Vous êtes sur la page 1sur 15

Cancer Immunol Immunother (2007) 56:303–317

DOI 10.1007/s00262-006-0180-4

O RI G I NAL ART I C LE

Efficient inhibition of EGFR signalling and of tumour growth


by antagonistic anti-EGFR Nanobodies
Rob C. Roovers · Toon Laeremans · Lieven Huang ·
Severine De Taeye · Arie J. Verkleij ·
Hilde Revets · Hans J. de Haard ·
Paul M. P. van Bergen en Henegouwen

Received: 14 December 2005 / Accepted: 1 May 2006 / Published online: 30 May 2006
© Springer-Verlag 2006

Abstract The development of a number of diVerent Nanobodies that speciWcally competed for EGF bind-
solid tumours is associated with over-expression of ing to the EGFR were isolated from ‘immune’ phage
ErbB1, or the epidermal growth factor receptor Nanobody repertoires. The selected antibody frag-
(EGFR), and this over-expression is often correlated ments were found to eYciently inhibit EGF binding to
with poor prognosis of patients. Therefore, this recep- the EGFR without acting as receptor agonists them-
tor tyrosine kinase is considered to be an attractive tar- selves. In addition, they blocked EGF-mediated signal-
get for antibody-based therapy. Indeed, antibodies to ling and EGF-induced cell proliferation. In an in vivo
the EGFR have already proven their value for the murine xenograft model, the Nanobodies were eVec-
treatment of several solid tumours, especially in combi- tive in delaying the outgrowth of A431-derived solid
nation with chemotherapeutic treatment regimens. tumours. This is the Wrst report describing the success-
Variable domains of camelid heavy chain-only antibod- ful use of untagged Nanobodies for the in vivo treat-
ies (called Nanobodies™) have superior properties ment of solid tumours. The results show that functional
compared with classical antibodies in that they are phage antibody selection, coupled to the rational
small, very stable, easy to produce in large quantities design of Nanobodies, permits the rapid development
and easy to re-format into multi-valent or multi-spe- of novel anti-cancer antibody-based therapeutics.
ciWc proteins. Furthermore, they can speciWcally be
selected for a desired function by phage antibody dis- Keywords EGFR · Nanobody · Tumour · Therapy ·
play. In this report, we describe the successful selection Signalling
and the characterisation of antagonistic anti-EGFR
Nanobodies. By using a functional selection strategy,
Introduction

R. C. Roovers · A. J. Verkleij · The epidermal growth factor (EGF) receptor (EGFR,


Paul M. P. van Bergen en Henegouwen (&) or ErbB1) is a receptor tyrosine kinase (RTK) belong-
Department of Molecular Cell Biology,
ing to a family of four receptors (ErbB1 to 4). The
Institute of Biomembranes, Utrecht University,
Padualaan 8, 3584 Utrecht, CH, The Netherlands EGFR has an important role in the regulation of
e-mail: bergenp@bio.uu.nl growth and diVerentiation of a large number of diVer-
ent cell types (for review, see [55]). Many models also
T. Laeremans · S. De Taeye · H. J. de Haard
predict the EGFR to be at the convergence point of
Ablynx N.V., Technologiepark 4, 9052 Zwijnaarde, Belgium
several signal transduction pathways: e.g. the prolifera-
L. Huang · H. Revets tive signalling of diVerent G-protein coupled receptors
Laboratory of Cellular and Molecular Immunology, has been shown to be dependent on EGFR [11].
Department of Molecular and Cellular Interactions,
Because of the role of the EGFR in proliferation,
Flanders Interuniversity Institute for Biotechnology (VIB),
Free University of Brussels, Pleinlaan 2, cell-survival and angiogenesis, over-expression of the
1050 Brussels, Belgium receptor confers advantages to tumour cells at diVerent

