SlideShare a Scribd company logo
1 of 9
Download to read offline
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 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .        .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1262
   Brief history of protein therapeutics . . . . . . . . . . . . . . . . . . .         .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1262
       Protein therapeutics enabled by protein engineering . . . . . . . . .           .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1262
       Insulin — the first recombinant human protein therapeutic . . . . .             .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1262
       Beginning of engineered proteins as therapeutics . . . . . . . . . . .          .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1263
       Fc fusion proteins . . . . . . . . . . . . . . . . . . . . . . . . . . .        .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1263
       Protein conjugation to polyethylene glycol . . . . . . . . . . . . . .          .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1263
       The rise of antibody therapeutics . . . . . . . . . . . . . . . . . . .         .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1263
   Rationale for next generation protein therapeutics . . . . . . . . . . . .          .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1264
       Strengths and limitations of antibody and other protein therapeutics            .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1264
       Clinical rationale . . . . . . . . . . . . . . . . . . . . . . . . . . .        .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1264
       Scientific rationale . . . . . . . . . . . . . . . . . . . . . . . . . . .      .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1264


 ⁎ 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                                    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 . . . . . . . . . . . . . . . . . . . .         .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1265
  Platform technologies for next generation protein therapeutics           .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1265
      Next generation antibodies and Fc fusion proteins . . . . .          .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1265
      Engineered protein scaffolds . . . . . . . . . . . . . . . .         .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1266
  Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . .        .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1267
  References . . . . . . . . . . . . . . . . . . . . . . . . . . . .       .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   .   1267




Introduction                                                                       constraints limit examples here to a sampling of technologies of
                                                                                   broad applicability — so called platform technologies. The inter-
Since the early 1980s proteins have emerged as a major new class                   ested reader is referred to many excellent articles on protein
of pharmaceuticals with ~200 marketed products that are mainly                     therapeutics including those in this special issue and others cited
therapeutics with a small number of diagnostics and vaccines [1].                  herein.
Protein therapeutics can be classified based upon their pharma-
cologic activity as drugs that: i) replace a protein that is deficient
or abnormal, ii) augment an existing pathway, iii) provide a novel                 Brief history of protein therapeutics
function or activity, iv) interfere with a molecule or organism, or
iv) deliver a payload such as a radionuclide, cytotoxic drug, or                   Protein therapeutics enabled by protein engineering
protein effector [2]. Alternatively, protein therapeutics can be
grouped into molecular types that include: antibody-based drugs,                   Protein therapeutics and its enabling sister discipline, protein
anticoagulants, blood factors, bone morphogenetic proteins,                        engineering, have emerged since the early 1980s. Protein engi-
engineered protein scaffolds, enzymes, Fc fusion proteins, growth                  neering by rational design or molecular evolution allows the
factors, hormones, interferons, interleukins, and thrombolytics                    systemic dissection of protein structure–function relationships and
(Fig. 1) [1,3]. Antibody-based drugs are the largest and fastest                   the generation of novel proteins with modified activities or entirely
growing class of protein therapeutics with 24 marketed antibody                    new properties. Indeed, protein engineering has revolutionized
drugs in the USA [4] and over 240 more in clinical development [5].                protein therapeutics by providing the tools to customize existing
    This introductory article provides a brief history of protein                  proteins or to create novel proteins for specific clinical applications.
therapeutics, offers a rationale for developing better protein drugs,                 A brief history of protein therapeutics is provided here from a
and identifies some major future opportunities as well as emerging                 protein engineering perspective. The focus is on technologies with
technologies that may meet them. The 30-year history of protein                    the greatest impact on current protein therapeutics — engineered
drugs is illustrated here with examples that pioneered their class or              versions of natural proteins, Fc fusion proteins, conjugation to the
have high clinical or commercial significance. The rationale for                   polymer, polyethylene glycol (PEG), and antibody therapeutics as
creating better protein drugs, including antibodies [6], comes from                the quintessential protein therapeutic platform.
the convergence of clinical, scientific, technological and commer-
cial drivers that collectively have identified major unmet needs and               Insulin — the first recombinant human protein therapeutic
provide strategies to address them. Major future opportunities for
protein therapeutics, are improved efficacy, greater safety, reduced               The first human protein therapeutic derived from recombinant
immunogenicity or improved delivery. A plethora of emerging                        DNA technology was human insulin (Humulin®) created at
technologies for the generation and optimization of protein ther-                  Genentech [7], developed by Eli Lilly, and approved by the US
apeutics are anticipated to help address these opportunities. Space                Food and Drug Administration (FDA) in 1982. Recombinant insulin



                                       Recombinant                             Antibodies                                                                      Fc fusion
                                         proteins                                                                                                              proteins
                                                                 Fab fragment                                  IgG
                                       Anticoagulants
                                       Blood factors                                                                                                                           Receptor
                                       Bone morphogenetic                                                                                                                      Ligand
                                          proteins                                                                                                                             Peptide
                                       Enzymes
                                       Growth factors
                                       Hormones                                                                                    Fc
                                       Interferons
                                       Interleukins
                                       Thrombolytics

               Optional              Conjugate PEG              Conjugate PEG                    Bispecific
             modifications          Engineer protein                                          Conjugate drug
                                  Engineer glycosylation                                   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                           1263


