Sie sind auf Seite 1von 15

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/237014993

Nanobodies and their potential applications

Article  in  Nanomedicine · June 2013


DOI: 10.2217/nnm.13.86 · Source: PubMed

CITATIONS READS

81 933

5 authors, including:

Gholamreza Hassanzadeh-Ghassabeh Nick Devoogdt


Vlaams Instituut voor Biotechnologie Vrije Universiteit Brussel
78 PUBLICATIONS   2,478 CITATIONS    138 PUBLICATIONS   2,263 CITATIONS   

SEE PROFILE SEE PROFILE

Pieter De Pauw Cécile Vincke


Vrije Universiteit Brussel Vrije Universiteit Brussel
19 PUBLICATIONS   1,219 CITATIONS    48 PUBLICATIONS   1,056 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

AAV9 gene therapy - Gelsolin amyloidosis View project

Ultrasensitive Measurement of Ca(2+) Influx into Lipid Vesicles Induced by Protein Aggregates View project

All content following this page was uploaded by Serge Muyldermans on 01 June 2014.

The user has requested enhancement of the downloaded file.


Perspective

Nanobodies and their potential applications

Nanobodies are recombinant, antigen-specific, single-domain, variable fragments of camelid heavy chain-
only antibodies. The innate supremacy of nanobodies as a renewable source of affinity reagents, together
with their high production yield in a broad variety of expression systems, minimal size, great stability,
reversible refolding and outstanding solubility in aqueous solutions, and ability to specifically recognize
unique epitopes with subnanomolar affinity, have combined to make them a useful class of biomolecules
for research and various medical diagnostic and therapeutic applications. This review speculates on a
number of technological innovations that might be introduced in the nanobody identification platform

f
to streamline the generation of more potent nanobodies and to expand their application range.

oo
Keywords: Camelidae n crystallization chaperone n deep sequencing n enzyme Gholamreza
inhibitor n heavy chain-only antibody n immune repertoire n intracellular delivery Hassanzadeh-
n nanobody n single-domain antibody
Ghassabeh1,2,
Nick Devoogdt1,3,
From heavy chain-only antibodies to Therefore, antigen-specific scFvs are preferably Pieter De Pauw1,4,
nanobodies
Pr
obtained in a two-step procedure: first, a large
Cécile Vincke1,4
The natural immune system receives much inter- single-pot repertoire of naive or synthetic VH
& Serge Muyldermans*1,4
est because of its importance in various funda- and VL genes are cloned in an scFv format; 1
Vrije Universiteit Brussel, Research
mental and applied sciences. The soluble IgG second, the antigen-specific scFvs are retrieved group Cellular & Molecular
glycoproteins circulating in the blood play an from this repertoire after phage display. It is well Immunology, Pleinlaan 2,
1050 Brussels, Belgium
essential role in the humoral immune system of established that phage display panning selects for
or

2
VIB, Nanobody Service Facility, Vrije
mammals. These antibodies, produced by B lym- binders with the highest specificity and affinity Universiteit Brussel, Brussels, Belgium
phocytes, recognize with high specificity the to the target, in addition to the better producers 3
Vrije Universiteit Brussel, Research
group ICMIC, Laarbeeklaan 109,
foreign biological or chemical substance against and more stable constructs. With the objective 1090 Jette, Belgium
which they are affinity matured, and might alter of making even smaller antigen binders, similar 4
VIB, Department of Structural Biology,
h

Vrije Universiteit Brussel, Brussels,


the characteristics (e.g., function) of this cognate single-pot libraries have been constructed based Belgium„
antigen. The basic structure of an IgG molecule on soluble, stable, VHs with good expression, *Author for correspondence:
Tel.: +32 2 629 19 69
comprising two identical heavy (H) polypeptide where codons for amino acids within antigen-
ut

Fax: +32 2 629 19 81


chains and two identical light (L) polypeptide binding loops were randomized. While such svmuylde@vub.ac.be
chains is highly conserved among mammals. synthetic VH libraries seem to be a good source
The molecular weight of an IgG is approxi- to identify single-domain antibody fragments,
mately 160,000 Da, and it is a complex mol- they can be substituted equally well by other
A

ecule in which H and L chains fold into four and immunoglobulin folds such as VL or fibronectin
two domains, respectively (Figure 1) . Compared domains, as well as by engineered nonimmu-
with smaller antigen-binding fragments, such noglobulin scaffolds, including ankyrin repeat
as Fab (molecular weight: 50,000 Da) or scFv proteins (DARPins), lipocalins (anticalins) or
(molecular weight: 30,000 Da), only low yields protein A (affibodies), to name just a few [1] .
of functional recombinant IgG are obtained Each of these scaffolds has its benefits and prob-
from microorganisms. The scFv is a manmade ably limitations as well.
product composed of an N‑terminal, variable At the time that first successes were experi-
domain of the H chain (VH) and an N‑terminal, enced in retrieving antigen-specific scFvs or Fabs
variable domain of the L chain (VL), joined by from large synthetic or naive libraries displayed
a short synthetic peptide linker (Figure 1) . The on a phage, it was discovered serendipitously
scFv is the smallest intact antigen-binding frag- that dromedaries incorporate a unique class of
ment that can be derived from a conventional antibodies devoid of L chains in their sera, in
IgG molecule. Unfortunately, bacterial expres- addition to the conventional H 2L 2‑type IgGs
sion of scFvs cloned from previously identi- [2] . Thus, these homodimeric H2‑type antibod-
fied hybridomas often suffers from low yields. ies were called H chain-only antibodies (HCAbs) part of

10.2217/NNM.13.86 © 2013 Future Medicine Ltd Nanomedicine (2013) 8(6), 1–14 ISSN 1743-5889 1
Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

bactrianus, Lama glama, Lama pacos, Lama gua-


ScFv
Nb nicoe and Lama vicugna). However, similar func-

VH
tional antibodies devoid of L chains are also pres-

VL
ent in an assortment of non-mammals, such as

VH
H
sharks (Orectolobus maculates and Ginglymostoma
VH

VH
cirratum) and ratfish. In these cartilaginous fish,

VH

H
VL
VL

VH
these antibodies, commonly known as Ig‑NARs,
CH

H
1
H
C
1

L
have an architecture fairly different from that of
CL

Fab
C

camelid HCAbs: an Ig‑NAR is a homodimeric

CH2

CH2
H chain, each chain consisting of one single vari-
CH2

CH2

Fc able antigen-binding domain (V‑NAR) and five


Fc
constant domains (Figure 1) [3] .

CH3

CH3
Inspection of the molecular organization of
CH3

CH3

the H2L2 antibodies, the camelid HCAbs and


sdAb Ig‑NARs, teaches us that the H2‑type antibod-
ies recognize their cognate antigen by one single
AR

f
N domain, the VHH or the V‑NAR, respectively
VH
VH

V-

(Figure 1) . Crystal structures of VHHs reveal a

oo
VL
VL

AR
V-

prolate (rugby ball) shape of approximately


C1

N
C

N
AR

V-
2.5 nm in diameter and 4.2 nm in length [4] .
L
CL

Hence, these ‘nanometer-sized antibody frag-


ments’ are referred to as ‘nanobodies’ (Nbs)
C2

C2

C2

C2

[5] . Although V‑NARs and VHHs have similar


Pr sizes, the term Nb is only used for (recombinant)
camelid single-domain antibody fragments.
C3

C3

C3

C3

The fascination of nanotechnology originates


from altered mechanical, thermal and catalytic
C4

C4

C4

C4

properties of materials as their size decreases


from the micro- to nano-meter scale, enabling
or

novel applications. Likewise, the size reduction


C5

C5

C5

C5

of an HCAb into an Nb (and the concomitant


reduction in valency from bivalent to monova-
lent) can cause a dramatic change in biologi-
h
C6

C6

C6

C6

cal activity. Indeed, while a monoclonal HCAb


with specificity to trypanosomes is harmless to
the parasite (in the absence of complement), the
ut

Figure 1. Mammalian and shark antibodies. (A) Conventional mammalian IgG


antibodies, (B) camelid heavy chain-only antibodies, (C) cartilaginous fish IgW and isolation of Nbs from these HCAbs makes these
(D) shark Ig‑NARs. The smallest practical antigen-binding entity of IgG, the scFv, is antigen-binding entities highly trypanolytic
also shown, as well as the single-domain antigen-binding fragments of the camelid by blocking a very early endocytosis step via a
heavy chain-only antibodies and shark Ig‑NAR, known as nanobody and V‑NAR
mechanism that has not been elucidated yet [6] .
A

