5,817
Views
67
CrossRef citations to date
0
Altmetric
Technology Evaluation

Shark variable new antigen receptor biologics – a novel technology platform for therapeutic drug development

, PhD, , PhD, , , PhD & , PhD

Abstract

Introduction: Biologics drugs have succeeded in achieving a commercial dominance in the global market for new therapies and large pharmaceutical companies' interest remains strong through a continued commitment to pipeline development. It is not surprising, therefore, that next-generation biologics, particularly antibody-like scaffolds that offer many of the advantages of the original biologic drugs but in simplified formats, have entered the clinic as competing substitute therapeutic products, to capture market share.

Areas covered: Specifically, this paper will position shark-derived variable new antigen receptors (VNARs) within an overview of the existing biologics landscape including the growth, diversity and success to date of alternative scaffolds. The intention is not to provide a comprehensive review of biologics as a whole but to discuss the main competing single-domain technologies and the exciting therapeutic potential of VNAR domains as clinical candidates within this context.

Expert opinion: The inherent ability to specifically bind target and intervene in disease-related biological processes, while reducing off-site toxicity, makes mAbs an effective, potent and now proven class of therapeutics. There are, however, limitations to these ‘magic bullets’. Their size and complexity can restrict their utility in certain diseases types and disease locations. In contrast, a number of so-called alternative scaffolds, derived from both immunoglobulin- and non-immunoglobulin-based sources have been developed with real potential to overcome many of the shortcomings documented for mAb treatments. Unlike competing approaches such as Darpins and Affibodies, we now know that shark VNAR domains (like camel VHH nanobody domains), are an integral part of the adaptive immune system of these animals and have evolved naturally (but from very different starting molecules) to exhibit high affinity and selectivity for target. In addition, and again influenced by the environment in which they have evolved naturally, their small size, simple architecture, high solubility and stability, deliver additional flexibility compared to classical antibodies (and many non-natural alternative scaffolds), thereby providing an attractive basis for particular clinical indications where these attributes may offer advantages.

1. Overview of the market

Since the 1970s, advances in recombinant DNA, protein engineering and phage display technologies Citation[1-10], coupled with a greater understanding of disease processes, have resulted in the development and production of the growing number of biopharmaceuticals or biologics currently used by clinicians in their assessment and treatment of patients. According to Leader et al. Citation[11], biologics can be divided into four main categories: i) replacement protein therapies with enzymatic or regulatory activity such as insulin; ii) specific targeting protein therapeutics including antibodies and alternative binding domains; iii) protein-based vaccines; and iv) protein-based diagnostics. Recent years have seen the biologics market go from strength to strength to a position currently where they dominate the top 10, billion dollar revenue generators or so-called blockbuster drugs. This growth is predicted to continue from a market value of $150.1 billion in 2011 to $251.8 billion by 2017, representing a compound annual growth factor (CAGR) of 9% Citation[12]. Worldwide spending on new drug development reflects this strong growth with reported drug development expenditure in 2012 to be in excess of $80 billion with spending on biopharmaceutical research accounting for around 50% of this total budget. The USA currently represents the greatest market segment for biologics, due in part to the high price for this drug class; however, this is likely to change over the next 5 years or so with developing countries such as China, Brazil, India and Russia gaining an increased importance in the global marketplace Citation[13].

2. Existing technologies

The past 10 years have been tough for many drug discovery and development companies. There has been a paucity of new small molecule entities entering the clinic and an urgent need to expand and identify new disease targets. In this climate the attractiveness of complex, multi-domain drug candidates which show shorter drug development timelines and are challenging to copy have provided a rare opportunity to many companies and this in turn has driven the recorded increase in biologic resource and development activity. Within the realm of biologics, mAbs are currently the king. In 2011, they represented close to 32% of overall revenue (∼ $48 billion). This is expected to grow further to a staggering $90 billion by 2017, giving an estimated CAGR of 11.8%. As of February 2013, 39 mAbs had been approved for therapeutic use and over 350 were in clinical development Citation[12,14].

Inherent advantages of mAbs over small molecules include high levels of specificity for target, reduced off-site toxicity and the efficacy benefits of induced cell death and long serum half-life, both achieved through the Fc domain. These latter attributes can be altered by molecular engineering to further tailor potency and pharmacokinetic properties, building flexibility into the design of mAbs, depending on the chosen application of the drug Citation[15-20].

Cancer and chronic inflammatory disease are currently the primary indications for antibody therapy due in part to the systemic accessibility of the target antigens. Of the 28 USA and Europe market-approved mAbs reported by Reichert in 2012 Citation[14], less than half recognised unique targets, with four targeting TNF-α and CD20 and two each targeting EGFR and VEGF. Whereas whole mAbs still dominate the list of clinically approved therapeutics, smaller, antibody fragments have been developed for both in vivo imaging and therapy. In particular, certolizumab pegol (Cimzia), which is a humanised anti-TNF-α Fab’ fragment conjugated to a PEG moiety, of ∼ 40 kDa, has shown good efficacy against rheumatoid arthritis and Crohn’s disease Citation[14,21-25].

Although the clinical successes of mAbs and buoyant market growth have resulted in a significant shift of investment towards biologic pipeline development, there remain biological limitations and economic barriers to overcome for many forms of antibody therapy Citation[26-28]. As small molecules are typically orally available, they can be readily prescribed and self-administered at home by the patient. This is simply unachievable for mAb therapy which currently has to be administered by intravenous infusion or subcutaneous injection often in a clinical setting by a medical professional. Although the frequency of dosing is greatly reduced compared to that of small molecules, the inconvenience, discomfort and cost of treatment has considerable impact on both the patient and the healthcare provider. Another limiting factor is the cost of manufacturing complex, large globular glycoproteins which requires considerable investment in good manufacturing practice level facilities, optimised eukaryotic expression systems and standardised purification protocols, to produce a consistent supply of clinical grade material. At 150 kDa, the size of mAbs also dictates their site of action within the human body, limiting this to systemically accessible targets or cell surface molecules with minimal tissue penetration. These considerations and perceived limitations has fuelled a continuous drive in the field of immunotherapeutic drug discovery to develop smaller, stable binding molecules that retain the specificity of antibodies but overcome some of the recognised cost and biological challenges. Examples of these antibody-based fragments and novel scaffolds have been the subject of many comprehensive reviews Citation[29-34] and as such this paper will only focus on a handful that have progressed towards the clinic and have similar attributes to variable new antigen receptor (VNAR) domains.