was used to replace its natural counterpart that can be deficient in               Beyond half-life extension, Fc fusion can provide several
diabetes mellitus. Other recombinant human proteins were soon                  additional benefits such as facilitating expression and secretion
developed as therapeutics to replace the natural proteins deficient            of recombinant protein, enabling facile purification by protein A
in some patients (e.g., growth hormone) or augment existing                    chromatography, improving solubility and stability, and enhancing
pathways (e.g., interferon-α, tissue plasminogen activator, and                potency by increasing valency. Optional additional properties
erythropoietin) [2].                                                           conferred by Fc fusion include binding to Fcγ receptors and/or
                                                                               complement to support secondary immune functions [13,14]. The
Beginning of engineered proteins as therapeutics                               first demonstration of the Fc fusion protein concept was CD4-Fc
                                                                               [16]. Five Fc fusion protein therapeutics were approved by 2010,
Beyond recombinant versions of natural proteins, the next major                the prototypical and most successful drug of this class being
step in the evolution of protein therapeutics was the redesign of              etanercept (Enbrel®), a TNF receptor 2-Fc fusion protein [1].
proteins to increase their clinical potential. Injected insulin fails to       Etanercept was the top-selling protein therapeutic in 2009 with
adequately mimic endogenous insulin in two major ways: rapid                   $6.6 billion in world-wide sales in rheumatoid arthritis and other
onset of action after meals and prolonged maintenance of low level             autoimmune diseases [1]. Romiplostim (Nplate®) is the first
insulin between meals [8]. Insulin has been engineered, commonly               peptide-Fc fusion (“peptibody”) approved as a human therapeutic.
by one to three amino acid replacements, to create analogs that are            Romiplostim is a thrombopoietin (TPO) receptor agonist approved
either rapid-acting or long-acting and better mimic different                  as a treatment for thrombocytopenia [17].
properties of endogenous insulin [8]. Multiple insulin analogs have
been approved for the treatment of diabetes mellitus including the             Protein conjugation to polyethylene glycol
rapid-acting analogs, insulin lispro (Humalog®), insulin aspart
(NovoRapid® and Novolog®) and insulin glulisine (Apidra®) and                  Protein conjugation to chemicals such as radionuclide chelators,
the long-acting analogs, insulin glargine (Optisulin® and Lantus®)             cytotoxic drugs and PEG can endow proteins with brand new
and insulin detemir (Levemir®) [1,8]. Insulin analogs offer mul-               capabilities to increase their clinical potential.
tiple benefits over first generation recombinant insulin drugs that                The most clinically and commercially successful protein
include improved physiologic profile, greater convenience for                  conjugates thus far have been with PEG. Protein conjugation to
patients, reduced risk of hypoglycemia, and less weight gain in                PEG – “PEGylation” – increases their hydrodynamic volume,
some cases [9]. The commercial importance of engineered insulins               prevents rapid renal clearance, and thereby increases the serum
is illustrated by Lantus®, which was one of the top ten selling                half-life, as extensively reviewed [15,18,19]. At least 8 PEGylated
biopharmaceuticals in 2009 [1].                                                proteins are approved as human therapeutics [18]. Indeed, long-
    Many protein therapeutics are produced in mammalian cells                  acting second generation interferon-α molecules that are PEGy-
and have one or more N- and/or O-linked glycans [10]. Engineering              lated have largely superseded their first generation non-PEGylated
a protein with additional glycosylation sites can increase the in              counterparts [20]. Benefits of PEGylation are that it is possible to
vivo serum half-life and thereby target exposure [11]. An early                tune the serum-half of proteins by varying the number of attached
example of such “glycoengineering” of proteins for half-life                   PEG molecules, their size and extent of branching [21]. PEGylation
extension is provided by darbepoetin-α (Aranesp®), administered                can also reduce the immunogenicity of proteins [18,19], although
to anemic patients to stimulate the production of red blood cells in           PEGylated proteins can still sometimes be immunogenic [22]. A
the bone marrow. The first generation drug, epoetin-α, has 3 N-                draw back to PEGylation is that it can impair protein function, a
linked glycosylation sites and was engineered with 2 additional                limitation that is offset by the greatly increased half-life, and
glycosylation sites to create darbepoetin-α, a commercially                    potentially ameliorated by site-specific PEGylation and/or tailoring
successful longer-acting second generation drug [12]. Protein                  the PEG [18,19]. PEG is a non-natural and apparently non-
glycosylation can sometimes reduce bioactivity, albeit offset by               biodegradable polymer, a molecular attribute that has unclear
longer half-life. Darbepoetin-α has higher specific activity in                clinical significance. PEGylated proteins can induce renal tubular
patients than epoetin-α, reflecting that the 3-fold increase in                vacuolation in animals, albeit with unaltered clinical pathology
serum half-life that increases drug exposure more than offsets a 5-            [23] and unknown impact on long term safety.
fold reduction in receptor binding affinity [12].
                                                                               The rise of antibody therapeutics
Fc fusion proteins
                                                                               Antibody-based drugs are the largest and fastest growing class of
Another significant advance in protein therapeutics came with the              protein therapeutics with 24 marketed antibody therapeutics in
advent of fusion proteins by joining the genes encoding 2 or more              the USA (28 FDA-approved and 4 later withdrawn from the
different proteins. Fusion proteins combine properties of their                market) [4] and at least 240 more in clinical development [5].
component parts. The most clinically and commercially successful               Antibody-based drugs contributed $38 billion of $99 billion in
fusion protein therapeutics to date contain the Fc region of                   world-wide sales for protein biopharmaceuticals in 2009 [1].
immunoglobulins [1]. Such Fc fusions can endow peptides or                     Moreover, 5 of the 10 top-selling protein therapeutics in 2009
proteins with the IgG-like property of long serum half-life (days to           were antibodies, namely, infliximab (Remicade®), bevacizumab
weeks), by virtue of binding to the salvage receptor, FcRn [13,14]. By         (Avastin®), rituximab (Rituxan® and MabThera®), adalimumab
contrast, proteins of ~70 kDa and smaller are typically eliminated             (Humira®) and trastuzumab (Herceptin®) [1].
rapidly from circulation by renal filtration and have half-lives of a             The rapid growth of antibody therapeutics since the mid-1990s
few minutes to a few hours, that can in many cases render them                 was made possible by the advent of antibody chimerization
marginal or unsuitable for therapeutic applications [15].                      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             other drug formats is that the success rate has been somewhat
the emergence of facile routes to human antibodies using                     higher — 17% for humanized antibodies from the first clinical trial
transgenic mice [25,26] or phage display [25,27] technologies.               to regulatory approval [35].
Other advances that have contributed to the success of antibodies                Proteins have several significant limitations as therapeutics. For
as therapeutics include the industrialization of antibody produc-            example, protein therapeutics are very expensive, reflecting high
tion technologies [28] that can provide sufficient quantities of             production costs, that may limit patient access and also clinical
antibodies to allow higher doses to be used in patients. The de-             applications [28]. This high cost issue is exacerbated for protein
velopment of methods to express and secrete functional antibody              therapeutics where multi-gram doses are needed for a treatment
fragments from E. coli [29,30] has significantly stimulated the field        course, as is the case for some antibodies. Intracellular delivery of
of antibody engineering and enabled subsequent technologies                  proteins is possible in the laboratory setting, e.g., by protein
such as phage display. The next major advance in the expression of           transduction [36]. However, targeted pharmaceutic delivery of
antibody fragments in E. coli was high titer (gram per liter)                proteins to reach intracellular targets is in its infancy, so protein
production in a fermentor [31], that facilitated the development of          therapeutics are currently limited to cell surface or extracellular
antibody fragment drugs such as the anti-VEGF Fab, ranibizumab               targets.
(Lucentis®). The emergence of antibody as therapeutics likely also               Another shortcoming of proteins therapeutics is that they are
reflects improved choices of antibody and target antigen for                 denatured and/or proteolyzed in the gut and are thus not orally
intervention in the pathobiology of disease.                                 bioavailable. Proteins, including antibodies [37], show very poor
    The majority of approved antibody drugs are unmodified                   ability to penetrate the specialized endothelial structure of the
(“naked”) IgG molecules. However, other antibody formats have                neurovasculature known as the blood-brain barrier. The integrity
been approved for therapy including IgG “armed” with radio-                  of the blood-brain barrier can be partially compromised in some
nuclides or cytotoxic drugs (antibody-drug conjugates) for cancer            pathological settings such as chronic neurodegenerative diseases
treatment, as well as Fab fragments and a PEGylated Fab′ fragment            [38]. Nevertheless, proteins with current technologies are poorly
[1].                                                                         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
Rationale for next generation protein therapeutics                           other proteins and this large size may limit their tissue penetration
                                                                             and ability to localize to their targets. For example, antibody
A major quest with protein therapeutics, especially antibodies               localization to tumors in patients is typically very inefficient:
[6,32], is the development of even better next generation drugs              ≤0.01% of the injected dose per gram of tumor [39]. Comparison of
with enhanced efficacy, greater safety or improved delivery.                 mathematical modeling with experimental data, strongly suggests
Current antibody therapeutics reveal major strengths that provide            that molecular size and binding affinity are major determinants of
a foundation upon which to build, as well as significant limitations         tumor targeting [40]. Many other factors likely contribute to tumor
to overcome that offers new opportunities [6,33]. Some antibody              penetration including antigen expression level and turnover rate
strengths and many of their limitations apply more broadly to                [41].
protein therapeutics and are discussed below as a compass to guide
the development of next generation protein therapeutics. The                 Clinical rationale
impetus to develop better protein therapeutics is also discussed
here as a convergence of clinical, scientific, and commercial forces         From a clinical standpoint, first generation protein drugs can
that identify unmeet needs (this section), in conjunction with               provide substantial patient benefit. However, in many cases there
technological tools that many help address them (next section).              is room for improvement in efficacy and/or safety. For example,
                                                                             several anti-cancer antibodies provide an overall survival benefit,
Strengths and limitations of antibody and other protein                      including bevacizumab [42] and trastuzumab [43] in combination
therapeutics                                                                 with chemotherapy in metastatic colorectal and breast cancers,
                                                                             respectively. Nevertheless, there remains plenty of opportunity for
One of the greatest strength of antibodies as a therapeutic platform         enhancing antibodies for cancer treatment, such as elevating the
is the potential ease of generating high affinity and specificity            overall survival rate of patients, as well as many strategies being
human antibodies to virtually any target of therapeutic interest,            explored to do so [44]. All therapeutic proteins are potentially
e.g., using transgenic mice [25,26] or phage display [25,27]                 immunogenic in patients and occasionally an anti-drug antibody
technologies. Moreover, antibodies are readily tailored for thera-           response can lead to a major safety issue. For example, a truncated
peutic applications by modulating (enhancing or attenuating)                 and PEGylated form of TPO (MGDF-PEG) elicited an antibody
their existing properties or endowing them with new activities as            response in some patients that neutralized endogenous TPO
discussed in a later section and reviewed more extensively                   leading to severe and persistent anemia, i.e., the opposite of the
elsewhere [33,34]. As for other protein therapeutics, antibodies             desired effect of increasing platelet counts [22]. This issue was
are commonly well tolerated by patients, but not invariably so.              overcome with the peptibody, Romiplostim (see Fc fusion proteins
Antibody drug development benefits from recombinant produc-                  section) [17].
tion methods that are well-established, broadly applicable and
commonly support expression at high titer (multi-gram per liter)             Scientific rationale
[28]. Widely available knowledge and experience across all facets
of antibody drug development also facilitates the generation of              Scientifically, our expanding knowledge suggests drug mechanisms
future antibody therapeutics. An advantage of antibodies over                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                            1265