(sdAb), respectively.
C: Constant domain; CH: Constant domain of the heavy chain; CL: constant
domain of the light chain; Nb: Nanobody; SdAb: Single-domain antibody; Biochemical & biophysical properties
V-NAR: Variable antigen-binding domain; VH: Variable domain of the heavy chain; of Nbs
VHH: N‑terminal variable domain of the heavy chain-only antibody; VL: Variable After vaccinating a camelid (or a cartilaginous
domain of the light chain.
shark) to raise an H2‑type antibody response,
(Figure 1) . It was also noticed that the H chain the VHH (or V‑NAR) gene fragments from
of HCAbs is folded into three domains: an lymphocytes are amplified by PCR, ligated in
N‑terminal domain that is variable in sequence phage display vectors and transformed in bac-
(VHH), followed by a hinge region and two teria to construct an immune VHH library.
constant domains. Therefore, the equivalent of Expression of the cloned VHHs in fusion with
the first constant domain of the H chain of a the phage coat protein pIII, its assembly at the
conventional antibody is missing in H chains of tip of phage particles and enrichment of phage-
H2 antibodies. The occurrence of such functional displayed, antigen-specific VHHs on immobi-
HCAbs within mammals as a component of their lized antigens in two or three rounds of panning
humoral immune system seems to be limited to allows the identification, with a high success
Camelidae species (Camelus dromarius, Camelus rate, of Nbs targeting a wide variety of antigens

2 Nanomedicine (2013) 8(6) future science group


Nanobodies & their potential applications Perspective
with nanomolar to even picomolar affinities Streamlining the Nb identification
within weeks of the first antigen injection [7] . platform
The retrieved Nbs are successfully expressed Successful identification of antigen-specific Nbs
in microorganisms, thereby providing an easy is straightforward after immunizing a camelid
route to obtain large amounts of in vivo affinity- (or shark for V‑NAR) with purified, soluble,
matured, antigen-specific single-domain anti- properly folded proteinaceous antigens and
bodies. This is an important advantage over scFv phage display [7] . However, immunization with
or manmade scaffolds where in vivo affinity mat- more difficult targets, such as multipass mem-
uration cannot be accomplished easily by immu- brane proteins, receptor complexes, intrinsically
nization. In cases where vaccination is difficult disordered proteins or unstructured peptides and
(e.g., chemical and nonimmunogenic targets), it the subsequent selections against such targets
is possible to construct large single-pot synthetic remain critical steps.
VHH (or V‑NAR) libraries to identify antigen
binders [8] . However, the antigen-specific binders „„ Immunization of camelids or
retrieved from synthetic libraries are not affin- transgenic mice
ity matured and might benefit from additional The procedures to raise HCAbs in camelids are

f
sequence diversification steps and subsequent similar to those used to elicit conventional anti-
selections to generate more potent binders [9] . bodies in other animals. Various adjuvants as

oo
The crystal structures of VHHs in complex well as different immunization routes and boost
with their antigen [10] confirmed that the side of intervals successfully induce a HCAb response
the domain (known as the framework‑2 region) in camelids. Camelid immunization simultane-
that contacts a VL in an H2L2‑type antibody is ously elicits conventional and HCAb responses,
resurfaced from a hydrophobic region in VH to a with the former often being dominant. Any opti-
more hydrophilic region in a VHH. This frame-
Pr
mized immunization protocol (e.g., a particular
work‑2 resurfacing by amino acid substitutions adjuvant and/or immunization route) favoring
contributes to the strict monomeric behavior and the HCAb response is highly desired. Taking
the high solubility of VHH domains. Moreover, into account that camelids are large and out-
it explains the stickiness of many VHs in the bred animals, this immunization optimization
absence of a VL partner. However, the reduced is challenging, but feasible.
or

solubility of isolated VHs in aqueous solution Most antibody targets are membrane pro-
can be remediated by ‘camelizing’ the mouse teins, which are difficult to express and purify
or human VH, whereby hallmark amino acids as functional recombinant proteins. The avail-
of the framework‑2 region of a camelid VHH ability of such proteins in sufficient quantities for
h

are imprinted in the VH sequence [11] . Con- immunization and selection is, therefore, a hur-
versely, although Nbs are poorly immunogenic dle in all antibody technology platforms. Since
in humans, due to their high sequence identity the vast majority of HCAbs and Nbs recognize
ut

to human VHs, it is probably good practice conformational epitopes, the immunization of


for prolonged therapeutic applications involv- camelids with peptides (even coupled to larger
ing multiple administrations to first ‘human- protein carriers) is not recommended as it raises
ize’ the framework‑2 region of Nbs to imprint poor H2‑type antibody responses.
A

an amino acid sequence closer to human VHs DNA immunization, cell immunization and
in that region [12] . The humanization of Nbs, DNA prime/cell boost are potential alternatives
like any other site-directed mutagenesis on Nbs, to protein immunization. Both DNA prime fol-
is straightforward as the gene is only approxi- lowed by protein boost or cell immunization
mately 380 base pairs. have been used to generate Nbs [13,14] . Never-
The crystal structure of VHHs further theless, the former method still requires proteins,
revealed new structural organizations of the whereas cell boost or cell immunization results
antigen-binding loops, so Nbs are more suitable in an undesired antibody response to nontarget
than scFv paratopes for interacting with grooves cellular antigens, thereby complicating panning
on the surface of the antigen, such as the cata- and screening steps. To optimize DNA immuni-
lytic site of enzymes [10] . Indeed, it is well estab- zation protocols without the need for protein or
lished that many Nbs act as enzyme inhibitors or cell boosts, it will be necessary to systematically
modulate the function of the target and that the analyze the target-specific Nbs identified after
preferred antigenic sites for scFvs and VHHs are such immunization.
distinct. Of note, the V‑NARs and VHHs seem As testified by the large number of publica-
to share this preferential binding to clefts [3] . tions, immunization of camelids – or better to

future science group www.futuremedicine.com 3


Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

say, the size and exotic nature of camelids – has find the best pairs as they were affinity-matured
not been prohibitive in generating Nbs and in in vivo during immunization.
exploiting these Nbs in various applications.
However, immunizing transgenic mice, with the Selection of Nbs against proteome
exon for the first constant domain in H chain fractions
gene(s) deleted, might streamline the Nb iden- It is common practice to immunize camelids
tification platform. The proof of principle for with a mixture of five to ten antigens and to sort
such mice as a source of Nbs has already been by panning antigen-specific binders to each com-
given [15,101] , but the concept of transgenic mice ponent afterwards. With this in mind, it should
as substitutes for camelids to rapidly generate be possible to immunize with a crude extract of
potent HCAbs and/or high-affinity Nbs still any particular cell, possibly containing a cur-
remains to be demonstrated. rently unknown ‘active’ protein. The camelid
will raise HCAbs to immunogenic proteins and
„„ Nb selection the cloned VHH library can be panned subse-
Current Nb identification strategies involve quently on the same proteome extract to enrich
enrichment of antigen-specific binders from for Nbs against any component within the crude

f
immune or naive libraries by phage display, extract. After the last round of panning, indi-
bacterial two-hybrid or bacterial surface dis- vidual clones can be screened by ELISA [19] or

oo
play [7,8,16] , and screening of individual colonies by a functional assay to identify Nbs that bind
from enriched phage populations, primarily by or modulate the ‘activity’ of a particular compo-
ELISA. However, advanced approaches are in nent of the mixture (see ‘Nb-based phenotypic
sight, and examples of such approaches are pro- screening’ section). Once such Nbs are isolated,
posed below. they can be purified and immobilized to cap-
Pr ture the antigen from the mixture. The captured
Deep sequencing to avoid repertoire antigen can then be identified, for example, by
cloning & selection mass spectrometry [20] .
Recently, next-generation sequencing (NGS) If the biomarker is known, it is possible to
was used to calculate the relative frequencies of use a minimal component fraction (i.e., a cell
genes encoding VH and VL in a cDNA pool fraction enriched for the biomarker and depleted
or

of plasma cells from immunized mice. Subse- of irrelevant proteins) for immunization and
quently, the most abundant VH and VL genes subsequent phage selections. However, in cases
were synthesized and paired to obtain antigen- where the biomarker of interest is unknown,
specific scFvs [17] . This study provides an exam- the possibility to use crude, unfractionated
h

ple of bypassing tedious library construction, cell extracts has an advantage over the use of
biopanning and ELISA screening steps. Moreo- minimal component fractions because the iden-
ver, the NGS screening identified antibodies that tification of the fraction containing the (unde-
ut

were lost during panning or went undetected by termined) biomarker causing the phenotype of
ELISA [18] . Interestingly, the recombinant anti- interest can be very tedious (especially when it
bodies identified by NGS, but not by standard is a multicomponent biomarker).
panning and ELISA screening, were expressed
A

in Escherichia coli at relatively high levels under Nb-based phenotypic screening


optimized conditions. The potential of antibodies for high-throughput
We anticipate that implementing the NGS phenotypic screening to discover new targets
approach will be extremely favorable to rapidly of therapeutic value still remains unexplored.
discover antigen-specific Nb sequences. Indeed, This is partly due to the inability of antibod-
the antigen-binding fragment of HCAbs involves ies to access intracellular targets and the low
only one single exon (VHH) instead of two (VH throughput of antibody discovery platforms.
and VL) in conventional antibodies. Therefore, Despite these shortcomings, antibodies repre-
the most abundant VHH sequences in a pool of sent an interesting class of molecules for high-
lymphocytes should correspond to those from throughput phenotypic screening, because of
HCAbs on B cells that maximally proliferated their high affinity, fine specificity and almost
during immunization. Moreover, the single- limitless repertoire diversity. Indeed, the feasibil-
domain Nb format immediately corresponds to ity to use antibodies for the discovery of novel
the intact antigen-binding site of HCAbs, and drugs or drug targets by phenotypic screening
avoids the requirement to combine all abundant was recently demonstrated by using both naive
VHs and VLs in all possible combinations to and immune scFv libraries [21] . In this work,