3. How the technology works

To replace current gold standard therapies, next-generation biologics not only have to be as good as antibodies but also need to demonstrate additional features that differentiate them in this highly competitive landscape. Decreased size to enhance tissue penetration and reduce immunogenicity, increased solubility to assist formulation, simple molecular architecture that facilitates multiple re-formatting options and promotes greater yields for cost-effective manufacturing are just some examples of additional desirable properties. One candidate biologic and the subject of this technology review is the immunoglobulin (Ig)-based VNAR derived from the Ig new antigen receptor (IgNAR) found in cartilaginous fish Citation[35,36]. IgNAR is known to play a role in the adaptive immune system of these animals and shares structural homology with cell adhesion molecules, antibody variable light (VL) chains and T-cell receptors, indicating a possible common ancestral receptor molecule that has evolved and been recruited by the immune system to function as antibody-like structures Citation[35,37-39]. The secreted form of the IgNAR molecule is a heavy-chain protein dimer, each polypeptide comprising five constant domains and a single variable domain, with no associated light chain Citation[35,36]. The variable domain of IgNAR or VNAR defines the specificity of the molecule and can be classified into different types based on the position and number of non-canonical cysteine resides within the hypervariable (HV) and framework (FW) regions Citation[40,41]. To date, three main categories of IgNAR variable domains (VNARs) have been defined in the literature, as isotypes I, II and III. Interestingly, the type I VNAR has only been identified in nurse sharks and may be unique to this species Citation[37]. This isotype possesses two cysteine residues in FWs 2 and 4, and two paired cysteines in CDR3. From analysis of the crystal structure of a type I VNAR in complex with lysozyme, it was determined that germline cysteine residues present in FWs 2 and 4 are required to form disulphide bridges with those in CDR3, thereby creating a tightly packed structure within which the CDR3 loop is constrained towards the HV2 region () Citation[42].

Figure 1. Illustration of VNAR isotypes showing the similarities and differences between different isotypes of VNAR domain.

Figure 1. Illustration of VNAR isotypes showing the similarities and differences between different isotypes of VNAR domain.

Type II VNAR domains possess the characteristic protruding CDR3, created by intra-molecular disulphide bonds formed between cysteine residues in CDR1 and CDR3 Citation[36,37,43]. Type II sequences typically have shorter CDR3s than type I with an average of 15 and 21 residues, respectively.

The remaining class of VNAR described in the literature is type III, the expression of which is confined to young sharks (< 1 year old) Citation[44]. Type III VNARs have highest similarity to the type II VNARs with the addition of a conserved tryptophan residue in CDR1. The D regions of shark genes, which form CDR3 and give rise to such diversity in types I and II VNARs, are fused with the V-gene in type III neonates and results in short (∼ 15 residues) limited CDR3s lacking in sequence diversity. It is hypothesised that type III VNARs are a first line of defence to protect newborn pups against a common pathogen or may play a role in the development of the adult immune system. This isotype is then quickly overtaken by the more mature classes of VNAR which provide a more expansive immune repertoire Citation[44]. In addition to types I, II and III, there are also variants that contain only the highly conserved canonical cysteine residues at positions 35 and 107 that act to hold the Ig fold and lack any other disulphide bridges throughout the molecule. Liu et al. Citation[45] and Streltsov et al. Citation[37] referred to these as type IIb, a nomenclature that we have also adopted to assign these domains to different isotypes. An expansion of this designation has also been employed to describe a very similar domain to the type IIb which lacks the non-canonical cysteine residues but retains an invariant tryptophan in CDR1, like the type III isotype – this has therefore been called type IIIb. All currently named isotypes are shown in . Unlike conventional antibodies, VNARs have a severely truncated CDR2 region Citation[42]. To compensate for this, diversity is achieved by long variable protruding CDR3s, with additional diversity evident in CDR1 and also through two HV regions (HV2 and 4), unique to this class of molecule. The benefit of a structurally complex CDR3 combined with supporting intermolecular disulphide bridging is that VNARs form more compact structures with unusual paratopes which can access cryptic epitopes such as pockets or grooves within the target antigen Citation[42,46-48]. In addition to this, the deleted CDR2 makes VNARs the smallest naturally occurring antigen-specific binding domains in the vertebrate kingdom with a molecular mass of ∼ 12 kDa Citation[42]. A comparison of relative sizes of antigen-binding domains currently being developed for therapeutic applications is shown in .

Figure 2. Relative size of Ig and Ig-like biologics is shown. Molecular models depicting the relative size of four different antibody-based scaffolds currently under clinical development are shown. VNAR and VHH are illustrated in two potential therapeutic formats. In general, the VNAR protein is ∼ 20% smaller than the VHH equivalent. The authors wish to thank Springer Science and Business Media, LLC and Landes Bioscience for their kind permission for inclusion of this modified figure from Barelle et al., 2009 Citation[41].

Figure 2. Relative size of Ig and Ig-like biologics is shown. Molecular models depicting the relative size of four different antibody-based scaffolds currently under clinical development are shown. VNAR and VHH are illustrated in two potential therapeutic formats. In general, the VNAR protein is ∼ 20% smaller than the VHH equivalent. The authors wish to thank Springer Science and Business Media, LLC and Landes Bioscience for their kind permission for inclusion of this modified figure from Barelle et al., 2009 Citation[41].

4. VNARs as therapeutic candidates

Naturally designed to bind antigen with high affinity and selectivity, VNARs are inherently good candidates for therapeutic development Citation[37,49-52]. Functioning naturally in the harsh environment of shark sera that consists of 350 mM urea and 1000 mOsmol, they exhibit tremendous stability and solubility Citation[40,53,54]. Their simple single chain structures afford remarkable refolding properties after exposure to high temperatures (> 80°C), chemical and enzymic degradation Citation[49,53-55]. Perhaps the greatest differentiating attributes from classical antibodies are their small size and unique topologies, opening up opportunities to penetrate tissues and bind to novel, cryptic targets intractable to existing larger more globular binding domains.

