Alternative non-antibody scaffolds for molecular recognition

https://doi.org/10.1016/j.copbio.2007.04.010Get rights and content

Originally proposed one decade ago, the idea of engineering proteins outside the immunoglobulin family for novel binding functions has evolved as a powerful technology. Several classes of protein scaffolds proved to yield reagents with specificities and affinities in a range that was previously considered unique to antibodies. Such engineered protein scaffolds are usually obtained by designing a random library with mutagenesis focused at a loop region or at an otherwise permissible surface area and by selection of variants against a given target via phage display or related techniques. Whereas a plethora of protein scaffolds has meanwhile been proposed, only few of them were actually demonstrated to yield specificities towards different kinds of targets and to offer practical benefits such as robustness, smaller size, and ease of expression that justify their use as a true alternative to conventional antibodies or their recombinant fragments. Currently, the most promising scaffolds with broader applicability are protein A, the lipocalins, a fibronectin domain, an ankyrin consensus repeat domain, and thioredoxin. Corresponding binding proteins are not only of interest as research reagents or for separation in biotechnology but also as potential biopharmaceuticals, especially in the areas of cancer, autoimmune and infectious diseases as well as for in vivo diagnostics. The medical prospects have boosted high commercial expectations, and many of the promising scaffolds are under development by biotech start-up companies. Although some issues still have to be addressed, for example immunogenicity, effector functions, and plasma half-life in the context of therapeutic use or low-cost high-throughput selection for applications in proteomics research, it has become clear that scaffold-derived binding proteins will play an increasing role in biotechnology and medicine.

Introduction

Until recently, antibodies have almost uniquely served as a natural biomolecular scaffold for various applications in basic science and medicine. Their structurally variable antigen-binding site has evolved in the immune system of higher vertebrates to provide an effective humoral response against almost all kinds of foreign substances and invading pathogens [1]. Apart from their pivotal role in vivo, natural as well as engineered antibodies have yielded valuable reagents for virtually any type of antigen, and at least 10 000 antibodies are commercially available [2]. In addition, more than 20 antibody-based products have been approved as biopharmaceuticals, mainly for the treatment of cancer and autoimmune diseases, and several hundred drug candidates are currently under clinical development [3].

However, with increasing application in research, biotechnology, and medical therapy, it has become obvious that antibodies suffer from some fundamental disadvantages. Immunoglobulins (Igs) are rather large molecules, thus limiting tissue penetration. They are made of two different polypeptides, the light and heavy chains, which leads to unstable domain association when dealing with small Fv fragments and also requires complicated cloning steps for recombinant expression. The complex architecture of their antigen-binding site, which is formed by six hypervariable loops, three from each chain, is difficult to manipulate simultaneously if synthetic libraries are to be generated. The constant Fc region mediates immunological effector functions that are only crucial for few biopharmaceutical applications and often lead to undesired interactions. The manufacture of glycosylated full-size antibodies requires expensive eukaryotic cell culture, whose optimization and fermentation is time-consuming and faces limited capacities [4]. Finally, the complex intellectual property situation hampers the generation and production of recombinant antibodies [5]. Consequently, there is an emerging need for antibody alternatives with improved features.

Section snippets

The ‘scaffold’ idea: evolution and consolidation

The idea of employing proteins that do not belong to the Ig superfamily for the generation of novel binding proteins arose from the notion that antibodies owe their universal antigen recognition function to the combination of a structurally conserved framework with a spatially defined combining site composed of peptide segments that are hypervariable both in sequence and conformation [1]. Since the extensive development of antibody engineering techniques in conjunction with methods for library

Structural classes of promising protein scaffolds

At present, the different types of protein scaffolds that have been successfully exploited for the construction of artificial binding proteins can be classified into the following groups (Table 1).