123
304 Cancer Immunol Immunother (2007) 56:303–317

stages of tumour development (initiation, progression tumours [10] and for antibody-based therapy of cancer
and neo-vascularisation). Indeed, the EGFR is fre- [9]. Therefore, the Nanobody format provides a supe-
quently found to be over-expressed in a large number rior scaVold for the development of antibody-based
of epithelial tumours, including carcinomas of the head anti-cancer molecules (for review, see [42]).
and neck, breast, colon, lung, prostate, kidney, ovary, In this report, we describe the use of functional
brain, pancreas and bladder (reviewed in [26]). In addi- phage antibody selection using competitive elution with
tion, this over-expression is often correlated with poor the ligand EGF to develop antagonistic anti-EGFR
prognosis of patients [52]. For these reasons, the Nanobodies for cancer therapy. This selection for func-
EGFR constitutes an attractive target for cancer ther- tion resulted in the isolation of a panel of Nanobodies
apy. Two approaches are currently being undertaken that inhibited binding of EGF to its receptor without
to inhibit EGFR signalling, i.e. the development of acting as receptor agonists themselves. These Nano-
small molecule inhibitors of the intra-cellular tyrosine bodies performed excellently in vitro in inhibiting
kinase and the isolation of monoclonal antibodies EGF-induced signalling and EGF-induced cell prolifer-
directed to the extra-cellular domain of the EGFR ation, and they were eYcient in inhibiting tumour out-
[38, 45]. In recent years, several antibodies directed to growth in an in vivo model for solid tumours. These
the EGFR, such as Cetuximab (Erbitux) [24, 47], results show the great potential of combining functional
EMD72000 and ABX-EGF [17, 46], have already been phage antibody selection strategies with the favourable
successfully introduced into pre-clinical and clinical characteristics of Nanobodies for the development of
development. antibody-based cancer therapeutics.
Heavy-chain antibodies (HcAb’s) have been described
in species belonging to the family of camilidae (i.e.
Dromedary, Camel and Llama) as a second class of Materials and methods
antibodies next to the conventional (four-chain) anti-
body repertoire [21]. Since these antibodies are com- Cell lines
posed of two identical heavy chains and are devoid of
light chains, their antigen-binding part is composed of Human epidermoid squamous carcinoma cell line
only one single immunoglobulin (Ig) variable region A431 [19], carrying an ampliWcation of the EGFR gene
(termed VHH, or Nanobody™). These antigen-speciWc [36], was obtained from the ATCC (cat. nr. CRL-
antibody fragments have many inherent, favourable 1555). NIH 3T3, clone 2.2 murine Wbroblasts were
characteristics, such as high solubility and the capacity selected for low endogenous EGFR expression [27].
to refold after denaturation while retaining their bind- Her14 cells are derived from NIH 3T3 Wbroblasts and
ing capacity [14, 16]. In addition, the genes encoding stably express roughly 105 copies of the human EGFR
these fragments can easily be engineered to obtain on their cell surface [27]. The cervix adenocarcinoma
multi-valent and multi-speciWc formats, or can be re- cell line Hela was obtained from the ATCC (cat. nr.
cloned as fusion to other (eVector) proteins (for CCL-2). All cells were cultured in Dulbecco’s ModiW-
review, see [39]). And since these fragments are com- cation of Eagle medium (DMEM: Gibco, Invitrogen,
posed of only a single Ig fold, they do not have the Paisley, UK) containing 7.5% (v/v) foetal calf serum
disadvantage of partial unfolding, thereby exposing and 2 mM L-glutamine in a humidiWed atmosphere at
hydrophobic patches, as have single-chain Fv (scFv) 37°C under 5% CO2. Cells were regularly tested for the
fragments. Furthermore, the absence of protease- presence of mycoplasma and consistently found to be
sensitive linker sequences makes them more stable mycoplasma-free.
than scFv’s.
Ever since the Wrst description of phage display of Immunisation of Llama glama with EGFR-containing
peptides [50] and of antibody fragments [35], this tech- cell preparations and construction of phage Nanobody
nique has been successfully used to isolate antibodies repertoires
against a broad range of antigens (for review, see [28,
29]). Because of the ease of cloning Nanobody-encod- All animal experiments were conducted with the
ing genes, they can easily (and eYciently) be displayed approval of the Ethical Committee of the Faculty of
on Wlamentous phage. Indeed, phage display of Nano- Veterinary Medicine (University of Ghent, Belgium).
bodies has already proven to yield speciWc antibody To induce a humoral immune response directed
fragments directed to both hapten- and protein anti- towards the EGFR in Llama glama, animals were
gens [1, 32]. Finally, Nanobodies have already been injected with EGFR-containing cell preparations. Two
shown to be potent vehicles for the targeting of animals were injected with intact human A431 cells