4.1 Drug discovery

Identifying a naturally occurring antibody-like scaffold with the potential for clinical development is the end point of a process that could now be considered validated and de-risked. The VNAR immune platform and drug pipelines now boast: robust means of isolating therapeutically relevant VNARs and selecting those with the desired binding affinity, the ability to engineer and adapt the biophysical properties of lead clones so they are fit for purpose and the means of high expression and purification in both eukaryotic and prokaryotic systems. Two platforms are available for the isolation of VNARs. The first is based on the role of IgNAR as part of the adaptive immune system of sharks. It has been shown now, in at least three different species of shark, that an IgNAR response can be elicited in response to antigen challenge Citation[49,51,56]. As VNAR domains are amenable to phage display, a simple blood sample can be taken from these animals after an iterative process of immunisation followed by boosts, RNA extracted and the VNAR repertoire amplified from cDNA generated from this total message Citation[57,58]. From this point cloning into a standard phagemid vector and selecting against target should provide positive hits. A second and complimentary means of isolating VNAR domains is the construction of a naïve or semi-synthetic phage display library based on naturally occurring VNAR domains that often include significant additional diversity through the engineering of the CDR regions Citation[45-48,55,59-64]. Both these methods have pros and cons. The evolutionarily distant position of the shark immune system encourages a good response to many mammalian proteins. Therefore, immunisation typically provides high-affinity domains directed against antigen following an in vivo maturation processes. This process is a little slower than the related process seen in mammals as it can take ∼ 4 – 6 months to achieve the desired response. Selection from a synthetic library shortens this time frame, however the level of success (specificity and affinity) is dependent on the quality and size of the library being screened and generally will deliver domains of lower affinity that may require further refinement through in vitro maturation. Both methods, however, have been used successfully to isolate a number of VNAR domains against multiple target classes as illustrated in .

Table 1. Published VNAR domains isolated by selection against specific targets.

Controlled mutagenesis through ribosome display has enabled the structure of a model type I VNAR to be interrogated and assessed based on retention of binding function Citation[65]. This study highlighted some key hallmark residues within VNAR isotypes across different species and has illustrated the tolerance of these domains for alterations within the sequence and retention of function, informing possible sites for conjugation and humanisation. A recent publication by Kovalenko et al. Citation[66], focussing on an anti-human serum albumin (HSA) VNAR domain isolated from an immunised dogfish Citation[51] showed that humanisation is achievable without loss of function. The question of the need to humanise has yet to be answered in vivo but the humanised versions of these VNAR domains show negligible antigenicity in human dendritic cell assays (personal communication). Their small, compact structure and lack of aggregation when expressed suggest that this may not be as significant an issue as one might expect for a shark-derived protein. This position was further supported by a study in non-human primates designed to address the serum half-life of the anti-HSA VNAR mentioned above. Müller et al. Citation[51] found no evidence of an immune response after two administrations (intravenous followed by subcutaneous) of the VNAR fusion protein. It is not always easy to predict but the key question of immunogenicity will also be affected by the target, the route of administration, serum half-life and the number of doses required to treat the patient. Ultimately, the true test of immunogenicity against a particular biologic in the patient population will only become apparent during clinical trials.

4.2 Drug development

The single chain composition of VNAR domains makes any molecular alteration relatively simple as the binding domain can be expressed as a single cassette without the requirement for two chains to associate and fold correctly. Dimer and trimer constructs can be expressed and purified and still exhibit functionality Citation[51]. They are also amenable to re-formatting as Fc if the cell killing function and half-life extension is required Citation[66].

Size, affinity and tissue penetration are correlated and in a cancer tissue penetration model, provided a high-enough affinity is achieved, the advantages of a smaller protein-binding domain will be realised Citation[67]. This profile perfectly matches that of the VNAR making it a highly plausible candidate technology for solid tumour therapies. Being small, however, does usually mean that the protein will have a short serum half-life. Typically, any molecule below the renal filtration cut-off of ∼ 60 kDa will be cleared rapidly from the body Citation[68]. This has value if the domain is designed to be an in vivo imaging agent where signal–to-noise ratios determine assay sensitivity but can be potentially limiting for the development of an efficacious drug. There are multiple ways to improve the pharmacokinetic profile of small proteins or peptides Citation[69-80]. Fusion to an antibody Fc domain, as mentioned previously, is one way Citation[81]; however, if being small is advantageous, then adding multiple extra domains (Fc) counteracts this size benefit. To circumvent this problem, the team at the University of Aberdeen have developed a VNAR-based half-life extension tool that increases the serum circulating time of a fusion partner from hours to weeks Citation[51]. The domain, known as E06, was isolated from a spiny dogfish immunised with HSA and has exhibited efficacy across three different models of pharmacokinetics (PK) (mouse, rat and monkey). Tolerating both N- and C-terminal fusions, this opens up the opportunities to re-format not only VNARs but also scFvs and peptides as dimers or trimers, increasing affinity for target through avidity or increasing utility through bi- or tri-functionality. Development of this PK bio-tool increases the repertoire of re-formatting options open to VNAR biologics, correlating optimal design with clinical application as shown in .

Figure 3. Schematic representation of VNAR formats showing different re-formatting options for VNAR domains.

Figure 3. Schematic representation of VNAR formats showing different re-formatting options for VNAR domains.

Tailoring PK is a highly effective way of increasing the therapeutic window of short-lived molecules, decreasing doses and reducing the frequency of administrations. This provides a huge benefit to patients who currently have to undergo invasive transfusions of drugs resulting in both discomfort and increased risk of complications and infection. Taking the combined PK data for E06 in fusion with a partner VNAR, and applying allometric extrapolation, the predicted half-life of this molecule in humans is ∼ 19 days (). This value correlates extremely well with that measured for the half-life of albumin itself validating the potential use of E06 as an effective bio-tool to improve the PK of partner therapeutics Citation[51].

Figure 4. Allometric scaling of the anti-albumin VNAR domain, E06 is shown. Comparison of the published half-life of albumin in mouse, rat and cynomolgus monkey (⧫) and that of 2V-E06 (▪) from the data published in Müller et al., 2012 Citation[51] is depicted.

Figure 4. Allometric scaling of the anti-albumin VNAR domain, E06 is shown. Comparison of the published half-life of albumin in mouse, rat and cynomolgus monkey (⧫) and that of 2V-E06 (▪) from the data published in Müller et al., 2012 Citation[51] is depicted.

5. Alternative technologies

As we have already reported, the overwhelming success of antibodies as therapeutic drugs has ignited a great deal of interest in the biopharmaceutical sector. The goal of replicating an antibody’s exquisite specificity for target while bringing added value through a differentiating quality such as small size has been central to the recent progress seen for several alternative platforms. Many promising antibody-based binding domains and alternative scaffolds have resulted from this recent intense research activity. The critique of novel scaffolds that follows is by no means exhaustive but focuses on those entering or in clinical trials or with an approved product already on the market. specifically collates the current information available about next-generation biologics in clinical trials.