overcome. For example, anti-CD3 antibodies illustrate the benefits of         generation antibody and Fc fusion protein therapeutics and the
understanding and overcoming mechanisms of drug toxicity and                  emerging technologies of engineered protein scaffolds.
resistance. In 1986 the anti-CD3 antibody, muromonab-CD3
(OKT3®), became the first antibody approved by the FDA for                    Next generation antibodies and Fc fusion proteins
human therapy — for the treatment of acute allograft rejection in
renal transplant patients. In 1993 muromonab-CD3 was approved                 Many promising strategies are emerging to enhance the existing
for the treatment of acute rejection of heart and liver transplants in        properties of antibodies or to endow them with new activities, as
patients resistant to steroid treatment. Unfortunately, muromonab-            reviewed in detail elsewhere [6,32,33]. Antibodies (IgG) have
CD3 activates T cells resulting in transient cytokine release and acute       multiple binding partners including antigen, Fcγ receptors,
and severe flu-like symptoms [45]. Another major limitation of                complement and FcRn. Each of these interactions can be tailored
muromonab-CD3 is that its therapeutic benefit is reduced by                   in ways that may improve the clinical properties of antibodies
neutralizing antibodies that develop in ~50% of patients [45].                [32,34,49]. Platform technologies that seem likely to impact the
Muromonab-CD3 has since been voluntarily withdrawn from the                   next generation of antibody therapeutics include old concepts that
US market due to decreased sales. These major limitations of                  are finally coming to fruition, such as antibody-drug conjugates
muromonab-CD3 have been greatly reduced, if not overcome, in                  and bispecific antibodies. Technologies for improving Fc-mediated
some second generation anti-CD3 antibodies that have advanced to              functions, such as half-life extension and effector function en-
late stage clinical development [6]. For example, the anti-CD3                hancement, seem likely to benefit the next generation of antibody
antibodies, teplizumab and otelixizumab, combine Fc engineering to            therapeutics. Additionally, these technologies appear directly
attenuate Fcγ receptor binding with chimerization or humanization             applicable to the optimization of Fc fusion proteins.
to reduce immunogenicity [34].                                                    The 50-year old concept of bispecific antibodies [50] came of
    Drug development, including for protein therapeutics, seems               age in 2009 with the first regulatory approval of a bispecific
likely to gain from advances in the understanding of targets and              antibody — catumaxomab (Removab®, anti-EpCAM x anti-CD3) in
their role in the pathobiology of disease. For example, there is a            Europe. Catumaxomab is approved for the treatment of malignant
growing appreciation that targets are not isolated gene products              ascites in patients with EpCAM-positive carcinomas [1,51].
but integral components of networks of interconnected signaling               Recruitment of T cells via the anti-CD3 arm of catumaxomab and
pathways [46,47].                                                             other immune effector cells via its Fc region may contribute to
                                                                              antitumor activity. A tandem bispecific scFv format known as a
Commercial rationale                                                          “BiTE”, that binds CD3 and a tumor-associated antigen, is a
                                                                              particularly effective way to retarget T cells to kill tumor cells.
Clinical and commercial success with protein therapeutics has                 Blinatumomab, a BiTE specific for CD19 and CD3, gave several
bred intense competition between different organizations striving             complete responses in a phase I clinical trial in non-Hodgkin's
to develop protein therapeutics to the same antigen and/or                    lymphoma [52] and is currently in multiple phase II clinical trials.
overlapping therapeutic indications. Indeed, this is already                  Another major area of current interest with bispecific antibodies is
reflected in marketed drugs as illustrated by 5 different protein             in the simultaneous blockade of 2 disease mediators, which may
therapeutics that block TNF including 3 different IgG (infliximab,            lead to greater therapeutic benefit than targeting individual
adalimumab and golimumab), a PEGylated Fab′ fragment (certo-                  disease mediators [53]. Until recently, a major bottleneck in the
lizumab pegol) and a receptor-Fc fusion (etanercept) [6]. Such                development of bispecific antibodies as therapeutics was the
competition is increasingly commonplace as patents dominating                 challenge of producing these complex molecules in sufficient
individual targets expire. The generation of high affinity and                quantity and quality for clinical applications. This issue has been
potency IgG is often possible, arguably commoditized. This                    largely overcome by the advent of many alternative formats and
provides a strong commercial incentive to develop superior-                   production methods for bispecific antibodies [6].
performing antibody drugs to differentiate products from those of                 Antibody-drug conjugates, a concept that dates back to the
competitors. The success of innovator protein therapeutics has                1970s, use antibodies to deliver a cytotoxic payload to kill a target
brought a rise in biosimilar efforts (i.e., products that are deemed          such as tumor cells [54,55]. One antibody-drug conjugate,
highly similar to innovator protein therapeutics according to drug            gemtuzumab ozogamicin (Mylotarg®), was approved by the FDA
regulatory authorities). Indeed, several biosimilar approval appli-           for the treatment of acute myeloid leukemia. Gemtuzumab
cations have been filed in the European Union with 13 biosimilars             ozogamicin was withdrawn from the market in 2010 due to safety
being approved by European drug regulators by early 2009 [48].                concerns and insufficient patient benefit in post approval clinical
For creators of innovator drugs, the impetus to develop improved              trials. Much progress has been made over the decades in many
next generation protein therapeutics that out perform the                     different facets of antibody-drug conjugates, including better
previous generation drugs has increased substantially [48].                   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,
Platform technologies for next generation                                     [56]) in Hodgkin's disease and trastuzumab emtansine (T-DM1,
protein therapeutics                                                          [57]) in metastatic breast cancer over-expressing HER2/neu
                                                                              following demonstration of robust antitumor activity and accept-
Many protein engineering tools have been developed that allow                 able safety profiles in earlier clinical trials.
one to optimize favorable properties of proteins, attenuate                       Engineering proteins to increase their serum half-life has the
undesired attributes and create proteins with entirely novel                  potential benefits of increased exposure leading to greater target
activities. Space constraints limit the discussion here to next               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        ribosome display. Increased binding affinity – affinity maturation –
be extended by at least a few fold by engineering the Fc region for           is often needed and readily accomplished by further library