123
Cancer Immunol Immunother (2007) 56:303–317 305

(approximately 108 cells per injection), while A431- culture plates at roughly 10,000 cells per well and cul-
derived membrane vesicles (prepared from the same tured overnight. The next day, cells were washed with
number of cells, according to the method described PBS, Wxed with 4% (w/v) formaldehyde in PBS for
by Cohen et al. [8]) were administered to two other 30 min at room temperature (RT) and non-reacted
llamas. Each animal received seven doses of subcuta- Wxative was quenched with 100 mM glycine in PBS for
neously administered antigen at weekly intervals. Pre- 10 min at RT. Non-speciWc binding was prevented by
immune and immune sera were collected at days 0 blocking with 2% skimmed milk powder (Marvell) in
(before immunisation), and after 4 and 6 weeks of PBS (2% MBPS) for 30 min at RT and serial dilutions
immunisation. Four days after the last antigen injec- of pre-immune and immune sera were added in 2%
tion, a 150 ml blood sample was collected, and periferal MPBS. All further incubations were carried out for 1 h
blood lymphocytes (PBLs) were puriWed by density at RT and after every incubation, plates were washed
gradient centrifugation on Ficoll-Paque™ PLUS gradi- four times with PBS. Detection of bound antibody was
ents (Amersham Biosciences, Little Chalfont, UK), performed by incubation with a rabbit anti-llama Ig
resulting in the isolation of approximately 108 PBLs. antiserum (1:1,000 in 2% MPBS) and peroxydase-con-
As an alternative source of B-cells, a small biopsy was jugated donkey anti-rabbit Ig (1:5,000 in 2% MPBS;
taken from the lymph node draining the site of immu- Jackson Immunoresearch, West Grove, PA, USA).
nisation. Total RNA was extracted from these tissues Substrate used was o-phenylenediamine and the OD of
as described [6] and transcribed into cDNA using an the resulting stain was measured at 490 nm. PuriWed
oligo-dT primer and the SuperScript III First-Strand EGFR was purchased from Sigma-Aldrich (Zwijn-
Synthesis System for RT-PCR (Invitrogen, Carlsbad, drecht, The Netherlands) and coated overnight at 4°C
CA, USA) according to the manufacturers’ recommen- at 50 ng/ml in PBS in ELISA plates (Nunc, Rochester,
dations. Next, cDNA was treated with RNAse H to MN, USA). Next day, plates were washed with PBS,
deplete for residual RNA prior to puriWcation with the blocked with 2% MPBS and the assay was performed
QIAquick PCR PuriWcation Kit (Qiagen, Hilden, Ger- as described above.
many). The puriWed cDNA was then used as template
to amplify the repertoire of Ig heavy chain-encoding Selection of antagonistic Nanobody fragments
gene segments with the use of a framework 1 (FR1) by ligand-speciWc elution
speciWc primer and an oligo-dT primer. This ampliWca-
tion procedure introduces a SWI restriction site at the 5⬘ To select Nanobody fragments that would eVectively
end of FR1 and results in PCR fragments of approxi- compete with ligand (EGF) binding to the EGFR, the
mately 1.6 kb (representing conventional IgGs) and method of competitive elution [37] was employed.
fragments of 1.3 kb (comprising heavy-chain IgGs that BrieXy, A431-derived membrane vesicles were coated
lack a CH1 domain). The two classes of heavy chain- to 96 wells immunosorp plates (Costar, Corning, NY,
encoding genes were then size-separated on agarose USA) at 5 g/ml overnight at 4°C. Phage (approxi-
gels and genes encoding heavy-chain only IgG were mately 1010 colony forming units (cfu)), prepared from
puriWed. Since a BstEII restriction site naturally occurs the ‘immune’ libraries as described [34], were then
in approximately 90% of the FR4 of Nanobody genes, panned for binding to immobilised EGFR. After
the repertoire of PCR-ampliWed genes was cut with SWI extensive washing with PBS, phage that could compete
and BstEII and the resulting 300–400 bp cDNA frag- for EGF binding were eluted by the addition of 1 mM
ments were puriWed by gel electrophoresis. cDNA of EGF (Molecular Probes, Invitrogen) for 15 min at
fragments were Wnally ligated in a phagemid vector for RT. Displaced phage were used to infect exponentially
display on Wlamentous bacteriophage [12] and electro- growing E. coli TG1 and bacteria were plated on LB
transformed to Escherichia coli TG1 (K12, (lac-pro), agar plates containing 2% (w/v) glucose and 100 g/ml
supE, thi, hsdD5/F’traD36, proA+B+, lacIq, lacZM15). ampicillin.
This resulted in ‘immune’ Nanobody repertoires of
approximately 107 transformants each. Re-cloning, expression and puriWcation of anti-EGFR
Nanobody fragments
Testing pre-immune and immune llama sera
for the presence of anti-EGFR antibodies Expression of recombinant Nanobody fragments in the
by (cell-)ELISA periplasm of E. coli and puriWcation by means of immo-
bilised metal ion aYnity chromatography (IMAC)
Her14 cells and NIH 3T3 clone 2.2 cells were seeded in were performed as has been described for Fab anti-
gelatine-coated (0.25% (w/v) in PBS) 96 wells tissue body fragments [43]. In order to obtain biotinylated