4 Nanomedicine (2013) 8(6) future science group


Nanobodies & their potential applications Perspective
individual scFvs, from affinity selections on magnetic sorting. Nbs against common antigens
Pseudomonas aeruginosa whole cells, were tested (present on both transfected and nontransfected
by ELISA for binding to heterologous serotype cells) will bind mainly to the untransfected cells
strains. The scFvs exhibiting serotype-inde- due to their higher abundancy, and, therefore,
pendent binding to P. aeruginosa were recon- those Nbs will be eliminated by sorting the
stituted into an IgG1 format and assayed for labeled transfected cells.
opsonophagocytic killing of P. aeruginosa. The
target antigens of scFvs with such activity were Nbs with improved properties
„„
then identified by assaying the binding of scFvs In vitro affinity improvement of Nbs
to P. aeruginosa mutant strains [21] . In some instances, the immunization of a
The robustness of the Nb identification tech- camelid is impossible (owing to highly toxic
nology and the possibility to generate and iden- or pathogenic immunogens, lack of purified
tify Nbs on the proteome scale [19] , together with properly folded antigen and nonimmunogenic
numerous data on the potent and target-specific compounds) and, thus, alternatives to in vivo
effects of Nbs, suggest that Nbs can serve as maturation might be necessary to obtain
alternatives to small chemical compounds for high‑affinity antigen-specific binders.

f
phenotypic screening aimed at the discovery of Generating bivalent Nb constructs might
both novel drugs and drug targets. However, for increase the functional affinity (i.e., avidity)

oo
this concept to be fully realized, future develop- [23] , but does not change the intrinsic affin-
ments are vital such as incorporation of NGS ity between Nbs and antigens. In principle,
into the Nb identification platform to increase improvement of the intrinsic affinity of Nbs
its throughput and preferably the ability to tar- for their antigen should be easier than that
get intracellular molecules in a high-throughput with scFvs because of Nbs’ smaller size, single-
manner.
Pr
domain nature, easy folding and the presence of
only three antigen-binding loops. These char-
Selection on cells acteristics make the construction of second-
Purification of properly folded, multipass mem- generation libraries on a previously selected
brane receptors (e.g., pharmacologically impor- Nb scaffold less complex and allow more robust
tant G protein-coupled receptors) or receptor phage or yeast display selections instead of more
or

complexes (e.g., integrins) for immunization and demanding selection techniques, such as ribo-
selection is a major bottleneck. For such targets, some display. Libraries of Nb variants with
there are two solutions: either, immunization either localized or unbiased random mutations
is avoided and non-affinity-matured Nbs are need to be generated. The targeted options use
h

retrieved from naive or synthetic libraries [8] , site-specific randomization of the codons for
or immune libraries will be constructed after the amino acids of the antigen-binding loops.
immunizing with DNA only [14] or transgenic The availability of structural information of the
ut

cells that overexpress such receptors. For the lat- antigen-binding loops of the Nb, preferably in
ter, transgenic DUBCA cell lines might be very complex with its antigen, is helpful, if not criti-
useful as these dromedary-derived cells are sup- cal, to pinpoint the mutagenesis hot spots. Such
posedly poorly immunogenic in camelids. Dur- antigen-binding hotspots can also be identified
A

ing the subsequent Nb selection process, special by Ala-scanning mutagenesis [9] . Apart from
measures need to be taken to retrieve binders these techniques based on evolution, rational
targeting the (perhaps less immunodominant) approaches might also be successful. Indeed, it
receptor of interest from a vast pool of poten- has been shown that mutating a limited num-
tial binders that target unwanted membrane ber of amino acids, carefully selected at the
proteins. It is imperative to eliminate the Nbs periphery of the paratope, guided by param-
to these irrelevant antigens. This is achieved to eterized quantitative descriptors (using a mul-
some extent by the biopanning and rapid ana­ tivariate experimental design), and measuring
lysis of selective interactive ligands, the ‘BRA- the effect of these mutations on the antigen-
SIL’ method [22] , or by panning on two different binding kinetics could be used to construct
transgenic cell-lines with very different origins. an algorithm that predicts the kinetic binding
Otherwise, multiple rounds of selections can parameters as well as the equilibrium binding
be performed on a cell mixture, in which fluo- constant of other possible mutations at those
rescently or magnetically labeled transfected positions [24] . Conversely, the unbiased, rand-
cells are diluted into unlabeled, untransfected omized Nb libraries are obtained via error prone
cells and then subjected to flow cytometric or PCR or the use of mutator strains. These fully

future science group www.futuremedicine.com 5


Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

random approaches have the disadvantage that compared with larger antibody formats. Their
a vast number of dysfunctional Nbs are formed monovalent mode of binding allows mild elution
and the advantage that occasionally a mutant conditions, which is particularly important for
might emerge with an amino acid substitution sensitive molecules. Moreover, their high stabil-
outside the antigen-binding loops with indirect ity and refolding capacity permits repeated and
favorable binding properties. stringent regeneration conditions.
Finally, the implementation of Nbs in the Nb-based affinity capture has been used to
phage-assisted continuous evolution technol- study protein–protein interactions [30] , ana-
ogy, an elegant technique in which phage fitness lyze in vivo DNA–protein interactions at the
constitutes the driving force to select perma- genomic scale after chromatin immunoprecipita-
nently novel mutants evolving spontaneously, tion, track protein conformations, trace bacterial
rapidly and continuously [25] , is expected to infections and purify recombinant proteins. Nbs
produce Nbs of greatly improved potency. have been employed for scavenging toxin from
plasma [31] and for depletion of abundant serum
Cell-penetrating Nbs proteins, a step required for proteomic ana­lysis
The availability of transducing Nbs (i.e., Nbs of blood samples [32] . The use of KDEL-specific

f
with the capacity to penetrate the membrane of Nbs to capture endoplasmic reticulum-resident
their specific target cell) that deliver functional proteins [23] raises the possibility to use signal

oo
molecules to the cytoplasm would offer an ele- peptide-specific Nbs as a generic tool for high-
gant solution to overcome the impermeability of throughput mapping of protein locations in
cells to most macromolecules [26] . These trans- physiological and pathophysiological conditions
ducing Nbs could be used to produce ‘Trojan by combining Nb-based affinity capture with,
horses’, directing proteins, DNA or siRNA spe- for example, anti-proteome Nb arrays. Unfortu-
Pr
cifically to diseased cells to provoke cytotoxicity, nately, generating Nbs against other signal pep-
to restore a lacking function in deficient cells, tides (e.g., palmytoylation, sumoylation, phos-
or to induce production of complementary pro- phorylation, S-nitrosylation and Y-nitration)
teins. To achieve transduction, one could take might be very challenging as these sites are less
advantage of the existing internalizing proteins well conserved and/or located in a flexible region
on the target cell surface (e.g., by inducing of the protein (i.e., poorly antigenic). Neverthe-
or

receptor dimerization and internalization with less, the KDEL and chromatin immunoprecipi-
a bivalent or bispecific Nb). Another approach tation examples enforce the idea that Nb-based
would be the addition of protein transduction affinity capture, alone or in combination with
domains, such as the one derived from the HIV other proteomics tools, might serve many pur-
h

Tat protein or penetratin, or by increasing the poses of modern proteomics.