Table 2. Next-generation scaffolds currently in the clinic.

5.1 Antibody-based biologics

As discussed, whole mAb sales currently dominate and represent ∼ 75% of the total biologics market in the USA and Europe Citation[14]. Of this 75%, the largest grouping is humanised mAbs (36%), with their origins firmly linked to monoclonal production via hybridomas. However, almost the same number are now fully human (32%) indicating a shift in technology preference and the use of approaches that enable the selection of human binding domains from phage display libraries or the innovative creation of genetically modified mice that express ‘human’ Igs in response to antigen challenge.

Antibody fragments such as Fabs, scFvs and Fc fusions have been developed for clinical use and have been comprehensively described in a recent book by William and Lila Strohl in addition to a number of review papers Citation[82-86]. For the purposes of this technical review, we will concentrate on camelid nanobodies and the single human domains known as dAbs. We consider these to be the most important antibody-based competitors to the VNAR approach. Discovered in the early 1990s, the naturally occurring single-domain antibodies in Camelidae (camels, dromedaries and llamas) known as VHHs or nanobodies were shown to form part of the adaptive immune system in these mammals Citation[87]. The ability to raise antigen-specific nanobodies through immunisation led to the isolation of domains against multiple targets and the launch of a Belgium-based company, Ablynx that successfully developed these domains into drug candidates. Showing similarities to VNARs in terms of their structure and properties, these single heavy-chain binding domains exist naturally and exhibit high affinity for target. However, in sharp contrast to VNARs, VHHs have evolved directly from an IgG lineage and at some point they have lost their partner VL domain and have undergone a truncation of the CH1 region resulting in the remaining VH variable domain being fused directly to the hinge region. Isolated VHH domains have been the subject of extensive research for almost 20 years (VNARs around 10 years) and there are currently seven nanobodies in clinical trials () with more than 25 programs in company pipelines across six major therapeutic areas, including cardiovascular, infectious disease, musculoskeletal, oncology, respiratory and immunology.

Single-domain human antibodies or dAbs were the basis of the technology platform developed by Domantis which was subsequently acquired by GlaxoSmithKline (GSK) in 2006. The technology comprises large synthetic phage display libraries built from isolated heavy or light human variable domains into which significant diversity was introduced through randomised engineering of the two CDR regions Citation[88]. Three reported dAbs are currently in the clinic (). One interesting programme is the fusion of an anti-HSA dAb (GSK2374697) to GLP-1 for the possible treatment of diabetes. The inclusion of an anti-HSA dAb as part of this drug formulation has validated the approach of ‘piggy-backing’ on the long-lived serum protein as a method of increasing the half-life of biologically active but short-lived peptides in the body Citation[89,90].

Just as fully human mAb can be derived from immunisation of an engineered mouse, so also a second platform developed by Crescendo Biologics Ltd allows for the isolation of fully human single domains from a transgenic mouse strain. This approach used as a starting point a ‘triple knockout’ (TKO) mouse where all three murine Ig loci were functionally silenced. TKO mice were then crossed with mice containing a yeast artificial chromosome construct that included both human heavy chain genes and murine constant genes. Following immunisation, these animals produce human heavy chain-only antibodies fused to a mouse Fc region. Initial success with the “Crescendo Mouse” was announced in January 2013, and the company is currently developing their own early stage pipeline of therapeutic products.

5.2 Novel scaffolds

In the past 20 years, around 30 different engineered binding proteins have been proposed as potential FWs for therapeutic drug development Citation[29,30,33,91-93]. The generation of engineered protein scaffolds generally begins with a natural protein structure that is used as a scaffold for modification. Typically, a core molecular FW provides the structure and stability onto which a diverse library of flexible loops or exposed residues can be introduced to create a synthetic library of binding proteins analogues to the variable domains found in antibodies. When compared to classical antibodies, these novel binding protein repertoires can offer a number of potential advantages. With the more obvious advantages of a small size (penetration, expression, re-formatting), the synthetic approach allows for the selection of a core scaffold that is chosen because it possess desirable characteristics such as low immunogenicity, expressability, solubility, thermal stability and/or protease resistance. Although many remain in the earlier, exploratory stages of platform technology optimisation, the exploitation of engineered protein scaffolds is proving to be an attractive route for binding molecules suitable for therapeutic development. Approaches, such as Affibodies, DARPins, Kunitz domains, Adnectins and Anticalins have progressed towards clinical candidates, either for therapy or for in vivo diagnostic or imaging applications.

Kunitz-type inhibitors are the earliest example of successful modifications to pre-existing binding molecules to deliver a library of related binding structures. They are robust scaffolds of human origin, based on a small engineered disulphide-bridged serine protease inhibitor of 60 amino acids in length with a molecular mass of ∼ 7 kDa. Diverse binding can be achieved through the engineering of an exposed peptide loop. Kalbitor (Escallantide) became the first approved domain in this class in 2009 for the treatment of acute attacks of hereditary angiooedema Citation[94]. A second drug candidate DX-890 (Depelestat, EPI-hNE4) is an inhibitor of human neutrophil elastase and is currently being assessed for the treatment of patients with impaired lung function Citation[95-97].

Libraries of Affibody-binding sites can be created by the mutagenesis of 13 amino acid positions exposed on the surface of α-helixes 1 and 2 in the Z-domain of protein A from Staphylococcus aureus. Although binders to a number of targets have been successfully isolated and characterised, one potential disadvantage of this class of protein scaffold is their bacterial origin which may increase the risk of immunogenicity after repeated therapeutic administration to patients. However, their small size (6.5 kDa) offers wide applicability for in vivo diagnostic imaging, where rapid distribution and elimination are desirable features of a disease-specific biologic Citation[98]. To date, the only Affibody enrolled in a clinical study is ABY-025 for PET imaging of HER2 expression in metastatic breast cancer.

DARPins are derived from natural ankyrin proteins and consist of at least three or more repeat motifs which generate a series of binding site loops at a large cysteine-free surface Citation[34,92]. This structural arrangement encourages the construction of diverse DARPin repertoires and the selection from these libraries of DARPins with picomolar affinities for their target proteins. They have a molecular size range of 14 – 20 kDa (four- or five-repeat DARPins units). Molecular Partners has developed a portfolio of drug candidates based on DARPins for ophthalmology, oncology and inflammation. DARPin MP0112 is an anti-angiogenic drug that has now been used clinically to treat wet age-related macular degeneration and more recently diabetic macular oedema. In both cases, MP0112 reduces VEGF-A concentrations in the aqueous humour of the eye after intraocular injection and appears to stabilise visual acuity and reduce retinal oedema.