Reagents for biomedical research and biotechnology

The predominant applications for engineered binding proteins in bioanalytics and biotechnology are the detection and separation of ‘analytes’ or other kinds of functionally interesting target molecules. Engineered protein scaffolds are receiving increasing attention for the preparation of bioseparation affinity matrices [36]. Similarly as protein A has become a standard reagent for the purification of antibodies from animal sera or cell culture supernatants, Affibodies derived from this

Applications for medical therapy and in vivo diagnostics

Although generally considered highly promising [18], advanced clinical applications for biological drugs derived from protein scaffolds are still rare. So far, just two candidates, the two engineered Kunitz-type protease inhibitors DX-88 and DX-890, are in later stage clinical trials. However, many other potential biopharmaceuticals are currently under preclinical development, and a few are about to enter the clinics soon or are already in phase I study, for example an AdNectin antagonizing

Conclusions and prospects

Probably the best indication that a protein scaffold has gained practical relevance is its commercial exploitation, either by a biotech start-up business that is devoted to its development or by a larger pharmaceutical or chemical company that sponsors the technology. Indeed, this is the case for most of the protein scaffolds discussed in this review (Table 1). Currently, the value in this field is largely driven by the medical prospects of alternative binding proteins [18], which is also

References and recommended reading

Papers of particular interest, published within the annual period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

References (51)

  • S. Attucci et al.

    EPI-hNE4, a proteolysis-resistant inhibitor of human neutrophil elastase and potential anti-inflammatory drug for treating cystic fibrosis

    J Pharmacol Exp Ther

    (2006)
  • S. Krause et al.

    Grafting of thrombopoietin-mimetic peptides into cystine knot miniproteins yields high-affinity thrombopoietin antagonists and agonists

    FEBS J

    (2007)
  • J. Silverman et al.

    Multivalent avimer proteins evolved by exon shuffling of a family of human receptor domains

    Nat Biotechnol

    (2005)
  • M. Wiederstein et al.

    Protein sequence randomization: efficient estimation of protein stability using knowledge-based potentials

    J Mol Biol

    (2005)
  • D. Steiner et al.

    Signal sequences directing cotranslational translocation expand the range of proteins amenable to phage display

    Nat Biotechnol

    (2006)
  • I. Van Walle et al.

    Immunogenicity screening in protein drug development

    Expert Opin Biol Ther

    (2007)
  • G.A. Michaud et al.

    Analyzing antibody specificity with whole proteome microarrays

    Nat Biotechnol

    (2003)
  • P.J. Carter

    Potent antibody therapeutics by design

    Nat Rev Immunol

    (2006)
  • M. Fiedler et al.

    Non-antibody scaffolds

  • A. Skerra

    Engineered protein scaffolds for molecular recognition

    J Mol Recognit

    (2000)
  • A. Rothe et al.

    In vitro display technologies reveal novel biopharmaceutics

    FASEB J

    (2006)
  • H.K. Binz et al.

    Engineering novel binding proteins from nonimmunoglobulin domains

    Nat Biotechnol

    (2005)
  • H.K. Binz et al.

    Engineered proteins as specific binding reagents

    Curr Opin Biotechnol

    (2005)
  • R.J. Hosse et al.

    A new generation of protein display scaffolds for molecular recognition

    Protein Sci

    (2006)
  • Nygren PÅ et al.

    Binding proteins from alternative scaffolds

    J Immunol Meth

    (2004)
  • Cited by (262)

    • Review of phage display: A jack-of-all-trades and master of most biomolecule display

      2024, International Journal of Biological Macromolecules
    • Developing a computational pharmacokinetic model of systemic snakebite envenomation and antivenom treatment

      2022, Toxicon
      Citation Excerpt :

      In vitro selection averts the requirement of animal immunisation by capturing high affinity binders against specific toxins from large, hypervariable antibody libraries. The use of synthetic antibody libraries can also facilitate the application of alternative scaffolds with novel pharmacokinetic properties (Roncolato et al., 2015; Skerra, 2007). Nanobodies (12–15 kDa) and scFvs (27 kDa) are two alternative antibody fragment scaffolds which are being applied to antivenom development.

    View all citing articles on Scopus
    View full text