Nbs’ arginine content, a condition that facilitates
internalization [27] . Finally, a promising tech- „„ Nbs as crystallization chaperones
ut

nology was developed whereby Nbs equipped Nbs exhibit a good track record as chaperones
with a short peptide are delivered into human to crystallize challenging proteins [33–35] . Indeed,
cells, directly from E. coli via its type III protein these robust, single-domain antibody fragments
secretion system [28] . It is surmised that expres- often lock proteins in a particular conformation
A

sion of a cell type-specific Nb on the surface [36] . The ability of Nbs to stabilize intrinsically
of attenuated, invasive bacterial strains might flexible regions or shield aggregating surfaces
direct these engineered cells to specific organs from contact with solvents are key features to
or tissues of the host. Colonization of the target allow the crystallization process [37] . DARPins
organ (e.g., solid tumors, infected or diseased and anticalins appear to serve the same purpose,
cells) might then allow prolonged delivery of a whereas crystallization with scFvs appear to be
protein therapy by a relevant second Nb (i.e., less successful, possibly because of the intrinsic
intrabody), injected to interfere or reprogram flexibility of VH and VL interactions, or the
the diseased cells. linker. The favorable properties of Nbs allowed
an impressive list of structures of highly dynamic
Nbs in research proteins to be elucidated and identification of
„„ Nbs as affinity capture reagents unknown conformations, providing insight into
Nbs are suitable affinity capture reagents [29] the mechanisms of action of their antigens. In
because their small size and single-domain for- addition, Nbs might become an essential tool to
mat provide higher capacity binding surfaces obtain valuable structural information on par-
and lower nonspecific background binding, as tially disordered or amyloid proteins [38,39] . For

6 Nanomedicine (2013) 8(6) future science group


Nanobodies & their potential applications Perspective
example, conformation-sensitive Nbs stabilize advantage over other antibody fragments, and
Amyloidb protofibrils and prevent the forma- they will probably become a valuable research
tion of mature amyloid fibrils [40] . Structural tool. However, the progression of Nb-based
data of such trapped amyloid intermediates intrabodies towards clinical application is less
might unravel the aggregation process of these certain and relies on the development of spe-
tangled targets and improve our understanding cific and efficient transduction methods. In this
of the mechanisms leading to their pathological respect, RNAi methodologies are certainly more
conversion. advanced. However, we believe that Nbs might
find a niche in cases where post-translationally
„„ Intracellular target imaging & modified protein variants or isoforms that
immunomodulation cannot be discriminated via RNAi need to be
Antibodies acting inside living cells are known targeted.
as ‘intrabodies’. Nbs fold surprisingly well into
functional entities, even in the reducing intra- Nbs in diagnosis
cellular environment. Therefore, intracellular „„ Nbs as probes in novel biosensors
expression of Nbs fused to fluorescent proteins Biosensors development is a popular area of his-

f
produces useful chromobodies or fluobod- tory because sensitive analyte detecting devices
ies that can be used to trace their antigen in are required for on-site application in many

oo
living cells [41,42] . This technology has been major fields, such as medical diagnosis, environ-
adapted to advanced super-resolution imaging mental and food ana­lysis. Many of these innova-
techniques [43] . tive biosensors rely on small, robust, highly spe-
The intracellular robustness, together with cific affinity probes, where size and directional
the target-modulating activity of Nbs, makes immobilization matter. Nbs are particularly
them an ideal tool to impair antigen activity
Pr
suited for controlled and reproducible coupling
inside the cells, producing functional knockouts because site-specific functional groups are eas-
[41,44–46] . Apart from being a target validation ily introduced, allowing covalent and oriented
tool, such Nbs might become valuable as leads binding with minimal loss of specificity and
to investigate molecules currently considered to affinity [50] . The Nbs’ small size provides high
be ‘undruggable’ (by definition, an undruggable binding capacity surfaces resulting in higher
or

target cannot be modulated appropriately by any sensitivity, and the high stability and refolding
currently known, available and safe molecule) . competence of Nbs allow stringent washing and
Nbs fused with leader signal peptides can be regeneration conditions.
directed to specific subcellular compartments. Moreover, Nbs directionally immobilized
h

The expression of Nbs, even those without onto solid sensor surfaces allow immunoreac-
intrinsic target inhibitory activity, at these sites tions to occur very close to the sensor–solu-
might sequester their targets from their normal tion interface, resulting in increased sensitivity.
ut

subcellular compartments, thereby inhibit- Alternatively, the analyte-capturing Nbs can


ing the function of the antigen. One example be joined spontaneously to magnetic particles
involves the intracellular retention of otherwise [51] , which are suspended in the sample solution
secreted proteins such as viral coat proteins [47] . to bind analyte before being attracted to the
A

Nbs, fused to a secretory signal peptide and a electrode by magnetic fields. The concentra-
C-terminal KDEL peptide, sequester both the tion enhancement of the analyte at the surface
Nb and its antigen in the endoplasmic reticu- of the electrode then leads to amplification of
lum. In another setting, Nb-based intrabodies the signal.
containing a PEST motif redirected the target
antigen to the proteasomal degradation pathway „„ Nbs for noninvasive in vivo imaging
[48] . Likewise, green fluorescent protein-tagged Noninvasive molecular imaging uses labeled
proteins could be depleted in vivo via the ubiq- tracers to visualize molecules and cellular bio-
uitin depletion pathway by the intracellular markers in the whole body without disturbing
expression of a green fluorescent protein-specific the host. This technology is very useful to inves-
Nb fused to the F‑box domain [49] . Despite these tigate biological processes in animal models and
successful examples, the broad applicability of is expected to become a major tool in nuclear
Nb-based PEST and F‑box-mediated protein medicine departments of hospitals to diagnose
knockout awaits independent confirmation. and stratify patients following PET and SPECT
In summary, because of their extraordinary scans. An ideal (clinical) tracer should be sta-
intracellular functionality, Nbs have a clear ble in vivo, with the ability to reach all sites in

future science group www.futuremedicine.com 7


Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

the body, generate signals over a long range of animals [53] or via coinjection of an unlabeled
biomarker concentrations, interact with the bio- competitor [54] . In preclinical animal models,
marker with high affinity and specificity, easy we have shown focal uptake of radiolabeled Nbs
to label with dyes or radionuclides, inexpensive, in tumors targeting markers of cancer cells [52]
and nontoxic. In clinical imaging, the generation or tumor-infiltrating macrophages (Figure 2) [53] ,
of contrast at the site of biomarker expression in arthritic lesions, atherosclerotic plaques [54]
within hours of tracer administration is essential. and immune organs [53] . A Phase I study with
An Nb meets most, but not all, requirements to an anti-HER2 Nb for PET imaging of breast
become a generic imaging tracer [52] . Nbs are cancer patients is currently ongoing.
usually stable, easily labeled, have low immuno- Nb-based imaging also has some limitations.
genicity and have affinities in the low nanomolar First, their fast renal excretion leads to high
to high picomolar range. Their small size assures signals in the kidneys and bladder, and imag-
optimal tissue penetration and fast blood clear- ing of specific tracer uptake at nearby sites is
ance, so that high contrast is attained within obstructed. Second, Nbs rarely penetrate the
hours of administration. Their target specific- blood–brain barrier [55] and, therefore, might be
ity has been demonstrated using gene-deficient suboptimal in the field of neuroimaging. Third,

f
while Nb-based imaging of cell surface receptors
is straightforward, tracing intracellular or solu-

oo
ble targets is more challenging. Finally, GMP
production of Nbs is far more expensive than
other useful tracers that are based on peptides
Pr or chemical compounds.

Nbs for therapy


Liver „„ Serum half-life extension
Antibody fragments, including Nbs, display
short serum half-life profiles. Although this is
highly desirable for certain applications, such as
imaging with radiolabeled tracers, many thera-
or

peutic applications require a slow drug clearance


Kidneys
rate to avoid high doses and frequent administra-
tion. PEGylation or modification with a serum
albumin-specific small chemical compound, and
h

genetic fusion with conformationally disordered


polypeptides (e.g., HAPylation and PASylation),
Tumor with a protein having an inherently long serum
ut

half-life (e.g., serum albumin) or with a mol-


ecule binding to an abundant serum protein
are among the many different approaches used
Bladder to extend the serum half-life of therapeutics
A

[56] . However, these modifications might have


adverse effects on antibody functionality [57] .
Injection site Nbs specific for abundant serum proteins,
such as albumin and IgG, have a long serum half-
life [58,59] , and fusion of such Nbs to other thera-
Figure 2. Maximal Intensity projection of peutic compounds provides a generic approach
fused pinhole SPECT/micro-computed
tomography image of a mouse bearing a
to extend the plasma half-life of this therapeutic.
TSA tumor implanted in the mammary fat Other studies described the extension of Nbs’
pad. The image was taken 3 h after injection serum half-life by PEGylation [60] , pentameriza-
with a 99mTc radio-labeled anti‑macrophage tion and fusion with IgFc [61] . The small amount
mannose receptor nanobody together with an of data on Nbs’ serum half-life extension makes
excess of bivalent, unlabeled macrophage
mannose receptor nanobody to reduce tracer
it difficult to speculate on the preferred method
uptake in organs other than the tumor. Note the for each application. However, it is evident that
accumulation of the tracer inside the tumor, methods that adversely affect Nbs’ advantages
visualizing mannose receptor-positive should be avoided. For instance, fusion of Nbs to
tumor‑infiltrating macrophages [53] . albumin or IgFc may complicate the expression