Adnectins are based on a fibronectin type III backbone with molecular mass of 10 kDa. They are structurally very similar to antibody variable domains, but without a core disulphide bond Citation[99]. CT-322 (Angiocept) is the first engineered adnectin developed by Adnexus/Bristol-Myers Squibb and is an antagonist of VEGFR-2. CT-322 is currently in Phase II clinical trials for treatment of glioblastoma having completed Phase I studies without any significant adverse effects and with promising anti-tumour effects seen in some patients Citation[100,101].

Anticalins® are proteins derived from human lipocalins which naturally bind, store and transport a diverse range of molecules in the body Citation[102]. Recombinant Anticalins are capable of binding both proteins and haptens. Structurally they are barrel-shaped domains which are formed by eight anti-parallel β-strands connected by flexible loops and an attached α-helix. Diversity is engineered into the loop regions giving a repertoire of molecules from which those with the desired binding properties can be selected. Anticalin libraries have been constructed by Pieris, and clinical Phase I testing of Angiocal (PRS-050), an anti-VEGF PEGylated protein of 40 kDa, has been successfully completed in 26 patients with advanced solid tumours. PRS-050 was safe and well tolerated and had a half-life of 6 days without immunogenicity.

6. Conclusion

Biologic drug development is an exciting and dynamic field where innovation is driving the next wave of new products. Antibodies are big players in this industry and exemplify perfectly how nature can be harnessed and exploited to benefit humans. We are proposing that another naturally occurring class of molecules, shark VNAR domains, can become the next ‘small’ big thing. VNARs are perfectly designed and adapted to fit the requirements of drug discovery development and exhibit a range of significant additional attributes to give confidence that they too will become a valuable platform for multiple medical treatments.

7. Expert opinion

Antibody-based biologics have gone from strength to strength, demonstrated by their current and growing dominance in the therapeutic drug market. Such success breeds opportunity in the development of the next generation of these molecules to enhance their clinical impact and increase their economic value. A common thread across these new biological domains is the desire to retain affinity and selectivity for target while decreasing the size and complexity. There appears to be two main categories for these new molecules: naturally occurring antibody or antibody-like domains and alternative scaffolds that have been re-engineered to adopt the features of their natural counterparts.

Taking a current snapshot of these new biologic domains shows quite clearly that the nanobodies have progressed significantly with seven candidates presently in the clinic. Perhaps one aspect of this success can be attributed to the timeframe since these single-domain antibodies were discovered; however, it is clear from the breadth of the utility and application of these domains that a key strength is that they naturally exhibit the attributes required for drug development with additional benefits such as their size providing added value for many indications. Although this review has focussed on the therapeutic arm of development, these single-domain antibodies have also shown utility as in vivo imaging agents and in vitro diagnostics and are, therefore, excellent candidates for stratified medicine in the form of companion diagnostics Citation[103,104].

It is the opinion of the authors that this broad utility and success can be translated directly from the camel VHH to the shark VNAR platform. In fact, the nanobodies by trailblazing a path into patients have signposted a rapid route for shark VNAR into clinical studies.

Correlations can be made regarding the natural role of shark VNARs and nanobodies as components of the adaptive immune systems in their relative hosts. It is fascinating that convergent evolution has resulted in two very different species developing a single chain binding domain repertoire to protect against external challenge. This is a huge advantage when it comes to desirable attributes in a therapeutic drug platform as the domains can be raised through immunisation resulting in in vivo maturation, with high affinity, highly selective binders. An important additional benefit of immunisation in the shark system is that these animals are evolutionary distinct from mammals (∼ 450 million years) providing a robust platform for raising a response against proteins that are highly conserved between warm-blooded animals.

A greater understanding of the natural repertoire of multiple shark species coupled with in vitro characterisation has resulted in the ability to construct and screen semi-synthetic VNAR phage display libraries. In contrast to the VHH approach, the increased number of isotypes present in different shark species has allowed the construction of more structurally and clonally diverse ‘naïve’ libraries from which single domain nanomolar affinity antibodies, against varied targets, have been quickly isolated. As such, VNAR domains can be isolated from a series of complementary platforms providing a route to additional diversity and candidate generation.

Size is another key attribute of these shark single domains. As discussed previously, there is a desire to retain the affinity and specificity of whole antibodies but capturing this within the smallest binding domain possible. VNARs are the smallest naturally occurring binding domains in the vertebrate kingdom (up to 20% smaller than VHH) and, therefore, have achieved this goal without the need for re-engineering. They possess four regions of high variability per domain (VHH has three) and, therefore, have at their disposal the greatest density of binding loops by size of any competing technology. When this is combined with the natural propensity of multiple different structural families to target these loops into crevices and pockets (as well as additional more flexible structures that demonstrate induced fit) Citation[42,43], the potential of the VNAR technology as a potent novel drug discovery ‘engine’ is clear.

Article highlights.

  • Biologics are dominating the therapeutic drug market with mAbs showing the greatest commercial impact and most sustainable market growth.

  • There has been a shift in the development of therapeutic antibodies towards smaller, more stable, more flexible formats to increase their efficacy and broaden their clinical utility.

  • There are a growing number of next-generation antibody-like and novel scaffolds entering biotechnology and biopharmaceutical company pipelines designed to address some of the limitations of conventional antibodies and open up new opportunities for novel disease targeting and alternative modes of administration.

  • Shark single domains (variable new antigen receptors) have multiple natural and downstream attributes such as small size, high affinity, selectivity for target and ease of production that makes them attractive candidates for drug development.

Declaration of interest

The authors were supported by Scottish Enterprise and the Biotechnology and Biological Sciences Research Council (BBSRC). The authors have no other relevant affiliations or financial involvement with any organisation or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

Notes

This box summarizes key points contained in the article.