higher affinity binding to the salvage receptor (FcRn), whilst                building and selection.
preserving the pH dependence of binding [58]. Half-life extension                 Over 50 different engineered protein scaffolds have been
of Fc-engineered antibodies in non-human primates was subse-                  described with a few advancing into clinical development [64–
quently demonstrated by several different groups [59]. Antibody               66] including: Adnectins® from type III fibronectin domains,
half-life extension can result in increased target exposure and               Affibodies® from the synthetic Z domain of staphylococcal protein
improved preclinical in vivo efficacy as recently shown for anti-             A, Anticalins® from lipocalins, Avimers® from LDLR-A modules,
EGFR and anti-VEGF antibodies [60]. Engineering antibodies for                DARPins® from ankyrin repeat proteins, and engineered Kunitz
pH-dependent interaction with their target antigen has also been              domains. Engineered protein scaffolds also include single-domain
shown to extend antibody half-life in vivo [61]. The first half-life          antibodies from human (dAbs®) and camelids (Nanobodies®).
extended antibody to enter clinical trials was the humanized anti-            One engineered protein scaffold protein has been approved for
RSV antibody, MEDI-557, although pharmacokinetic data have yet                therapy, namely, ecallantide (Kalbitor®), a potent and selective
to be reported.                                                               inhibitor of plasma kallikrein based upon a Kunitz domain [67].
    Another major area of interest for improving antibody per-                    Many potential advantages of engineered protein scaffolds over
formance is effector function enhancement, as reviewed exten-                 antibodies have been proposed [64–66], albeit with limited
sively elsewhere [34,62] and discussed briefly below. Antibody                supporting evidence to date in the context of developing protein
effector functions include ADCC (antibody-dependent cellular                  therapeutics. For example, the smaller size and higher stability of
cytotoxicity), ADCP (antibody-dependent cellular phagocytosis)                some engineered protein scaffolds may allow for alternative routes
and complement-dependent cytotoxicity (CDC). Effector functions               of administration such as pulmonary delivery. The smaller size of
may be advantageous where destruction of the target cells is                  engineered protein scaffolds over antibodies favors more efficient
desired, as is often the case for oncology targets.                           tissue penetration, but this can be more than offset by much faster
    Indeed many antibodies approved for oncologic indications                 clearance. Engineered protein scaffolds may be more readily
(e.g., rituximab, trastuzumab, alemtuzumab, cetuximab and                     expressed in microbial hosts reflecting their high stability and
ofatumumab) support one or many effector functions that may                   solubility, small size, simple structure (single polypeptide), and
contribute to their clinical antitumor activity, with the strongest           that they commonly lack disulfide bonds and/or glycosylation
evidence being for rituximab [63].                                            sites. The small and modular architecture of engineered protein
    For antibody and Fc fusion protein therapeutics potential                 scaffolds seems particularly well suited to the construction of
benefits from enhancing effector functions include greater efficacy,          multivalent and/or multispecific protein therapeutics (Fig. 2).
lower or less frequent dosing and decreased cost-of-goods. As for             Engineered proteins are commonly associated with independent
risks, these include more frequent or severe adverse events. ADCC,            intellectual property that is outside the scope of antibody patents.
ADCP and CDC have been augmented by Fc protein engineering as                 Several of these proposed advantages of engineered protein
extensively reviewed elsewhere [62]. ADCC has also been                       scaffolds have been achieved with antibodies by engineering for
augmented to a similar level by modifying the glycan attached to              improved stability or expression, or by exploiting the modular
the Fc, e.g., afucosylation achieved through cell line engineering            architecture of antibodies to create different formats including
[62]. At least 8 effector function enhanced antibodies have                   ones that are smaller and simpler than IgG [6,32,33].
advanced into early clinical development [34].                                    Engineered protein scaffolds lack Fc-mediated properties of
                                                                              antibodies such as long serum half-life and effector functions. This
Engineered protein scaffolds                                                  does not appear to be a major limitation, as many different
                                                                              strategies have been developed to extend the half-life of proteins
The success of antibodies as protein therapeutics has inspired the            as comprehensively reviewed [15]. For example, single-domain
development of so-called “engineered protein scaffolds” as alterna-           antibodies binding to the long-circulating protein, serum albumin,
tive platform technologies for the development of novel next                  have been identified and fused to small proteins, including other
generation protein therapeutics [64–66]. Engineered protein scaf-             single-domain antibodies to extend their half-life [68,69]. As for
folds provide binding sites that can be tailored to specifically              effector functions, these are desired in some clinical settings but
recognize desired target molecules, in an analogous way that                  not others. Fc fusion offers a simple way to endow engineered
antibodies can be developed to recognize their target antigens.               protein scaffolds with effector functions if needed. Engineered
Here the basic features of engineered protein scaffolds are described,        protein scaffolds, like antibodies, offer significant opportunity for
together with their strengths, limitations and some possible future           sequence diversity but individual protein scaffolds appear to
directions.                                                                   provide less opportunity for structural diversity than do anti-
    Protein scaffolds chosen for engineering have known 3-                    bodies. The functional impact of lower structural diversity on the
dimensional structures that together represent diverse protein                ease of being able to generate binding proteins to specific sites on
folds [64–66]. Protein scaffolds are commonly selected with the               targets of interests remains to be determined.
following attributes: small, monomeric, high solubility and                       All protein therapeutics, including engineered protein scaffolds,
stability, and that lack disulfide bonds and glycosylation sites.             are potentially immunogenic in patients and may elicit an anti-
Protein scaffolds are typically readily expressed in microbial hosts.         drug antibody response that may neutralize or otherwise interfere
Sequence diversity is introduced into the protein scaffold, e.g., by          with the drug's activity. This issue can be more significant for
randomizing surface accessible residues, to create a so-called                chronic therapy involving repeated drug administration. Clinical
library. Binders to a target of therapeutic interest are then iden-           trials are ultimately needed to understand the extent to which
tified by selection using technologies such as phage, yeast or                individual engineered protein scaffold drugs are immunogenic in
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                                  1267