8 Nanomedicine (2013) 8(6) future science group


Nanobodies & their potential applications Perspective
of Nbs in microorganisms, and pentamerization established that polyclonal antibodies are more
(by fusion to verotoxin) may render Nbs immu- effective at combatting such infections than
nogenic. It is also worth noting that fusion with monoclonal antibodies. However, the possibil-
IgG-specific Nbs for serum half-life extension ity to screen thousands of antigen-specific Nbs
may provoke Ig-mediated immune effector cell from immune VHH libraries, in conjunction
activation at undesired locations. with their intrinsic capacity to target epitopes,
which are cryptic for conventional antibodies,
„„ Nbs against envenoming has provided access to a number of neutralizing
The preferred serotherapeutic application of Nbs against a myriad of pathogens [66,67] . A suc-
Nbs over other antibody formats is illustrated cessful Phase I clinical trial with Nbs targeting
in the field of envenoming caused by scorpions or respiratory syncytial virus has demonstrated
snakes [62,63] . Current antivenoms are polyclonal the potential of Nbs to combat infections [102] .
immunoglobulin fragments purified from the Since Nbs are devoid of any Fc region, they do
blood of venom-immunized horses and sheep. not have Fc-mediated effector functions and are
However, these antivenoms are often associated unable to neutralize and eliminate the patho-
with low potency, variable efficacy and severe gen. However, several Nbs have an inherent

f
adverse effects (e.g., serum sickness). This low neutralizing effect that is masked in HCAbs
potency is mainly attributed to a poor immune because pathogens have evolved to escape this

oo
response of host animals because most toxic antibody-mediated threat [6] . Alternatively, the
compounds within venom are small and poor strict monomeric nature of Nbs facilitates their
immunogens. Owing to their small size and fusion with molecules with innovative effector
the absence of an Fc region, Nbs diffuse rapidly functions, such as enzymes or toxic molecules,
through the body and reach a tissue biodistribu- in order to eradicate the pathogen. Additional
tion that closely matches that of the small venom
Pr
success with Nb therapies will come from more
toxins. In addition, even after capturing and patient-friendly administration (topical, inhala-
neutralizing the toxin, the toxin–Nb complex tion or oral delivery, and colonizing bacteria-
remains sufficiently small to be eliminated rap- releasing therapeutic Nbs) and from the variety
idly via the kidneys. As a result, a bispecific Nb of strategies used to obtain improved blood levels
against the Androctonus australis hector AahI and over a prolonged time.
or

AahII toxin – the most toxic molecules in scor-


pion venom – was shown to possess, in mouse „„ Nbs against amyloidosis
models, a higher neutralization capacity than The search for lasting treatments that target the
the current anti-scorpion toxin immunotherapy underlying causes and alter the natural course
h

[64] . Recently, in vivo echocardiography in rats of amyloidosis disorders (e.g., Alzheimer’s,


demonstrated the ability of this 30‑kDa con- Parkinson’s and Huntington’s diseases), linked
struct to successfully prevent the fatal hemody- with misfolding or aggregation of proteins into
ut

namic disturbances provoked by a lethal dose of fibrillar deposits, is an extremely active area of
venom [65] . Intact camelid IgG antibodies and, clinical investigation. Regarding Alzheimer’s
in particular, their Nb derivatives are equally or disease, active and passive immunotherapy
more potent than the conventional antivenom in have been suggested for more than a decade
A

neutralizing the lethal, hemorrhagic and coagu- as potential strategies to retard or even reverse
lopathic effects of west African viper (Echis ocel- disease progression. The form of the amyloid‑b
latus) venom [62] . Further research to define the peptide that leads to synaptic, neurotic and
pathology-inducing compounds of the complex behavioral abnormalities has been difficult to
snake venoms might lead to the identification define, although growing evidence suggests that
of Nbs against conserved epitopes, which may oligomeric amyloid‑b intermediates are prob-
eventually result in the development of polyreac- ably more toxic than the polymeric fibrils [68] .
tive toxin-specific antivenoms. Finally, the pro- Antibody-mediated interference must prevent
duction of sustainable Nb-based recombinant the conformational transitions that lead to the
antivenoms is expected to contribute favorably assembly of oligomers, protofibrils and fibrils,
to the reproducibility, safety and treatment cost which ultimately lead to plaque deposition. The
of envenomed victims. clearance should, therefore, preferably be per-
formed before the proteins cluster into the toxic
„„ Nbs against infections oligomers, by only targeting a specific subset
Antibodies are natural defense molecules against of amyloid‑b precursors. The single-domain
bacterial, viral and parasite infections. It is well nature of Nbs and their availability to work

future science group www.futuremedicine.com 9


Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

with immune repertoires greatly facilitates their administration of different antibodies) or,
isolation as highly specific probes for patho- alternatively, by using multispecific antibodies.
logical target structures with excellent affinity Compared with combination therapy, multispe-
[69,70] . In addition, Nbs are readily available for cific antibodies provide the possibility to reduce
large-scale expression, and if penetration in the the costs of production, preclinical testing and
CNS appears to be necessary for efficacy, some clinical trials [83] . Bispecific antibodies hold
Nbs that recognize unique epitopes on epithelia great promise as next-generation therapeutics,
have been described to transmigrate through especially for cancer treatment [83–85] .
the human blood–brain barrier in vitro and The interesting properties of Nbs, in par-
in vivo via active transport [71,72] . ticular their strict monomeric behavior, single-
domain nature and minimal size, make them
„„ Nbs against pathologies & cancer attractive candidates for the development of
Therapeutic antibodies often rely on activation manifold constructs [86,87] . Based on the pub-
of immune effector mechanisms for optimal licly available data, this notion has surprisingly
efficacy. Due to the lack of Fc, which medi- largely stayed in the explorative phase. This
ates immune effector cell activation, Nbs in might be, at least in part, due to the restrictions

f
therapeutic intervention are entirely depend- resulting from intellectual property rights asso-
ent on intrinsic antagonistic antigen targeting, ciated with the use of Nbs, taking into account

oo
and possibly on agonistic signaling. Conversely, that the major driving force behind bispecific
lack of Fc in Nbs may result in fewer unwanted antibodies is the desire to finally bring them to
immune-mediated side effects. clinics. With some of these patents coming to
The application of Nbs to combat diseases an end soon, we anticipate much faster progress
and pathologies mainly focuses on the inhibi- in the field of bispecific Nbs in the near future.
Pr
tion of ligand–receptor interactions. Examples Bispecific antibodies have often been used to
include antagonizing anti-von Willebrand fac- activate immune effector cells and their rerout-
tor Nbs to block thrombosis initiation [73] , anti- ing to cancer cells [83] . An alternative approach
TNF‑a Nbs to treat arthritis [74] and EGF– to the use of bispecific antibodies for rerouting
EGFR blocking Nbs to treat cancer [75] . Other and activation of T cells is the T‑body approach
examples include Nbs that keep antigens in an (i.e., generation of T cells expressing chimeric
or

inactive receptor state [76] or provoke receptor receptors consisting of an antibody directed
activation (agonist action) for apoptosis induc- to a tumor-specific antigen) [88,89] . The small
tion of cancer cells. One advantage of Nbs is size and high stability/folding capacity of Nbs
their small size, which means that dense tissues suggest that they could offer advantages over
h

can be penetrated by bivalent or trivalent Nb other antibody formats for the generation of a
complexes [59] . Hence, they are effective even T‑body. Proof of concept for Nb-based T‑bodies
at occluded sites. Since Nbs are usually rapidly has been provided [90] , but further studies are
ut

excreted from the body, their serum half-life required to catalyze the translation of this con-
needs to be controlled (see ‘Serum half-life cept into effective therapies.
extension’ section).
Recently, Nbs have been employed as a cell Conclusion & future perspective
A

(re-)targeting moiety in larger theranostic The possibility to rapidly obtain in vivo affin-
vehicles, such as lentiviruses [77] , microbub- ity matured Nbs, combined with their strict
bles [78] , branched nanogold particles [79] and monomeric behavior, and favorable biochemi-
albumin nanoparticles [80] , bio-nanocapsules cal and physical properties, make Nbs unique
[81] , and polymeric micelles [82] . The toxicologi- among multitarget recognition molecules that
cal profile of these relatively large particles in are currently available. Therefore, it is expected
clinical settings still needs to be evaluated, as that Nbs will make a substantial difference in
well as their targeting capacity after systemic therapy, diagnostic screenings and research.
administration. Although no Nb-based product is currently
approved as a therapeutic agent, the fast identi-
„„ Bispecific Nbs & Nb-based T-body fication of a Nb-based lead gives Ablynx‑NV a
Complex diseases are often multifactorial in substantial advantage, as shown by the various
nature; therefore, necessitating simultaneous Nb-based products that are already in the pipe-
targeting of multiple factors or pathways for line and/or in advanced clinical phases. Besides
maximal therapeutic effect. This can be achieved Nbs developed to combat cancer, diseases
by combination therapy (i.e., simultaneous such as amyloidoses, viral infections or toxin