123
306 Cancer Immunol Immunother (2007) 56:303–317

Nanobodies, Nanobody-encoding gene segments were Mannheim, Germany) and blots were stained for the
re-cloned as SWI-BstEII fragments in vector pUR5850 EGFR as described below.
[54]. This vector allows expression of c-Myc- and His6-
tagged protein in the periplasmic space of E. coli and it Inhibition of EGF binding to EGFR by selected
adds a biotinylation sequence (LRSIFEAQKMEW) Nanobodies and functionality of the trivalent,
between the c-Myc and His6 tag. The lysine in this bispeciWc format
sequence is biotinylated upon expression of the Nano-
body in an E. coli strain that over-expresses the BirA To determine whether the selected EGFR-speciWc
gene (AVB101; Avidity, Denver, CO, USA). Bi- and Nanobodies were able to prevent binding of EGF to
trivalent Nanobody constructs were synthesised essen- the EGFR, a competition ELISA was designed. A431-
tially as described [15]. However, Nanobody fragments derived membrane vesicles (5 g/ml, as determined by
were separated by the N-terminal part of the long the bicinchoninic acid (BCA) protein assay; Perbio Sci-
hinge region found in IgG3 HcAb’s, followed by ence, Etten-Leur, The Netherlands) were immobilised
three alanine residues encoded by the Not1 site overnight at 4°C in 96 wells immunosorp plates
(EPKTPKPQPAAA). To make trivalent, bispeciWc (Costar, Corning, NY, USA). Plates were washed with
Nanobody constructs, the construct containing a biva- PBS containing 0.05% (v/v) Tween-20 and subse-
lent Nanobody fragment in vector pAX011 was PstI- quently blocked with PBS containing 1% (w/v) casein
digested. After agarose gel electrophoresis and subse- for 2 h at RT. After washing, equal volumes of 8 ng/ml
quent gel extraction, the 400 bp PstI fragment (1.6 nM) biotinylated EGF (Peprotech, New York,
(corresponding to one Nanobody-encoding gene fol- NY, USA) mixed with serial dilutions of puriWed solu-
lowed by the linker) was ligated in the PstI site of ble Nanobody (in Wnal concentrations of 160 nM to
pAX011 containing the anti-mouse serum albumin 9 pM) were simultaneously added in PBS containing
(MSA) Nanobody MSA21 (a kind gift of R. Klooster, 0.1% (w/v) casein and incubated for 1 h at room
Utrecht University, The Netherlands). The latter con- temperature. Receptor-bound EGF-biotin was Wnally
struct was obtained by ligation of MSA21 as a PstI- detected with an extravidine-alkaline phosphatase