                           Engineered scaffold             Representative            Target          Target         Long
                                proteins                     assemblies             valencies      specificities   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          [6] A.C. Chan, P.J. Carter, Therapeutic antibodies for autoimmunity
proteins are often selected for engineered protein scaffolds, which                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.
may help reduce the risk of immunogenicity in patients for ther-
                                                                                   Crea, T. Hirose, A. Kraszewski, K. Itakura, A.D. Riggs, Expression in
apeutic applications.                                                              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.
Conclusions                                                                    [9] J.S. Freeman, Insulin analog therapy: improving the match with
                                                                                   physiologic insulin secretion, J. Am. Osteopath. Assoc. 109 (2009)
Proteins are now well established as a clinically and commercially                 26–36.
important class of therapeutics. Experience with protein thera-               [10] R.J. Solá, K. Griebenow, Glycosylation of therapeutic proteins: an
                                                                                   effective strategy to optimize efficacy, BioDrugs 24 (2010) 9–21.
peutics has provided an understanding of their strengths and
                                                                              [11] S. Elliott, T. Lorenzini, S. Asher, K. Aoki, D. Brankow, L. Buck, L.
limitations and ways in which they might be improved. Clinical
                                                                                   Busse, D. Chang, J. Fuller, J. Grant, N. Hernday, M. Hokum, S. Hu, A.
and commercial success with protein therapeutics provides strong                   Knudten, N. Levin, R. Komorowski, F. Martin, R. Navarro, T.
motivation to develop better protein drugs, whereas new tools,                     Osslund, G. Rogers, N. Rogers, G. Trail, J. Egrie, Enhancement of
especially platform technologies provide the likely means to do so.                therapeutic protein in vivo activities through glycoengineering,
The next generation of protein therapeutics will likely include                    Nat. Biotechnol. 21 (2003) 414–421.
some of the many enhanced antibodies in early clinical develop-               [12] Z. Kiss, S. Elliott, K. Jedynasty, V. Tesar, J. Szegedi, Discovery and
                                                                                   basic pharmacology of erythropoiesis-stimulating agents (ESAs),
ment, optimized Fc fusion proteins, as well as new formats such as
                                                                                   including the hyperglycosylated ESA, darbepoetin alfa: an update
engineered protein scaffolds.                                                      of the rationale and clinical impact, Eur. J. Clin. Pharmacol. 66
                                                                                   (2010) 331–340.
                                                                              [13] C. Huang, Receptor-Fc fusion therapeutics, traps, and
REFERENCES                                                                         MIMETIBODY technology, Curr. Opin. Biotechnol. 20 (2009)
                                                                                   692–699.
                                                                              [14] J.A. Jazayeri, G.J. Carroll, Fc-based cytokines: prospects for
 [1] G. Walsh, Biopharmaceutical benchmarks 2010, Nat. Biotechnol.                 engineering superior therapeutics, BioDrugs 22 (2008) 11–26.
     28 (2010) 917–924.                                                       [15] R.E. Kontermann, Strategies to extend plasma half-lives of
 [2] B. Leader, Q.J. Baca, D.E. Golan, Protein therapeutics: a summary             recombinant antibodies, BioDrugs 23 (2009) 93–109.
     and pharmacological classification, Nat. Rev. Drug Discov. 7             [16] D.J. Capon, S.M. Chamow, J. Mordenti, S.A. Marsters, T. Gregory, H.
     (2008) 21–39.                                                                 Mitsuya, R.A. Byrn, C. Lucas, F.M. Wurm, J.E. Groopman, S. Broder,
 [3] N.C. Nicolaides, P.M. Sass, L. Grasso, Advances in targeted                   D.H. Smith, Designing CD4 immunoadhesins for AIDS therapy,
     therapeutic agents, Expert Opin. Drug Discov. 5 (2010)                        Nature 337 (1989) 525–531.
     1123–1140.                                                               [17] D.B. Cines, U. Yasothan, P. Kirkpatrick, Romiplostim, Nat. Rev.
 [4] J.M. Reichert, Metrics for antibody therapeutics development,                 Drug Discov. 7 (2008) 887–888.
     mAbs 2 (2010) 695–700.                                                   [18] S. Jevševar, M. Kunstelj, V.G. Porekar, PEGylation of therapeutic
 [5] J.M. Reichert, Antibodies to watch in 2010, mAbs 2 (2010) 28–45.              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                      antibody against HER2 for metastatic breast cancer that
     therapies, BioDrugs 22 (2008) 315–329.                                              overexpresses HER2, N. Engl. J. Med. 344 (2001) 783–792.
[20] G.B. Kresse, Biosimilars — science, status, and strategic                    [44]   L.M. Weiner, R. Surana, S. Wang, Monoclonal antibodies: versatile
     perspective, Eur. J. Pharm. Biopharm. 72 (2009) 479–486.                            platforms for cancer immunotherapy, Nat. Rev. Immunol. 10
[21] A.P. Chapman, P. Antoniw, M. Spitali, S. West, S. Stephens, D.J.                    (2010) 317–327.
     King, Therapeutic antibody fragments with prolonged in vivo                  [45]   D.J. Norman, L. Chatenoud, D. Cohen, M. Goldman, C.F. Shield III,
     half-lives, Nat. Biotechnol. 17 (1999) 780–783.                                     Consensus statement regarding OKT3-induced cytokine-release
[22] J. Li, C. Yang, Y. Xia, A. Bertino, J. Glaspy, M. Roberts, D.J. Kuter,              syndrome and human antimouse antibodies, Transplant. Proc. 25
     Thrombocytopenia caused by the development of antibodies to                         (1993) 89–92.
     thrombopoietin, Blood 98 (2001) 3241–3248.                                   [46]   D.K. Arrell, A. Terzic, Network systems biology for drug discovery,
[23] A. Bendele, J. Seely, C. Richey, G. Sennello, G. Shopp, Short                       Clin. Pharmacol. Ther. 88 (2010) 120–125.
     communication: renal tubular vacuolation in animals treated                  [47]   A.L. Barabási, N. Gulbahce, J. Loscalzo, Network medicine: a
     with polyethylene-glycol-conjugated proteins, Toxicol. Sci. 42                      network-based approach to human disease, Nat. Rev. Genet. 12
     (1998) 152–157.                                                                     (2011) 56–68.
[24] J.C. Almagro, J. Fransson, Humanization of antibodies, Front.                [48]   G.B. Kresse, Biosimilars — science, status, and strategic
     Biosci. 13 (2008) 1619–1633.                                                        perspective, Eur. J. Pharm. Biopharm. 72 (2009) 479–486.
[25] N. Lonberg, Fully human antibodies from transgenic mouse and                 [49]   P.J. Carter, Potent antibody therapeutics by design, Nat. Rev.