10 Nanomedicine (2013) 8(6) future science group


Nanobodies & their potential applications Perspective
envenoming could be treated with future Nb- Acknowledgements
based therapeutics. Evidently, the development The authors wish to thank all personnel from our laboratory
of Nbs into therapeutics remains a high-risk, and collaborators all over the world for sharing their excit-
high-reward business, and commercialization of ing data and for their much-appreciated contributions in
Nbs will be faster in the diagnostic field and as the past 20 years.
a research tool. For diagnostic applications, Nbs
developed for noninvasive in vivo imaging of Financial & competing interests disclosure
tumors and lesions have definitely a great poten- The authors give special thanks to granting authorities
tial. In addition, their application as a highly from the EU (AFFINOMICS project 241481), NATO
specific probe on µ‑arrays or in novel biosen- (Science for Peace), Belgium (Ministry of Health),
sors will grow steadily in the very near future. Flanders (IWT and FWO), VIB (Technology funds) and
Finally, as a research tool, Nbs are already quite OZR of Vrije Universiteit Brussel, ‘Vlaamse Liga tegen
useful as crystallization chaperones or when Kanker’ and ‘Stichting tegen kanker’ for their financial
fluorescently labeled as tracers of antigen traf- support. The authors have no other relevant affiliations or
ficking inside living cells at the highest possible financial involvement with any organization or entity with
resolution. The Nbs with an intrinsic antigen- a financial interest in or financial conflict with the subject

f
modulating activity (e.g., enzyme activation matter or materials discussed in the manuscript apart from
or inhibition), sometimes even leading to the those disclosed.

oo
production of functional antigen knockouts will No writing assistance was utilized in the production of
be a valuable research tool for target validation. this manuscript.

Executive summary
Biochemical & biophysical properties of nanobodies
Pr
ƒƒ An optimized technology platform has been developed to identify nanobodies (Nbs) against soluble, properly folded, immunogenic
proteins from immune variable domain of a heavy chain-only antibody phage display libraries.
ƒƒ Such Nbs are naturally endowed with good biochemical and biophysical properties.
Streamlining the Nb identification platform
or

ƒƒ Development of transgenic mice that can be immunized to elicit heavy chain-only antibodies and used to generate Nbs may further
streamline the Nb technology.
ƒƒ Novel immunization protocols to maximally stimulate the heavy chain-only antibodies’ immune response needs to be developed.
ƒƒ Further streamlining of the Nb identification platform for difficult targets (transmembrane biomarkers) is envisaged by DNA
immunization or cell immunization, and panning on cells.
h

ƒƒ Next-generation sequencing technologies should be introduced for rapid target-specific Nb identification obviating the need to
construct immune libraries and panning.
ut

ƒƒ Immune Nb libraries against (sub-)proteomes are predicted to constitute a valuable source for novel target identification, especially
when linked with phenotypic screening technologies that are still in their infancy.
ƒƒ Affinity maturation technologies are available, but introducing Nbs in the phage-assisted continuous evolution technology might be
developed to generate Nbs with novel innate effector functions.
A

ƒƒ Tools need to be developed for obtaining cell-specific and cell-penetrating Nbs for intracellular delivery of therapeutics.
Nbs in research
ƒƒ Nbs are excellent capturing agents to purify targets from complex mixtures or chromatin immunoprecipitation.
ƒƒ Nbs are key molecules as crystallization chaperones.
ƒƒ Nbs are useful tools for tracing and immunomodulation of intracellular antigens at the highest resolution.
Nbs in diagnosis
ƒƒ Nbs have a future as minimal-sized antigen-specific probes in µ-arrays and innovative biosensors.
ƒƒ Nbs are readily labeled and their fast renal clearance makes them ideal tools for noninvasive in vivo imaging.
Nbs for therapy
ƒƒ The very fast blood clearance of Nbs emphasizes the need for technologies to increase the blood half-life of Nbs for therapeutic
purposes.
ƒƒ The small, monomeric format of Nbs makes them a perfect plug-and-play tool to generate bispecific Nbs or larger pluripotent
constructs with an adaptable serum half-life, that are able to combat envenoming with small toxins, viral, bacterial or parasite
infections, and various pathologies.
ƒƒ Generation of Nbs that cross the blood–brain barrier, the attachment of Nbs to lentiviral particles or even on the surface of bacteria, or
creation of Nbs that are secreted by bacteria will create novel therapeutic opportunities.

future science group www.futuremedicine.com 11


Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

13 Jahnichen S, Blanchetot C, Maussang D et al. affinity predictions of a protein-protein


References CXCR4 nanobodies (VHH-based single interaction using multivariate experimental
1 Gill DS, Damle NK. Biopharmaceutical drug variable domains) potently inhibit chemotaxis design. J. Biol. Chem. 277(33), 29897–29907
discovery using novel protein scaffolds. Curr. and HIV-1 replication and mobilize stem cells. (2002).
Opin. Biotechnol. 17(6), 653–658 (2006). Proc. Natl Sci. USA 107(47), 20565–20570 25 Esvelt KM, Carlson JC, Liu DR. A system
2 Hamers-Casterman C, Atarhouch T, (2010). for the continuous directed evolution of
Muyldermans S et al. Naturally-occurring 14 Koch-Nolte F, Reyelt J, Schoessow B et al. biomolecules. Nature 472(7344), 499–503
antibodies devoid of light-chains. Nature Single domain antibodies from llama (2011).
363(6428), 446–448 (1993). effectively and specifically block T cell nn Introduces an elegant and powerful
3 Flajnik MF, Deschacht N, Muyldermans S. ecto‑ADP-ribosyltransferase ART2.2 in vivo.
technology for a phage-assisted continuous
A Case of convergence: why did a simple FASEB J. 21(13), 3490–3498 (2007).
directed evolution of genes linked to
alternative to canonical antibodies arise in 15 Janssens R, Dekker S, Hendriks RW et al. protein production in Escherichia coli.
sharks and camels? PLoS Biology 9(8), Generation of heavy-chain-only antibodies in
e1001120 (2011). 26 van den Berg A, Dowdy SF. Protein
mice. Proc. Natl Sci. USA 103(41),
transduction domain delivery of therapeutic
4 De Genst E, Saerens D, Muyldermans S, 15130–15135 (2006).
macromolecules. Curr. Opin. Biotechnol.
Conrath K. Antibody repertoire development 16 Pellis M, Pardon E, Zolghadr K et al. A 22(6), 888–893 (2011).
in camelids. Devel. Comp. Immunol. 30(1–2), bacterial-two-hybrid selection system for one-
187–198 (2006). 27 Mcnaughton BR, Cronican JJ, Thompson
step isolation of intracellularly functional
DB, Liu DR. Mammalian cell penetration,

f
5 Cortez-Retamozo V, Backmann N, Senter PD Nanobodies. Arch. Biochem. Biophys. 526(2),
siRNA transfection, and DNA transfection
et al. Efficient cancer therapy with a 114–123 (2012).

oo
by supercharged proteins. Proc. Natl Sci.
nanobody-based conjugate. Cancer Res. 64(8), 17 Reddy ST, Ge X, Miklos AE et al. Monoclonal USA 106(15), 6111–6116 (2009).
2853–2857 (2004). antibodies isolated without screening by
28 Blanco-Toribio A, Muyldermans S, Frankel
6 Stijlemans B, Caljon G, Natesan SKA et al. analyzing the variable-gene repertoire of plasma
G, Angel Fernandez L. Direct injection of
High affinity nanobodies against the cells. Nat. Biotechnol. 28(9), 965–969 (2010).
functional single-domain antibodies from
trypanosome brucei vsg are potent Direct high-throughput sequencing of V gene
nn
E. coli into human cells. PLoS ONE 5(12),
trypanolytic agents that block endocytosis.
PLoS Pathogens 7(6), e1002072 (2011).
Pr
repertoires has been used to bypass tedious e15227 (2010).
library generation and panning, in an effort nn Injecting nanobodies (Nbs) into
7 Vincke C, Gutiérrez C, Wernery U, Devoogdt to identify affinity-matured scFvs.
mammalian cells using nonpathogenic
N, Hassanzadeh-Ghassabeh G, Muyldermans
18 Ravn U, Gueneau F, Baerlocher L et al. bacterial strains carrying a T3SS is a
S. Generation of single domain antibody
By‑passing in vitro screening-next generation promising technology for in vitro and
fragments derived from camelids and
sequencing technologies applied to antibody in vivo intrabody applications that
generation of manifold constructs. Methods
or

display and in silico candidate selection. Nucleic target host cell functions and signaling
Mol. Biol. 907, 145–176 (2012).
Acids Res. 38(21), e193 (2010).
pathways.
8 Monegal A, Ami D, Martinelli C et al.
19 Hassanzadeh-Ghassabeh G, Saerens D,
Immunological applications of single-domain 29 Verheesen P, Ten Haaft MR, Lindner N,
Muyldermans S. Generation of anti‑infectome/
llama recombinant antibodies isolated from a Verrips CT, De Haard JJW. Beneficial
anti-proteome nanobodies. Methods Mol. Biol.
h

naive library. Protein Eng. Des. Sel. 22(4), properties of single-domain antibody
790, 239–259 (2011).
273–280 (2009). fragments for application in immunoaffinity
20 Abbady AQ, Al-Daoude A, Al-Mariri A, purification and immuno-perfusion
9 Koide A, Tereshko V, Uysal S, Margalef K,
ut

