Front. Biosci. 13 (2008) 1619–1633. [25] N. Lonberg, Fully human antibodies from transgenic mouse and phage display platforms, Curr. Opin. Immunol. 20 (2008) 450–459. [26] N. Lonberg, Human monoclonal antibodies from transgenic mice, Handb. Exp. Pharmacol. (2008) 69–97. [27] H. Thie, T. Meyer, T. Schirrmann, M. Hust, S. Dübel, Phage display derived therapeutic antibodies, Curr. Pharm. Biotechnol. 9 (2008) 439–446. [28] B. Kelley, Industralization of mAb production technology. The bioprocess industry at a crossroads, mAbs 1 (2009) 443–452. [29] M. Better, C.P. Chang, R.R. Robinson, A.H. Horwitz, Escherichia coli secretion of an active chimeric antibody fragment, Science 240 (1988) 1041–1043. [30] A. Skerra, A. Plückthun, Assembly of a functional immunoglobulin Fv fragment in Escherichia coli, Science 240 (1988) 1038–1041. [31] P. Carter, R.F. Kelley, M.L. Rodrigues, B. Snedecor, M. Covarrubias, M.D. Velligan, W.L. Wong, A.M. Rowland, C.E. Kotts, M.E. Carver, M. Yang, J.H. Bourell, H.M. Shepard, D. Henner, High level Escherichia coli expression and production of a bivalent humanized antibody fragment, Biotechnology 10 (1992) 163–167. [32] A. Beck, T. Wurch, C. Bailly, N. Corvaia, Strategies and challenges for the next generation of therapeutic antibodies, Nat. Rev. Immunol. 10 (2010) 345–352. [33] P. Chames, M. Van Regenmortel, E. Weiss, D. Baty, Therapeutic antibodies: successes, limitations and hopes for the future, Br. J. Pharmacol. 157 (2009) 220–233. [34] A.C. Chan, P.J. Carter, Therapeutic antibodies for autoimmunity and inflammation, Nat. Rev. Immunol. 10 (2010) 301–316. [35] J.M. Reichert, Monoclonal antibodies as innovative therapeutics, Curr. Pharm. Biotechnol. 9 (2008) 423–430. [36] A.L. Marschall, A. Frenzel, T. Schirrmann, M. Schüngel, S. Dübel, Targeting antibodies to the cytoplasm, mAbs 3 (2011) 3–16. [37] L. Lampson, Monoclonal antibodies in neuro-oncology: getting past the blood-brain barrier, mAbs 3 (2011) 156–163. [38] B.V. Zlokovic, The blood-brain barrier in health and chronic neurodegenerative disorders, Neuron 57 (2008) 178–201. [39] H.-H. Sedalacek, G. Seemann, D. Hoffmann, J. Czech, P. Lorenz, C. Kolar, K. Bosslet, Antibodies as Carriers of Cytotoxicity, Karger, Munich, Germany, 1992. [40] M.M. Schmidt, K.D. Wittrup, A modeling analysis of the effects of molecular size and binding affinity on tumor targeting, Mol. Cancer Ther. 8 (2009) 2861–2871. [41] M.E. Ackerman, D. Pawlowski, K.D. Wittrup, Effect of antigen turnover rate and expression level on antibody penetration into tumor spheroids, Mol. Cancer Ther. 7 (2008) 2233–2240. [42] H. Hurwitz, L. Fehrenbacher, W. Novotny, T. Cartwright, J. Hainsworth, W. Heim, J. Berlin, A. Baron, S. Griffing, E. Holmgren, N. Ferrara, G. Fyfe, B. Rogers, R. Ross, F. Kabbinavar, Bevacizumab plus irinotecan, fluorouracil, and leucovorin for metastatic colorectal cancer, N. Engl. J. Med. 350 (2004) 2335–2342. [43] D.J. Slamon, B. Leyland-Jones, S. Shak, H. Fuchs, V. Paton, A. Bajamonde, T. Fleming, W. Eiermann, J. Wolter, M. Pegram, J. Baselga, L. Norton, Use of chemotherapy plus a monoclonal [44] [45] [46] [47] [48] [49] [50] [51] [52] [53] [54] [55] [56] [57] [58] [59] [60] [61] [62] [63] [64] [65] antibody against HER2 for metastatic breast cancer that overexpresses HER2, N. Engl. J. Med. 344 (2001) 783–792. L.M. Weiner, R. Surana, S. Wang, Monoclonal antibodies: versatile platforms for cancer immunotherapy, Nat. Rev. Immunol. 10 (2010) 317–327. D.J. Norman, L. Chatenoud, D. Cohen, M. Goldman, C.F. Shield III, Consensus statement regarding OKT3-induced cytokine-release syndrome and human antimouse antibodies, Transplant. Proc. 25 (1993) 89–92. D.K. Arrell, A. Terzic, Network systems biology for drug discovery, Clin. Pharmacol. Ther. 88 (2010) 120–125. A.L. Barabási, N. Gulbahce, J. Loscalzo, Network medicine: a network-based approach to human disease, Nat. Rev. Genet. 