     phage display platforms, Curr. Opin. Immunol. 20 (2008)                             Immunol. 6 (2006) 343–357.
     450–459.                                                                     [50]   A. Nisonoff, W.J. Mandy, Quantitative estimation of the
[26] N. Lonberg, Human monoclonal antibodies from transgenic mice,                       hybridization of rabbit antibodies, Nature 194 (1962) 355–359.
     Handb. Exp. Pharmacol. (2008) 69–97.                                         [51]   R. Linke, A. Klein, D. Seimetz, Catumaxomab: clinical
[27] H. Thie, T. Meyer, T. Schirrmann, M. Hust, S. Dübel, Phage display                  development and future directions, mAbs 2 (2010) 129–136.
     derived therapeutic antibodies, Curr. Pharm. Biotechnol. 9 (2008)            [52]   R. Bargou, E. Leo, G. Zugmaier, M. Klinger, M. Goebeler, S.
     439–446.                                                                            Knop, R. Noppeney, A. Viardot, G. Hess, M. Schuler, H. Einsele, C.
[28] B. Kelley, Industralization of mAb production technology. The                       Brandl, A. Wolf, P. Kirchinger, P. Klappers,
     bioprocess industry at a crossroads, mAbs 1 (2009) 443–452.                         M. Schmidt, G. Riethmüller, C. Reinhardt, P.A. Baeuerle,
[29] M. Better, C.P. Chang, R.R. Robinson, A.H. Horwitz, Escherichia coli                P. Kufer, Tumor regression in cancer patients by very low
     secretion of an active chimeric antibody fragment, Science 240                      doses of a T cell-engaging antibody, Science 321 (2008) 974–977.
     (1988) 1041–1043.                                                            [53]   J.S. Marvin, Z. Zhu, Recombinant approaches to IgG-like bispecific
[30] A. Skerra, A. Plückthun, Assembly of a functional immunoglobulin                    antibodies, Acta Pharmacol. Sin. 26 (2005) 649–658.
     Fv fragment in Escherichia coli, Science 240 (1988) 1038–1041.               [54]   S.C. Alley, N.M. Okeley, P.D. Senter, Antibody-drug conjugates:
[31] P. Carter, R.F. Kelley, M.L. Rodrigues, B. Snedecor, M. Covarrubias, M.D.           targeted drug delivery for cancer, Curr. Opin. Chem. Biol. 14
     Velligan, W.L. Wong, A.M. Rowland, C.E. Kotts, M.E. Carver, M. Yang,                (2010) 529–537.
     J.H. Bourell, H.M. Shepard, D. Henner, High level Escherichia coli           [55]   P.J. Carter, P.D. Senter, Antibody-drug conjugates for cancer
     expression and production of a bivalent humanized antibody                          therapy, Cancer J. 14 (2008) 154–169.
     fragment, Biotechnology 10 (1992) 163–167.                                   [56]   A. Younes, N.L. Bartlett, J.P. Leonard, D.A. Kennedy, C.M. Lynch, E.L.
[32] A. Beck, T. Wurch, C. Bailly, N. Corvaia, Strategies and challenges                 Sievers, A. Forero-Torres, Brentuximab vedotin (SGN-35) for
     for the next generation of therapeutic antibodies, Nat. Rev.                        relapsed CD30-positive lymphomas, N. Engl. J. Med. 363 (2010)
     Immunol. 10 (2010) 345–352.                                                         1812–1821.
[33] P. Chames, M. Van Regenmortel, E. Weiss, D. Baty, Therapeutic                [57]   I. Niculescu-Duvaz, Trastuzumab emtansine, an antibody-drug
     antibodies: successes, limitations and hopes for the future, Br. J.                 conjugate for the treatment of HER2+ metastatic breast cancer,
     Pharmacol. 157 (2009) 220–233.                                                      Curr. Opin. Mol. Ther. 12 (2010) 350–360.
[34] A.C. Chan, P.J. Carter, Therapeutic antibodies for autoimmunity              [58]   V. Ghetie, S. Popov, J. Borvak, C. Radu, D. Matesoi, C. Medesan, R.J.
     and inflammation, Nat. Rev. Immunol. 10 (2010) 301–316.                             Ober, E.S. Ward, Increasing the serum persistence of an IgG fragment
[35] J.M. Reichert, Monoclonal antibodies as innovative therapeutics,                    by random mutagenesis, Nat. Biotechnol. 15 (1997) 637–640.
     Curr. Pharm. Biotechnol. 9 (2008) 423–430.                                   [59]   L.G. Presta, Molecular engineering and design of therapeutic
[36] A.L. Marschall, A. Frenzel, T. Schirrmann, M. Schüngel, S. Dübel,                   antibodies, Curr. Opin. Immunol. 20 (2008) 460–470.
     Targeting antibodies to the cytoplasm, mAbs 3 (2011) 3–16.                   [60]   J. Zalevsky, A.K. Chamberlain, H.M. Horton, S. Karki, I.W.L. Leung,
[37] L. Lampson, Monoclonal antibodies in neuro-oncology: getting                        T.J. Sproule, G.A. Lazar, D.C. Roopenian, J.R. Desjarlais, Enhanced
     past the blood-brain barrier, mAbs 3 (2011) 156–163.                                antibody half-life improves in vivo activity, Nat. Biotechnol. 28
[38] B.V. Zlokovic, The blood-brain barrier in health and chronic                        (2010) 157–159.
     neurodegenerative disorders, Neuron 57 (2008) 178–201.                       [61]   T. Igawa, S. Ishii, T. Tachibana, A. Maeda, Y. Higuchi, S. Shimaoka, C.
[39] H.-H. Sedalacek, G. Seemann, D. Hoffmann, J. Czech, P. Lorenz, C.                   Moriyama, T. Watanabe, R. Takubo, Y. Doi, T. Wakabayashi, A.
     Kolar, K. Bosslet, Antibodies as Carriers of Cytotoxicity, Karger,                  Hayasaka, S. Kadono, T. Miyazaki, K. Haraya, Y. Sekimori, T. Kojima,
     Munich, Germany, 1992.                                                              Y. Nabuchi, Y. Aso, Y. Kawabe, K. Hattori, Antibody recycling by
[40] M.M. Schmidt, K.D. Wittrup, A modeling analysis of the effects of                   engineered pH-dependent antigen binding improves the duration
     molecular size and binding affinity on tumor targeting, Mol.                        of antigen neutralization, Nat. Biotechnol. 28 (2010) 1203–1207.
     Cancer Ther. 8 (2009) 2861–2871.                                             [62]   T. Kubota, R. Niwa, M. Satoh, S. Akinaga, K. Shitara, N. Hanai,
[41] M.E. Ackerman, D. Pawlowski, K.D. Wittrup, Effect of antigen                        Engineered therapeutic antibodies with improved effector
     turnover rate and expression level on antibody penetration into                     functions, Cancer Sci. 100 (2009) 1566–1572.
     tumor spheroids, Mol. Cancer Ther. 7 (2008) 2233–2240.                       [63]   G. Cartron, H. Watier, J. Golay, P. Solal-Celigny, From the bench to
[42] H. Hurwitz, L. Fehrenbacher, W. Novotny, T. Cartwright, J.                          the bedside: ways to improve rituximab efficacy, Blood 104
     Hainsworth, W. Heim, J. Berlin, A. Baron, S. Griffing, E. Holmgren,                 (2004) 2635–2642.
     N. Ferrara, G. Fyfe, B. Rogers, R. Ross, F. Kabbinavar, Bevacizumab          [64]   H.K. Binz, P. Amstutz, A. Plückthun, Engineering novel binding
     plus irinotecan, fluorouracil, and leucovorin for metastatic                        proteins from nonimmunoglobulin domains, Nat. Biotechnol. 23
     colorectal cancer, N. Engl. J. Med. 350 (2004) 2335–2342.                           (2005) 1257–1268.
[43] D.J. Slamon, B. Leyland-Jones, S. Shak, H. Fuchs, V. Paton, A.               [65]   M. Gebauer, A. Skerra, Engineered protein scaffolds as
     Bajamonde, T. Fleming, W. Eiermann, J. Wolter, M. Pegram, J.                        next-generation antibody therapeutics, Curr. Opin. Chem. Biol. 13
     Baselga, L. Norton, Use of chemotherapy plus a monoclonal                           (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                                       1269