Bibliography

  • Cohen SN, Chang AC, Boyer HW, Helling RB. Construction of biologically functional bacterial plasmids in vitro. Proc Natl Acad Sci USA 1973;70:3240-4
  • Kohler G, Milstein C. Continuous cultures of fused cells secreting antibody of predefined specificity. Nature 1975;256:495-7
  • Wallace RB, Schold M, Johnson MJ, et al. Oligonucleotide directed mutagenesis of the human beta-globin gene: a general method for producing specific point mutations in cloned DNA. Nucleic Acids Res 1981;9:3647-56
  • Smith GP. Filamentous fusion phage: novel expression vectors that display cloned antigens on the virion surface. Science 1985;228:1315-17
  • McCafferty J, Griffiths AD, Winter G, Chiswell DJ. Phage antibodies: filamentous phage displaying antibody variable domains. Nature 1990;348:552-4
  • Bass S, Greene R, Wells JA. Hormone phage: an enrichment method for variant proteins with altered binding properties. Proteins 1990;8:309-14
  • Choo Y, Klug A. Toward a code for the interactions of zinc fingers with DNA: selection of randomized fingers displayed on phage. Proc Natl Acad Sci USA 1994;91:11163-7
  • Hiipakka M, Poikonen K, Saksela K. SH3 domains with high affinity and engineered ligand specificities targeted to HIV-1 nef. J Mol Biol 1999;293:1097-106
  • Christ D, Winter G. Identification of protein domains by shotgun proteolysis. J Mol Biol 2006;358:364-71
  • Hwang WY, Foote J. Immunogenicity of engineered antibodies. Methods 2005;36:3-10
  • Leader B, Baca QJ, Golan DE. Protein therapeutics: a summary and pharmacological classification. Nat Rev Drug Discov 2008;7:21-39
  • Highsmith J. Biologic therapeutic drugs: technologies and global markets. 2013; BIO079B, BCC Research, Wellesley, MA 02481, USA
  • Bosklopper E. Antibody drugs: technologies and global markets. 2012; BIO016H, BCC Research, Wellesley, MA 02481, USA
  • Reichert JM. Marketed therapeutic antibodies compendium. MAbs 2012;4:413-15
  • Dall’Acqua WF, Cook KE, Damschroder MM, et al. Modulation of the effector functions of a human IgG1 through engineering of its hinge region. J Immunol 2006;177:1129-38
  • Dall’Acqua WF, Kiener PA, Wu H. Properties of human IgG1s engineered for enhanced binding to the neonatal fc receptor (FcRn). J Biol Chem 2006;281:23514-24
  • Presta LG. Molecular engineering and design of therapeutic antibodies. Curr Opin Immunol 2008;20:460-70
  • Yeung YA, Leabman MK, Marvin JS, et al. Engineering human IgG1 affinity to human neonatal fc receptor: impact of affinity improvement on pharmacokinetics in primates. J Immunol 2009;182:7663-71
  • Kubota T, Niwa R, Satoh M, et al. Engineered therapeutic antibodies with improved effector functions. Cancer Sci 2009;100:1566-72
  • Hinton PR, Xiong JM, Johlfs MG, et al. An engineered human IgG1 antibody with longer serum half-life. J Immunol 2006;176:346-56
  • Schreiber S, Khaliq-Kareemi M, Lawrance IC, PRECISE 2 Study Investigators. Maintenance therapy with certolizumab pegol for crohn’s disease. N Engl J Med 2007;357:239-50
  • Winter TA, Sandborn WJ, de Villiers WJ, Schreiber S. Treatment of crohn’s disease with certolizumab pegol. Expert Rev Clin Immunol 2007;3:683-94
  • Rutgeerts P, Schreiber S, Feagan B, et al. CDP870 Crohn’s Disease Study Group. Certolizumab pegol, a monthly subcutaneously administered fc-free anti-TNFalpha, improves health-related quality of life in patients with moderate to severe crohn’s disease. Int J Colorectal Dis 2008;23:289-96
  • Weinblatt ME, Fleischmann R, Huizinga TW, et al. Efficacy and safety of certolizumab pegol in a broad population of patients with active rheumatoid arthritis: results from the REALISTIC phase IIIb study. Rheumatology (Oxford) 2012;51:2204-14
  • Deeks ED. Certolizumab pegol: a review of its use in the management of rheumatoid arthritis. Drugs 2013;73:75-97
  • Qiu XQ, Wang H, Cai B, et al. Small antibody mimetics comprising two complementarity-determining regions and a framework region for tumor targeting. Nat Biotechnol 2007;25:921-9
  • Chames P, Van Regenmortel M, Weiss E, Baty D. Therapeutic antibodies: successes, limitations and hopes for the future. Br J Pharmacol 2009;157:220-33
  • Hansel TT, Kropshofer H, Singer T, et al. The safety and side effects of monoclonal antibodies. Nat Rev Drug Discov 2010;9:325-38
  • Skerra A. Alternative non-antibody scaffolds for molecular recognition. Curr Opin Biotechnol 2007;18:295-304
  • Gebauer M, Skerra A. Engineered protein scaffolds as next-generation antibody therapeutics. Curr Opin Chem Biol 2009;13:245-55
  • Wesolowski J, Alzogaray V, Reyelt J, et al. Single domain antibodies: promising experimental and therapeutic tools in infection and immunity. Med Microbiol Immunol 2009;198:157-74
  • Lofblom J, Frejd FY, Stahl S. Non-immunoglobulin based protein scaffolds. Curr Opin Biotechnol 2011;22:843-8
  • Wurch T, Pierre A, Depil S. Novel protein scaffolds as emerging therapeutic proteins: from discovery to clinical proof-of-concept. Trends Biotechnol 2012;30:575-82
  • Boersma YL, Pluckthun A. DARPins and other repeat protein scaffolds: advances in engineering and applications. Curr Opin Biotechnol 2011;22:849-57
  • Greenberg AS, Avila D, Hughes M, et al. A new antigen receptor gene family that undergoes rearrangement and extensive somatic diversification in sharks. Nature 1995;374:168-73
  • Roux KH, Greenberg AS, Greene L, et al. Structural analysis of the nurse shark (new) antigen receptor (NAR): molecular convergence of NAR and unusual mammalian immunoglobulins. Proc Natl Acad Sci USA 1998;95:11804-9
  • Streltsov VA, Varghese JN, Carmichael JA, et al. Structural evidence for evolution of shark ig new antigen receptor variable domain antibodies from a cell-surface receptor. Proc Natl Acad Sci USA 2004;101:12444-9
  • Richards MH, Nelson JL. The evolution of vertebrate antigen receptors: a phylogenetic approach. Mol Biol Evol 2000;17:146-55