BstEII fragment in pAX011. This procedure allows conjugate (Sigma-Aldrich, Zwijndrecht, The Nether-
cloning of (repeating units of) Nanobody-linker encod- lands), followed by staining using para-nitrophenyl
ing segments amino-terminally to the MSA21 Nano- phosphate (PNP).
body. To demonstrate the simultaneous reactivity of the
two antigen speciWcities present in the bispeciWc, triva-
Immuno-precipitation of EGFR using biotinylated lent Nanobody constructs, a sandwich ELISA was per-
Nanobody formed. Coating of A431 vesicles and blocking were as
described above. 10 nM of the trivalent, bispeciWc mol-
Hela cells were seeded at 800,000 cells per dish in Ø ecules was then incubated for an hour at RT in PBS
10 cm tissue culture dishes (Costar, Corning, NY, containing 0.1% casein and bound Nanobody was
USA) and grown for 24 h. Total cell lysates were pre- detected with biotinylated MSA (100 nM in PBS con-
pared in 750 l lysis buVer (50 mM Tris–HCl, pH 7.5; taining 0.1% casein) and alkaline phosphatase-coupled
150 mM NaCl; 5 mM Na-EDTA, pH 8.0; 1% (v/v) Tri- extravidin as described earlier.
ton X100), containing a mix of protease-inhibitors
(Complete™, EDTA-free: Roche, Mannheim, Ger- Staining of diVerent cell lines by Xuorescein-labelled
many) by scraping the cells oV the plate. Nuclei were Nanobodies using FACS
spun down (14,000 rpm, 5 min, 4°C) and an aliquot of
the lysate was set apart. Streptavidin-coated agarose PuriWed, recombinant, monovalent Nanobodies (approx-
beads (50 l per precipitation; Uptima (Interchim), imately 200 g per reaction) were labelled with Xuo-
Montluçon, France) were washed twice with lysis rescein using a commercially available Xuorescein
buVer and saturated with biotinylated Nanobody labelling kit (Roche, Mannheim, Germany). Fluores-
(10 g per precipitation). Beads were washed again cence-labelled Nanobody (100 nM) was then used to
with lysis buVer twice, added to the cellular lysate and stain EGFR-expressing (Her-14 and A431) cell lines
incubated for 1 h at 4°C with gentle mixing. Beads and the EGFR-negative cell line 3T3 clone 2.2. Stain-
were washed four times with lysis buVer and boiled in ing was performed on live, non-Wxed cells for 30 min
2£ Laemmli sample buVer [30]. Proteins were then at 4°C in DMEM. Excess Nanobody was then
size-separated on 8% (w/v) poly-acrylamide gels removed by washing with Dulbecco’s balanced salt
(SDS-PAGE), blotted to PVDF membrane (Roche, solution (four times) and cells were analyzed on a