[66] S.D. Nuttall, R.B. Walsh, Display scaffolds: protein engineering                 albumin domain antibodies in the development of highly potent,
     for novel therapeutics, Curr. Opin. Pharmacol. 8 (2008)                          efficacious and long-acting interferon, Protein Eng. Des. Sel. 23
     609–615.                                                                         (2010) 271–278.
[67] B. Zuraw, U. Yasothan, P. Kirkpatrick, Ecallantide, Nat. Rev. Drug          [69] L.J. Holt, A. Basran, K. Jones, J. Chorlton, L.S. Jespers, N.D. Brewis, I.M.
     Discov. 9 (2010) 189–190.                                                        Tomlinson, Anti-serum albumin domain antibodies for extending
[68] A. Walker, G. Dunlevy, D. Rycroft, P. Topley, L.J. Holt, T. Herbert, M.          the half-lives of short lived drugs, Protein Eng. Des. Sel. 21 (2008)
     Davies, F. Cook, S. Holmes, L. Jespers, C. Herring, Anti-serum                   283–288.

More Related Content

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

Introduction to current and future protein therapeutics - a protein engineeri...
Introduction to current and future protein therapeutics - a protein engineeri...Introduction to current and future protein therapeutics - a protein engineeri...
Introduction to current and future protein therapeutics - a protein engineeri...National Institute of Biologics
 
Brown adipose tissue updates
Brown adipose tissue updatesBrown adipose tissue updates
Brown adipose tissue updatesAnahi Garcia
 
Cellular immunity and immunopathology in autoimmune addison’s disease
Cellular immunity and immunopathology in autoimmune addison’s diseaseCellular immunity and immunopathology in autoimmune addison’s disease
Cellular immunity and immunopathology in autoimmune addison’s diseaseAlifiani Rohma
 
Toxicology paper 2010
Toxicology paper 2010Toxicology paper 2010
Toxicology paper 2010Nasrin Vassel
 
Toxicology paper 2010
Toxicology paper 2010Toxicology paper 2010
Toxicology paper 2010Nasrin Vassel
 
Pharmacol Rev-2014 Ratner
Pharmacol Rev-2014 RatnerPharmacol Rev-2014 Ratner
Pharmacol Rev-2014 RatnerMarcia Ratner
 
Glicolisis y el cancer
Glicolisis y el cancerGlicolisis y el cancer
Glicolisis y el cancerGean Arroyo
 
Clinical aspects of cobalamin deficiency in elderly patients
Clinical aspects of cobalamin deficiency in elderly patientsClinical aspects of cobalamin deficiency in elderly patients
Clinical aspects of cobalamin deficiency in elderly patientsJosep Vidal-Alaball
 
Nutrition for the ageing brain: Towards evidence for an optimal diet
Nutrition for the ageing brain: Towards evidence for an optimal dietNutrition for the ageing brain: Towards evidence for an optimal diet
Nutrition for the ageing brain: Towards evidence for an optimal dietNutricia
 
Pbpk models for the prediction of in vivo performance of oral dosage forms
Pbpk models for the prediction of in vivo performance of oral dosage formsPbpk models for the prediction of in vivo performance of oral dosage forms
Pbpk models for the prediction of in vivo performance of oral dosage formsAlberto Palomino Huarhua
 
Poin of care testing
Poin of care testingPoin of care testing
Poin of care testingLAB IDEA
 
Platelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdf
Platelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdfPlatelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdf
Platelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdfroomno3
 
Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...
Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...
Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...Andrea Porter
 
Handbook of capsule endoscopy
Handbook of capsule endoscopyHandbook of capsule endoscopy
Handbook of capsule endoscopy兆涛 宫
 
2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdf2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdfJonh85
 
Papel de la Vitamina K en la prevención de los embolismos
Papel de la Vitamina K en la prevención de los embolismos Papel de la Vitamina K en la prevención de los embolismos
Papel de la Vitamina K en la prevención de los embolismos Hospital Guadix
 
Ultralow doses (history of one research)
Ultralow doses (history of one research)Ultralow doses (history of one research)
Ultralow doses (history of one research)xiu2k
 

Similar to Introduction to current and future protein therapeutics a protein engineering perspective (20)

Introduction to current and future protein therapeutics - a protein engineeri...
Introduction to current and future protein therapeutics - a protein engineeri...Introduction to current and future protein therapeutics - a protein engineeri...
Introduction to current and future protein therapeutics - a protein engineeri...
 