Zarkawi M, Muyldermans S. Chaperonin chromatography. Biochim. Biophys. Acta


Kossiakoff AA, Koide S. Exploring the
GroEL a Brucella immunodominant antigen 1624(1–3), 21–28 (2003).
capacity of minimalist protein interfaces:
identified using nanobody and MALDI-TOF-
interface energetics and affinity maturation to 30 Rothbauer U, Zolghadr K, Muyldermans S,
MS technologies. Vet. Immunol. Immunopathol.
picomolar K-D of a single-domain antibody Schepers A, Cardoso MC, Leonhardt H.
146(3–4), 254–263 (2012).
A versatile nanotrap for biochemical and
A

with a flat paratope. J. Mol. Biol. 373(4),


941–953 (2007). 21 Digiandomenico A, Warrener P, Hamilton M functional studies with fluorescent fusion
et al. Identification of broadly protective proteins. Mol. Cell. Proteomics 7(2), 282–289
10 De Genst E, Silence K, Decanniere K et al.
human antibodies to Pseudomonas aeruginosa (2008).
Molecular basis for the preferential cleft
exopolysaccharide Psl by phenotypic screening.
recognition by dromedary heavy-chain 31 Adams H, Brummelhuis W, Maassen B et al.
J. Exp. Med. 209(7), 1273–1287 (2012).
antibodies. Proc. Natl Sci. USA 103(12), Specific immuno capturing of the
4586–4591 (2006). 22 Giordano RJ, Cardo-Vila M, Lahdenranta J, staphylococcal superantigen toxic-shock
Pasqualini R, Arap W. Biopanning and rapid syndrome toxin-1 in plasma. Biotechnol.
11 Davies J, Riechmann L. Camelising
ana­lysis of selective interactive ligands. Nat. Bioeng. 104(1), 143–151 (2009).
Human‑antibody fragments – NMR-studies
Med. 7(11), 1249–1253 (2001).
on Vh domains. FEBS Letts 339(3), 285–290 32 Klooster R, Maassen BTH, Stam JC et al.
(1994). 23 Klooster R, Eman Mr, Le Duc Q et al. Improved anti-IgG and HSA affinity ligands:
Selection and characterization of KDEL- Clinical application of VHH antibody
12 Vincke C, Loris R, Saerens D,
specific VHH antibody fragments and their technology. J. Immunol. Meth. 324(1–2),
Martinez‑Rodriguez S, Muyldermans S,
application in the study of ER resident protein 1–12 (2007).
Conrath K. General strategy to humanize a
expression. J. Immunol. Meth. 342(1–2), 1–12
camelid single-domain antibody and 33 Loris R, Marianovsky I, Lah J et al. Crystal
(2009).
identification of a universal humanized structure of the intrinsically flexible
nanobody scaffold. J. Biol. Chem. 284(5), 24 De Genst E, Areskoug D, Decanniere K, addiction antidote MazE. J. Biol. Chem.
3273–3284 (2009). Muyldermans S, Andersson K. Kinetic and 278(30), 28252–28257 (2003).

12 Nanomedicine (2013) 8(6) future science group


Nanobodies & their potential applications Perspective
34 Tereshko V, Uysal S, Koide A, Margalef K, antibodies from diverse libraries emphasizing blood–brain barrier. Br. J. Pharmacol. 165(7),
Koide S, Kossiakoff AA. Toward chaperone- dual functions of LMO2 protein interactions 2341–2353 (2012).
assisted crystallography: protein engineering using a single VH domain. J. Biol. Chem.
56 Kontermann RE. Strategies for extended
enhancement of crystal packing and x‑ray 286(5), 3707–3716 (2011).
serum half-life of protein therapeutics. Curr.
phasing capabilities of a camelid single- 46 De Clercq S, Zwaenepoel O, Martens E, Opin. Biotechnol. 22(6), 868–876 (2011).
domain antibody (V(H)H) scaffold. Protein Vandekerckhove J, Guillabert A, Gettemans J.
Sci. 17(7), 1175–1187 (2008). 57 Kubetzko S, Sarkar CA, Pluckthun A.
Nanobody-induced perturbation of LFA-1/L-
Protein PEGylation decreases observed target
35 Baranova E, Fronzes R, Garcia-Pino A et al. plastin phosphorylation impairs MTOC
association rates via a dual blocking
SbsB structure and lattice reconstruction docking, immune synapse formation and
mechanism. Mol. Pharmacol. 68(5),
unveil Ca 2+ triggered S‑layer assembly. Nature T cell activation. Cell. Mol. Life Sci. 70,
1439–1454 (2005).
487(7405), 119–122 (2012). 909–922 (2013).
58 Harmsen MM, Van Solt CB, Fijten HPD,
36 Rasmussen SGF, Choi H-J, Fung JJ et al. 47 Serruys B, Van Houtte F, Farhoudi-
Van Setten MC. Prolonged in vivo residence
Structure of a nanobody-stabilized active state Moghadam A, Leroux-Roels G,
times of llama single-domain antibody
of the beta(2) adrenoceptor. Nature Vanlandschoot P. Production,
fragments in pigs by binding to porcine
469(7329), 175–180 (2011). characterization and in vitro testing of
immunoglobulins. Vaccine 23(41),
37 Koide S. Engineering of recombinant HBcAg-specific VHH intrabodies. J. Gen.
4926–4934 (2005).
crystallization chaperones. Curr. Opin. Struct. Virol. 91, 643–652 (2010).
59 Tijink BM, Laeremans T, Budde M et al.
Biol. 19, 449–457 (2009). 48 Joshi SN, Butler DC, Messer A. Fusion to a
Improved tumor targeting of anti-epidermal

f
38 Dumoulin M, Last AM, Desmyter A et al. highly charged proteasomal retargeting
growth factor receptor nanobodies through
A camelid antibody fragment inhibits the sequence increases soluble cytoplasmic

oo
albumin binding: taking advantage of
formation of amyloid fibrils by human expression and efficacy of diverse anti-
modular nanobody technology. Mol. Cancer
lysozyme. Nature 424(6950), 783–788 synuclein intrabodies. Mabs 4(6), 686–693
Ther. 7(8), 2288–2297 (2008).
(2003). (2012).
60 Vugmeyster Y, Entrican CA, Joyce AP et al.
39 Domanska K, Vanderhaegen S, Srinivasan V 49 Caussinus E, Kanca O, Affolter M.
Pharmacokinetic, biodistribution and
et al. Atomic structure of a nanobody-trapped Fluorescent fusion protein knockout mediated
biophysical profiles of TNF nanobodies
domain-swapped dimer of an amyloidogenic
beta 2-microglobulin variant. Proc. Natl Sci.
Pr
by anti-GFP nanobody. Nat. Struct. Mol.
Biol. 19(1), 117–121 (2012).
conjugated to linear or branched
poly(ethyleen glycol). Bioconj. Chem. 23(7),
USA 108(4), 1314–1319 (2011). n Genetically encoded method for direct and 1452–1462 (2012).
40 Habicht G, Haupt C, Friedrich RP et al. fast depletion of target green fluorescent 61 Iqbal U, Trojahn U, Albaghdadi H et al.
Directed selection of a conformational protein fusions via the ubiquitin pathway in Kinetic ana­lysis of novel mono- and
antibody domain that prevents mature any eukaryotic system has been developed. multivalent VHH-fragments and their
or

amyloid fibril formation by stabilizing A beta 50 Sukhanova A, Even-Desrumeaux K, Kisserli application for molecular imaging of brain
protofibrils. Proc. Natl Sci. USA 104(49), A et al. Oriented conjugates of single-domain tumours. Br. J. Pharmacol. 160(4),
19232–19237 (2007). antibodies and quantum dots: toward a new 1016–1028 (2010).
41 Romer T, Leonhardt H, Rothbauer U. generation of ultrasmall diagnostic 62 Harrison RA, Cook DA, Renjifo C, Casewell
Engineering antibodies and proteins for nanoprobes. Nanomed. Nanotechnol. Biol. NR, Currier RB, Wagstaff SC. Research
h

molecular in vivo imaging. Curr. Opin. Med. 8(4), 516–525 (2012). strategies to improve snakebite treatment:
Biotechnol. 22(6), 882–887 (2011). 51 Pollithy A, Romer T, Lang C et al. challenges and progress. J. Proteomics 74(9),
42 Olichon A, Surrey T. Selection of genetically Magnetosome expression of functional 1768–1780 (2011).
ut

encoded fluorescent single domain antibodies camelid antibody fragments (nanobodies) in 63 Bouhaouala-Zahar B, Ben Abderrazek R,
engineered for efficient expression in magnetospirillum gryphiswaldense. Appl. Hmila I, Abidi N, Muyldermans S, El Ayeb
Escherichia coli. J. Biol. Chem. 282(50), Env. Microbiol. 77(17), 6165–6171 (2011). M. Immunological aspects of scorpion
36314–36320 (2007). 52 Vaneycken I, D’huyvetter M, Hernot S et al. toxins: current status and perspectives.
A