12 (2011) 56–68. G.B. Kresse, Biosimilars — science, status, and strategic perspective, Eur. J. Pharm. Biopharm. 72 (2009) 479–486. P.J. Carter, Potent antibody therapeutics by design, Nat. Rev. Immunol. 6 (2006) 343–357. A. Nisonoff, W.J. Mandy, Quantitative estimation of the hybridization of rabbit antibodies, Nature 194 (1962) 355–359. R. Linke, A. Klein, D. Seimetz, Catumaxomab: clinical development and future directions, mAbs 2 (2010) 129–136. R. Bargou, E. Leo, G. Zugmaier, M. Klinger, M. Goebeler, S. Knop, R. Noppeney, A. Viardot, G. Hess, M. Schuler, H. Einsele, C. Brandl, A. Wolf, P. Kirchinger, P. Klappers, M. Schmidt, G. Riethmüller, C. Reinhardt, P.A. Baeuerle, P. Kufer, Tumor regression in cancer patients by very low doses of a T cell-engaging antibody, Science 321 (2008) 974–977. J.S. Marvin, Z. Zhu, Recombinant approaches to IgG-like bispecific antibodies, Acta Pharmacol. Sin. 26 (2005) 649–658. S.C. Alley, N.M. Okeley, P.D. Senter, Antibody-drug conjugates: targeted drug delivery for cancer, Curr. Opin. Chem. Biol. 14 (2010) 529–537. P.J. Carter, P.D. Senter, Antibody-drug conjugates for cancer therapy, Cancer J. 14 (2008) 154–169. A. Younes, N.L. Bartlett, J.P. Leonard, D.A. Kennedy, C.M. Lynch, E.L. Sievers, A. Forero-Torres, Brentuximab vedotin (SGN-35) for relapsed CD30-positive lymphomas, N. Engl. J. Med. 363 (2010) 1812–1821. I. Niculescu-Duvaz, Trastuzumab emtansine, an antibody-drug conjugate for the treatment of HER2+ metastatic breast cancer, Curr. Opin. Mol. Ther. 12 (2010) 350–360. V. Ghetie, S. Popov, J. Borvak, C. Radu, D. Matesoi, C. Medesan, R.J. Ober, E.S. Ward, Increasing the serum persistence of an IgG fragment by random mutagenesis, Nat. Biotechnol. 15 (1997) 637–640. L.G. Presta, Molecular engineering and design of therapeutic antibodies, Curr. Opin. Immunol. 20 (2008) 460–470. J. Zalevsky, A.K. Chamberlain, H.M. Horton, S. Karki, I.W.L. Leung, T.J. Sproule, G.A. Lazar, D.C. Roopenian, J.R. Desjarlais, Enhanced antibody half-life improves in vivo activity, Nat. Biotechnol. 28 (2010) 157–159. T. Igawa, S. Ishii, T. Tachibana, A. Maeda, Y. Higuchi, S. Shimaoka, C. Moriyama, T. Watanabe, R. Takubo, Y. Doi, T. Wakabayashi, A. Hayasaka, S. Kadono, T. Miyazaki, K. Haraya, Y. Sekimori, T. Kojima, Y. Nabuchi, Y. Aso, Y. Kawabe, K. Hattori, Antibody recycling by engineered pH-dependent antigen binding improves the duration of antigen neutralization, Nat. Biotechnol. 28 (2010) 1203–1207. T. Kubota, R. Niwa, M. Satoh, S. Akinaga, K. Shitara, N. Hanai, Engineered therapeutic antibodies with improved effector functions, Cancer Sci. 100 (2009) 1566–1572. G. Cartron, H. Watier, J. Golay, P. Solal-Celigny, From the bench to the bedside: ways to improve rituximab efficacy, Blood 104 (2004) 2635–2642. H.K. Binz, P. Amstutz, A. Plückthun, Engineering novel binding proteins from nonimmunoglobulin domains, Nat. Biotechnol. 23 (2005) 1257–1268. M. Gebauer, A. Skerra, Engineered protein scaffolds as next-generation antibody therapeutics, Curr. Opin. Chem. Biol. 13 (2009) 245–255.
  • 9. E XP E RI ME N T AL C E L L R E S EA RC H 31 7 ( 20 1 1) 1 2 6 1– 1 26 9 [66] S.D. Nuttall, R.B. Walsh, Display scaffolds: protein engineering for novel therapeutics, Curr. Opin. Pharmacol. 8 (2008) 609–615. [67] B. Zuraw, U. Yasothan, P. Kirkpatrick, Ecallantide, Nat. Rev. Drug Discov. 9 (2010) 189–190. [68] A. Walker, G. Dunlevy, D. Rycroft, P. Topley, L.J. Holt, T. Herbert, M. Davies, F. Cook, S. Holmes, L. Jespers, C. Herring, Anti-serum 1269 albumin domain antibodies in the development of highly potent, efficacious and long-acting interferon, Protein Eng. Des. Sel. 23 (2010) 271–278. [69] L.J. Holt, A. Basran, K. Jones, J. Chorlton, L.S. Jespers, N.D. Brewis, I.M. Tomlinson, Anti-serum albumin domain antibodies for extending the half-lives of short lived drugs, Protein Eng. Des. Sel. 21 (2008) 283–288.