Brown adipose tissue updates
Brown adipose tissue updatesBrown adipose tissue updates
Brown adipose tissue updates
 
SCDB2010
SCDB2010SCDB2010
SCDB2010
 
Cellular immunity and immunopathology in autoimmune addison’s disease
Cellular immunity and immunopathology in autoimmune addison’s diseaseCellular immunity and immunopathology in autoimmune addison’s disease
Cellular immunity and immunopathology in autoimmune addison’s disease
 
Toxicology paper 2010
Toxicology paper 2010Toxicology paper 2010
Toxicology paper 2010
 
Toxicology paper 2010
Toxicology paper 2010Toxicology paper 2010
Toxicology paper 2010
 
Pharmacol Rev-2014 Ratner
Pharmacol Rev-2014 RatnerPharmacol Rev-2014 Ratner
Pharmacol Rev-2014 Ratner
 
Glicolisis y el cancer
Glicolisis y el cancerGlicolisis y el cancer
Glicolisis y el cancer
 
Clinical aspects of cobalamin deficiency in elderly patients
Clinical aspects of cobalamin deficiency in elderly patientsClinical aspects of cobalamin deficiency in elderly patients
Clinical aspects of cobalamin deficiency in elderly patients
 
Nutrition for the ageing brain: Towards evidence for an optimal diet
Nutrition for the ageing brain: Towards evidence for an optimal dietNutrition for the ageing brain: Towards evidence for an optimal diet
Nutrition for the ageing brain: Towards evidence for an optimal diet
 
Pbpk models for the prediction of in vivo performance of oral dosage forms
Pbpk models for the prediction of in vivo performance of oral dosage formsPbpk models for the prediction of in vivo performance of oral dosage forms
Pbpk models for the prediction of in vivo performance of oral dosage forms
 
Poin of care testing
Poin of care testingPoin of care testing
Poin of care testing
 
Wiley et al PRER
Wiley et al PRERWiley et al PRER
Wiley et al PRER
 
Platelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdf
Platelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdfPlatelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdf
Platelet-rich-fibrin--Its-role-in-periodont_2014_The-Saudi-Journal-for-Denta.pdf
 
LiteratureReviewFinal
LiteratureReviewFinalLiteratureReviewFinal
LiteratureReviewFinal
 
Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...
Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...
Author S Personal Copy Peptide-Based Biopolymers In Biomedicine And Biotechno...
 
Handbook of capsule endoscopy
Handbook of capsule endoscopyHandbook of capsule endoscopy
Handbook of capsule endoscopy
 
2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdf2013 - The impact of coffee on health.pdf
2013 - The impact of coffee on health.pdf
 
Papel de la Vitamina K en la prevención de los embolismos
Papel de la Vitamina K en la prevención de los embolismos Papel de la Vitamina K en la prevención de los embolismos
Papel de la Vitamina K en la prevención de los embolismos
 
Ultralow doses (history of one research)
Ultralow doses (history of one research)Ultralow doses (history of one research)
Ultralow doses (history of one research)
 

Recently uploaded

Separation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesSeparation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesFatimaKhan178732
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application ) Sakshi Ghasle
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdfssuser54595a
 
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Celine George
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️9953056974 Low Rate Call Girls In Saket, Delhi NCR
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...Marc Dusseiller Dusjagr
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)eniolaolutunde
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionSafetyChain Software
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfsanyamsingh5019
 
Crayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon ACrayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon AUnboundStockton
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityGeoBlogs
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxGaneshChakor2
 
Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Educationpboyjonauth
 
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...M56BOOKSTORE PRODUCT/SERVICE
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxRoyAbrique
 
The basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptxThe basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptxheathfieldcps1
 
Science 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its CharacteristicsScience 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its CharacteristicsKarinaGenton
 

Recently uploaded (20)

Separation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and ActinidesSeparation of Lanthanides/ Lanthanides and Actinides
Separation of Lanthanides/ Lanthanides and Actinides
 
Hybridoma Technology ( Production , Purification , and Application )
Hybridoma Technology  ( Production , Purification , and Application  ) Hybridoma Technology  ( Production , Purification , and Application  )
Hybridoma Technology ( Production , Purification , and Application )
 
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
18-04-UA_REPORT_MEDIALITERAСY_INDEX-DM_23-1-final-eng.pdf
 
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
Incoming and Outgoing Shipments in 1 STEP Using Odoo 17
 
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
call girls in Kamla Market (DELHI) 🔝 >༒9953330565🔝 genuine Escort Service 🔝✔️✔️
 
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Tilak Nagar Delhi reach out to us at 🔝9953056974🔝
 
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
“Oh GOSH! Reflecting on Hackteria's Collaborative Practices in a Global Do-It...
 
Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1Código Creativo y Arte de Software | Unidad 1
Código Creativo y Arte de Software | Unidad 1
 
Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)Software Engineering Methodologies (overview)
Software Engineering Methodologies (overview)
 
Mastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory InspectionMastering the Unannounced Regulatory Inspection
Mastering the Unannounced Regulatory Inspection
 
Sanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdfSanyam Choudhary Chemistry practical.pdf
Sanyam Choudhary Chemistry practical.pdf
 
Crayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon ACrayon Activity Handout For the Crayon A
Crayon Activity Handout For the Crayon A
 
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝Model Call Girl in Bikash Puri  Delhi reach out to us at 🔝9953056974🔝
Model Call Girl in Bikash Puri Delhi reach out to us at 🔝9953056974🔝
 
Paris 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activityParis 2024 Olympic Geographies - an activity
Paris 2024 Olympic Geographies - an activity
 
CARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptxCARE OF CHILD IN INCUBATOR..........pptx
CARE OF CHILD IN INCUBATOR..........pptx
 
Introduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher EducationIntroduction to ArtificiaI Intelligence in Higher Education
Introduction to ArtificiaI Intelligence in Higher Education
 
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
KSHARA STURA .pptx---KSHARA KARMA THERAPY (CAUSTIC THERAPY)————IMP.OF KSHARA ...
 
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptxContemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
Contemporary philippine arts from the regions_PPT_Module_12 [Autosaved] (1).pptx
 
The basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptxThe basics of sentences session 2pptx copy.pptx
The basics of sentences session 2pptx copy.pptx
 
Science 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its CharacteristicsScience 7 - LAND and SEA BREEZE and its Characteristics
Science 7 - LAND and SEA BREEZE and its Characteristics
 

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