43 Ries J, Kaplan C, Platonova E, Eghlidi H, Immuno-imaging using nanobodies. Curr. Inflamm. Allergy Drug Targets 10(5), 358–
Ewers H. A simple, versatile method for GFP- Opin. Biotechnol. 22(6), 877–881 (2011). 368 (2011).
based super-resolution microspcopy via 53 Movahedi K, Schoonooghe S, Laoui D et al. 64 Hmila I, Saerens D, Ben Abderrazek R et al.
nanobodies. Nat. Methods 9(6), 582–584 Nanobody-based targeting of the macrophage A bispecific nanobody to provide full
(2012). mannose receptor for effective in vivo imaging protection against lethal scorpion
n Method for using any green fluorescent of tumor-associated macrophages. Cancer Res. envenoming. FASEB J. 24(9), 3479–3489
protein-tagged construct in single-molecule 72(16), 4165–4177 (2012). (2010).
super-resolution microscopy using green 54 Broisat A, Hernot S, Toczek J et al. n Bispecific Nbs specifically recognize one of
fluorescent protein-specific Nbs coupled to Nanobodies targeting mouse/human vcam1 each of the two most toxic proteins from
organic dyes is described. for the nuclear imaging of atherosclerotic scorpion venom, providing full protection
44 Jobling SA, Jarman C, Teh MM, Holmberg
lesions. Circ. Res. 110(7), 927–937 (2012). against lethal envenoming.
N, Blake C, Verhoeyen ME. n Radiolabeled Nbs against V-CAM1 have 65 Hmila I, Cosyns B, Tounsi H et al.
Immunomodulation of enzyme function in been developed for noninvasive detection of Pre-clinical studies of toxin-specific
plants by single-domain antibody fragments. atherosclerotic lesions. nanobodies: evidence of in vivo efficacy to
Nat. Biotechnol. 21(1), 77–80 (2003). prevent fatal disturbances provoked by
55 Caljon G, Caveliers V, Lahoutte T et al. Using
45 Tanaka T, Sewell H, Waters S, Phillips SEV, microdialysis to analyse the passage of scorpion envenoming. Toxicol. Appl.
Rabbitts TH. Single domain intracellular monovalent nanobodies through the Pharmacol. 264(2), 222–231 (2012).

future science group www.futuremedicine.com 13


Perspective Hassanzadeh-Ghassabeh, Devoogdt, De Pauw, Vincke, & Muyldermans

66 Wesolowski J, Alzogaray V, Reyelt J et al. 74 Coppieters K, Dreier T, Silence K et al. micelles for active drug targeting. J. Control.
Single domain antibodies: promising Formatted anti-tumor necrosis factor alpha Release 151(2), 183–192 (2011).
experimental and therapeutic tools in VHH proteins derived from camelids show 83 May C, Sapra P, Gerber H-P. Advances in
infection and immunity. Med. Microbiol. superior potency and targeting to inflamed bispecific biotherapeutics for the treatment
Immunol. 198(3), 157–174 (2009). joints in a murine model of collagen-induced of cancer. Biochem. Pharmacol. 84(9),
67 Rahbarizadeh F, Ahmadvand D, Sharifzadeh arthritis. Arthritis Rheum. 54(6), 1856–1866 1105–1112 (2012).
Z. Nanobody; an old concept and new vehicle (2006).
84 Baeuerle PA, Reinhardt C. Bispecific T-cell
for immunotargeting. Immunol. Invest. 40(3), 75 Roovers RC, Laeremans T, Huang L et al. engaging antibodies for cancer therapy.
299–338 (2011). Efficient inhibition of EGFR signalling and Cancer Res. 69(12), 4941–4944 (2009).
68 Broersens K, Rousseau F, Schymkowitz J. The of tumour growth by antagonistic anti-EGFR
85 Thakur A, Lum LG. Cancer therapy with
culprit behind amyloid beta peptide related nanobodies. Cancer Immunol. Immunother.
bispecific antibodies: clinical experience.
neurotoxicity in Alzheimer’s disease: oligomer 56(3), 303–317 (2007).
Curr. Opin. Mol. Ther. 12(3), 340–349
size or conformation? Alzheimers Res. Ther. 76 Roovers RC, Vosjan MJWD, Laeremans T (2010).
2(4), 12 (2010). et al. A biparatopic anti-EGFR nanobody
86 Saerens D, Hassanzadeh-Ghassabeh G,
69 Lafaye P, Achour I, England P, Duyckaerts C, efficiently inhibits solid tumour growth. Int.
Muyldermans S. Single-domain antibodies as
Rougeon F. Single-domain antibodies J. Cancer 129(8), 2013–2024 (2011).
building blocks for novel therapeutics.
recognize selectively small oligomeric forms of 77 Goyvaerts C, Robays L, De Groeve K et al. Curr. Opin. Pharmacol. 8(5), 600–608
amyloid beta, prevent Abeta-induced Targeting lentiviral vectors to dendritic cells (2008).

f
neurotoxicity and inhibit fibril formation. by the nanobody display technology. Human
87 Chames P, Baty D. Bispecific antibodies for
Mol. Immunol. 46(4), 695–704 (2009). Gene Therapy 21(10), 1440–1441 (2012).

oo
cancer therapy. The light at the end of the
70 De Genst E, Dobson CM. Nanobodies as 78 Hernot S, Unnikrishnan S, Du Z et al. tunnel? Mabs 1(6), 539–547 (2009).
structural probes of protein misfolding and Nanobody-coupled microbubbles as novel
88 Eshhar Z. Adoptive cancer immunotherapy
fibril formation. Methods Mol. Biol. 911, molecular tracer. J. Control. Release 158(2),
using genetically engineered designer T-cells:
533–558 (2012). 346–353 (2012).
first steps into the clinic. Curr. Opin. Mol.
71 Muruganandam A, Tanha J, Narang S, 79 Van De Broek B, Devoogdt N, D’Hollander Ther. 12(1), 55–63 (2010).
Stanimirovic D. Selection of phage-displayed
llama single-domain antibodies that
Pr
A et al. Specific cell targeting with nanobody
conjugated branched gold nanoparticles for
89 Baeuerle PA, Itin C. Clinical experience with
gene therapy and bispecific antibodies for T
transmigrate across human blood–brain photothermal therapy. ACS Nano 5(6),
cell-based therapy of cancer. Curr. Pharm.
barrier endothelium. FASEB J. 15(14), 4319–4328 (2011).
Biotechnol. 13(8), 1399–1408 (2012).
240–242 (2001). 80 Altintas I, Kok RJ, Schiffelers RM.
90 Bakhtiari SHA, Rahbarizadeh F, Hasannia
72 Li T, Bourgeois JP, Celli S et al. Cell- Targeting epidermal growth factor receptor
S, Ahmadvand D, Iri-Sofla FJ, Rasaee MJ.
or

penetrating anti-GFAP VHH and in tumors: from conventional monoclonal


Anti-MUC1 nanobody can redirect T-body
corresponding fluorescent fusion protein antibodies via heavy chain-only antibodies to
cytotoxic effector function. Hybridoma
VHH-GFP spontaneously cross the blood– nanobodies. Eur. J. Pharm. Sci. 45(4),
28(2), 85–92 (2009).
brain barrier and specifically recognize 399–407 (2012).
astrocytes: application to brain imaging. 81 Shishido T, Azumi Y, Nakanishi T et al.
h

FASEB J. 26(10), 3969–3979 (2012). Biotinylated bionanocapsules for displaying Websites


„„
73 Ulrichts H, Silence K, Schoolmeester A et al. diverse ligands toward cell-specific delivery.
101 Crescendo.
Antithrombotic drug candidate ALX-0081 J. Biochem. 146(6), 867–874 (2009).
ut

www.crescendo.com
shows superior preclinical efficacy and safety 82 Talelli M, Rijcken CJF, Oliveira S et al.
compared with currently marketed antiplatelet 102 Ablynx.
Nanobody–shell functionalized
drugs. Blood 118(3), 757–765 (2011). www.ablynx.com
thermosensitive core-crosslinked polymeric
A

14 Nanomedicine (2013) 8(6) future science group

View publication stats

Das könnte Ihnen auch gefallen