  • Dooley H, Flajnik MF. Shark immunity bites back: affinity maturation and memory response in the nurse shark, ginglymostoma cirratum. Eur J Immunol 2005;35:936-45
  • Dooley H, Flajnik MF. Antibody repertoire development in cartilaginous fish. Dev Comp Immunol 2006;30:43-56
  • Barelle C, Gill DS, Charlton K. Shark novel antigen receptors – the next generation of biologic therapeutics? Adv Exp Med Biol 2009;655:49-62
  • Stanfield RL, Dooley H, Flajnik MF, Wilson IA. Crystal structure of a shark single-domain antibody V region in complex with lysozyme. Science 2004;305:1770-3
  • Stanfield RL, Dooley H, Verdino P, et al. Maturation of shark single-domain (IgNAR) antibodies: evidence for induced-fit binding. J Mol Biol 2007;367:358-72
  • Diaz M, Stanfield RL, Greenberg AS, Flajnik MF. Structural analysis, selection, and ontogeny of the shark new antigen receptor (IgNAR): identification of a new locus preferentially expressed in early development. Immunogenetics 2002;54:501-12
  • Liu JL, Anderson GP, Delehanty JB, et al. Selection of cholera toxin specific IgNAR single-domain antibodies from a naive shark library. Mol Immunol 2007;44:1775-83
  • Nuttall SD, Krishnan UV, Doughty L, et al. A naturally occurring NAR variable domain binds the kgp protease from porphyromonas gingivalis. FEBS Lett 2002;516:80-6
  • Nuttall SD, Krishnan UV, Doughty L, et al. Isolation and characterization of an IgNAR variable domain specific for the human mitochondrial translocase receptor Tom70. Eur J Biochem 2003;270:3543-54
  • Nuttall SD, Humberstone KS, Krishnan UV, et al. Selection and affinity maturation of IgNAR variable domains targeting plasmodium falciparum AMA1. Proteins 2004;55:187-97
  • Dooley H, Flajnik MF, Porter AJ. Selection and characterization of naturally occurring single-domain (IgNAR) antibody fragments from immunized sharks by phage display. Mol Immunol 2003;40:25-33
  • Goodchild SA, Dooley H, Schoepp RJ, et al. Isolation and characterisation of ebolavirus-specific recombinant antibody fragments from murine and shark immune libraries. Mol Immunol 2011;48:2027-37
  • Müller MR, Saunders K, Grace C, et al. Improving the pharmacokinetic properties of biologics by fusion to an anti-HSA shark VNAR domain. MAbs 2012;4:673
  • Bojalil R, Mata-Gonzalez MT, Sanchez-Munoz F, et al. Anti-tumor necrosis factor VNAR single domains reduce lethality and regulate underlying inflammatory response in a murine model of endotoxic shock. BMC Immunol 2013;14:17
  • Griffiths K, Dolezal O, Parisi K, et al. Shark variable new antigen receptor (VNAR) single domain antibody fragments: stability and diagnostic applications. Antibodies 2013;2:66-81
  • Liu JL, Zabetakis D, Brown JC, et al. Thermal stability and refolding capability of shark derived single domain antibodies. Mol Immunol 2014;59:194-9
  • Liu JL, Anderson GP, Goldman ER. Isolation of anti-toxin single domain antibodies from a semi-synthetic spiny dogfish shark display library. BMC Biotechnol 2007;7:78
  • Camacho-Villegas T, Mata-Gonzalez T, Paniagua-Solis J, et al. Human TNF cytokine neutralization with a vNAR from heterodontus francisci shark: a potential therapeutic use. MAbs 2013;5:80-5
  • Flajnik MF, Dooley H. The generation and selection of single-domain, v region libraries from nurse sharks. Methods Mol Biol 2009;562:71-82
  • Muller MR, O’Dwyer R, Kovaleva M, et al. Generation and isolation of target-specific single-domain antibodies from shark immune repertoires. Methods Mol Biol 2012;907:177-94
  • Shao CY, Secombes CJ, Porter AJ. Rapid isolation of IgNAR variable single-domain antibody fragments from a shark synthetic library. Mol Immunol 2007;44:656-65
  • Simmons DP, Streltsov VA, Dolezal O, et al. Shark IgNAR antibody mimotopes target a murine immunoglobulin through extended CDR3 loop structures. Proteins 2008;71:119-30
  • Nuttall SD, Krishnan UV, Hattarki M, et al. Isolation of the new antigen receptor from wobbegong sharks, and use as a scaffold for the display of protein loop libraries. Mol Immunol 2001;38:313-26
  • Walsh R, Nuttall S, Revill P, et al. Targeting the hepatitis B virus precore antigen with a novel IgNAR single variable domain intrabody. Virology 2011;411:132-41
  • Ohtani M, Hikima J, Jung TS, et al. Variable domain antibodies specific for viral hemorrhagic septicemia virus (VHSV) selected from a randomized IgNAR phage display library. Fish Shellfish Immunol 2013;34:724-8
  • Ohtani M, Hikima J, Jung TS, et al. Construction of an artificially randomized IgNAR phage display library: screening of variable regions that bind to hen egg white lysozyme. Mar Biotechnol (NY) 2013;15:56-62
  • Fennell BJ, Darmanin-Sheehan A, Hufton SE, et al. Dissection of the IgNAR V domain: molecular scanning and orthologue database mining define novel IgNAR hallmarks and affinity maturation mechanisms. J Mol Biol 2010;400:155-70
  • Kovalenko OV, Olland A, Piche-Nicholas N, et al. Atypical antigen recognition mode of a shark IgNAR variable domain characterized by humanization and structural analysis. J Biol Chem 2013;288(24):17408-19
  • Schmidt MM, Wittrup KD. A modeling analysis of the effects of molecular size and binding affinity on tumor targeting. Mol Cancer Ther 2009;8:2861-71
  • Obeidat M, Obeidat M, Ballermann BJ. Glomerular endothelium: a porous sieve and formidable barrier. Exp Cell Res 2012;318:964-72
  • Kitamura K, Takahashi T, Yamaguchi T, et al. Chemical engineering of the monoclonal antibody A7 by polyethylene glycol for targeting cancer chemotherapy. Cancer Res 1991;51:4310-15
  • Chapman AP, Antoniw P, Spitali M, et al. Therapeutic antibody fragments with prolonged in vivo half-lives. Nat Biotechnol 1999;17:780-3
  • Jevsevar S, Kunstelj M, Porekar VG. PEGylation of therapeutic proteins. Biotechnol J 2010;5:113-28
  • Stork R, Zettlitz KA, Muller D, et al. N-glycosylation as novel strategy to improve pharmacokinetic properties of bispecific single-chain diabodies. J Biol Chem 2008;283:7804-12
  • Smith BJ, Popplewell A, Athwal D, et al. Prolonged in vivo residence times of antibody fragments associated with albumin. Bioconjug Chem 2001;12:750-6
  • Kurtzhals P, Havelund S, Jonassen I, et al. Albumin binding of insulins acylated with fatty acids: characterization of the ligand-protein interaction and correlation between binding affinity and timing of the insulin effect in vivo. Biochem J 1995;312(Pt 3):725-31
  • Makrides SC, Nygren PA, Andrews B, et al. Extended in vivo half-life of human soluble complement receptor type 1 fused to a serum albumin-binding receptor. J Pharmacol Exp Ther 1996;277:534-42
  • Muller D, Karle A, Meissburger B, et al. Improved pharmacokinetics of recombinant bispecific antibody molecules by fusion to human serum albumin. J Biol Chem 2007;282:12650-60
  • Markussen J, Havelund S, Kurtzhals P, et al. Soluble, fatty acid acylated insulins bind to albumin and show protracted action in pigs. Diabetologia 1996;39:281-8
  • Stork R, Muller D, Kontermann RE. A novel tri-functional antibody fusion protein with improved pharmacokinetic properties generated by fusing a bispecific single-chain diabody with an albumin-binding domain from streptococcal protein G. Protein Eng Des Sel 2007;20:569-76
  • Dennis MS, Zhang M, Meng YG, et al. Albumin binding as a general strategy for improving the pharmacokinetics of proteins. J Biol Chem 2002;277:35035-43
  • Nguyen A, Reyes AE II, Zhang M, et al. The pharmacokinetics of an albumin-binding fab (AB.fab) can be modulated as a function of affinity for albumin. Protein Eng Des Sel 2006;19:291-7
  • Alt M, Muller R, Kontermann RE. Novel tetravalent and bispecific IgG-like antibody molecules combining single-chain diabodies with the immunoglobulin gamma1 fc or CH3 region. FEBS Lett 1999;454:90-4
  • Strohl W, Strohl L. Therapeutic antibody engineering: current and future advances driving the strongest growth area in the pharmaceutical industry. Woodhead Publishing, Cambridge, UK; 2012
  • Teillaud JL. From whole monoclonal antibodies to single domain antibodies: think small. Methods Mol Biol 2012;911:3-13
  • Buss NA, Henderson SJ, McFarlane M, et al. Monoclonal antibody therapeutics: history and future. Curr Opin Pharmacol 2012;12:615-22
  • Ahmad ZA, Yeap SK, Ali AM, et al. scFv antibody: principles and clinical application. Clin Dev Immunol 2012;2012:980250
  • McAleese F, Eser M. RECRUIT-TandAbs: harnessing the immune system to kill cancer cells. Future Oncol 2012;8:687-95
  • Hamers-Casterman C, Atarhouch T, Muyldermans S, et al. Naturally occurring antibodies devoid of light chains. Nature 1993;363:446-8
  • Ignatovich O, Jespers L, Tomlinson IM, de Wildt RM. Creation of the large and highly functional synthetic repertoire of human VH and vkappa domain antibodies. Methods Mol Biol 2012;911:39-63
  • Holt LJ, Basran A, Jones K, et al. Anti-serum albumin domain antibodies for extending the half-lives of short lived drugs. Protein Eng Des Sel 2008;21:283-8
  • Walker A, Dunlevy G, Rycroft D, et al. Anti-serum albumin domain antibodies in the development of highly potent, efficacious and long-acting interferon. Protein Eng Des Sel 2010;23:271-8
  • Skerra A. Engineered protein scaffolds for molecular recognition. J Mol Recognit 2000;13:167-87
  • Binz HK, Amstutz P, Pluckthun A. Engineering novel binding proteins from nonimmunoglobulin domains. Nat Biotechnol 2005;23:1257-68
  • Nuttall SD, Walsh RB. Display scaffolds: protein engineering for novel therapeutics. Curr Opin Pharmacol 2008;8:609-15
  • Lehmann A. Ecallantide (DX-88), a plasma kallikrein inhibitor for the treatment of hereditary angioedema and the prevention of blood loss in on-pump cardiothoracic surgery. Expert Opin Biol Ther 2008;8:1187-99
  • Delacourt C, Herigault S, Delclaux C, et al. Protection against acute lung injury by intravenous or intratracheal pretreatment with EPI-HNE-4, a new potent neutrophil elastase inhibitor. Am J Respir Cell Mol Biol 2002;26:290-7
  • Honore S, Attalah HL, Azoulay E, et al. Beneficial effect of an inhibitor of leukocyte elastase (EPI-hNE-4) in presence of repeated lung injuries. Shock 2004;22:131-6
  • Attucci S, Gauthier A, Korkmaz B, 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;318:803-9
  • Feldwisch J, Tolmachev V. Engineering of affibody molecules for therapy and diagnostics. Methods Mol Biol 2012;899:103-26
  • Bloom L, Calabro V. FN3: a new protein scaffold reaches the clinic. Drug Discov Today 2009;14:949-55
  • Lipovsek D. Adnectins: engineered target-binding protein therapeutics. Protein Eng Des Sel 2011;24:3-9
  • Tolcher AW, Sweeney CJ, Papadopoulos K, et al. Phase I and pharmacokinetic study of CT-322 (BMS-844203), a targeted adnectin inhibitor of VEGFR-2 based on a domain of human fibronectin. Clin Cancer Res 2011;17:363-71
  • Richter A, Eggenstein E, Skerra A. Anticalins: exploiting a non-ig scaffold with hypervariable loops for the engineering of binding proteins. FEBS Lett 2014;588:213-18
  • De Meyer T, Muyldermans S, Depicker A. Nanobody-based products as research and diagnostic tools. Trends Biotechnol 2014;32:263-70
  • De Vos J, Devoogdt N, Lahoutte T, Muyldermans S. Camelid single-domain antibody-fragment engineering for (pre)clinical in vivo molecular imaging applications: adjusting the bullet to its target. Expert Opin Biol Ther 2013;13:1149-60
  • Clinical Trials.gov. 2014. Available from: http://www.clinicaltrials.gov/ [Last accessed 2 May 2014]
  • Molecular Partners. 2014. Available from: http://www.molecularpartners.com/public/index.php?id=23&lang=en [Last accessed 2 May 2014]
  • Ablynx. 2014. Available from: http://www.ablynx.com/en/research-development/pipeline/ [Last accessed 2 May 2014]

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.