123
Cancer Immunol Immunother (2007) 56:303–317 307

FACSVantage Xuorescence-activated cell sorter cells were serum-starved overnight. Next day (day 0),
(Becton & Dickinson, San Jose, CA, USA). dilutions of bispeciWc, trivalent Nanobodies (0, 1, 10,
100 and 1,000 nM) were mixed with 8 nM of EGF in
Activation of the EGFR and inhibition medium containing 0.1% FCS and mixtures were
of EGF-induced EGFR activation added to the cells in hexa-duplicate. After 4 days of
by multi-valent anti-EGFR Nanobodies growth, total cellular protein was precipitated by the
addition of 5% (w/v) of trichloro-acetic acid (TCA)
Her14 cells (approximately 105 cells per well) were and stained with sulpho-rhodamine B (SRB) as
seeded in 12-wells tissue culture clusters (Costar, Corn- described [49]. OD was read at 540 nm and the num-
ing, NY, USA) and allowed to adhere. After 8 h, cells ber of cells was measured relative to the number at
were rinsed once with DMEM containing 0.1% (v/v) day 0. In addition, a bio-assay was used in which cell-
FCS and serum-starved overnight in the same medium. proliferation was determined by measuring the
The day of the assay, medium was refreshed with the uptake of radio-active thymidine: A431 cells were
same medium containing 1% (w/v) BSA and EGF seeded at approximately 4,000 cells/well in 100 l ITS
(50 ng/ml, corresponding to approximately 8 nM) or medium (DMEM supplemented with insulin/transfer-
trivalent, bispeciWc Nanobodies (1 M) were added at ring/selenium; Gibco, Invitrogen) in 96-wells tissue
37°C. After 15 min of incubation, cells were quickly culture clusters and incubated overnight. After 24 h,
cooled down on ice and washed twice with ice-cold the medium was refreshed and dilutions of Nanobod-
PBS. Total cell lysates were prepared by scraping the ies (0, 1, 10, 100 and 1,000 nM) were added together
cells oV the plate in 50 l 2£ Laemmli protein sample with 1 ng/ml (corresponding to approximately
buVer and by boiling for 5 min at 100°C. Proteins were 170 pM) EGF. After 2 days of incubation, cells were
size-separated on 8% (w/v) poly-acrylamide gels and pulsed with 1–2 Ci [3H]-thymidine and incubated for
blotted to PVDF membrane (Roche, Mannheim, an additional 16 h, prior to freezing at ¡20°C. Cells
Germany). Blots were then stained for the total amount were subsequently thawed and embedded on glass
of EGFR with a rabbit polyclonal antiserum to the Wber membranes using a cell harvester (Perkin Elmer
receptor (Santa Cruz Biotechnology, Santa Cruz, CA, Life Sciences, Wellesley, MA, USA). After several
USA) and for phosphorylated receptor using a mouse washing with MiliQ-grade water, Wlters were air-dried
monoclonal anti-EGFR phospho-tyrosine 1068 anti- and counted using a -counter (Perkin Elmer Life Sci-
body (Cell Signalling, Beverly, MA, USA), followed by ences).
the respective peroxidase-conjugated secondary anti-
bodies (donkey anti-rabbit and donkey anti-mouse; EYcacy of trivalent, bispeciWc anti-EGFR Nanobodies
Jackson Immunoresearch, West Grove, PA, USA). As in in vivo tumour therapy
a loading control, the lower parts of the blots were
stained for actin with a monoclonal anti-actin antibody BispeciWc, trivalent Nanobody fragments were
(ICN Biomedicals, Irvine, CA, USA). Bound antibody expressed and endotoxin puriWed. At day 0, 1 day
was visualised by enhanced chemoluminescence using before the start of therapy, the left Xank of each mouse
Western Lightning™ substrate (Perkin Elmer Life Sci- was subcutaneously injected with 107 A431 carcinoma
ences, Wellesley, MA, USA). To measure the capacity cells. Eight female athymic (nude) mice (NMRI: nu/
of the diVerent bispeciWc, trivalent anti-EGFR Nano- nu) were used per treatment. As a placebo control,
bodies to inhibit EGF-induced signalling, dilutions of mice were treated with PBS; antibody fragments and
Nanobodies (1,000, 100, 10 or 1 nM) were mixed with placebo were administered intra-peritoneally. Animals
8 nM of human EGF and these were added to serum- were treated twice a week at indicated days (1, 4, 8, 11,
starved Her-14 cells for 15 min. EGF receptor phos- 15, 18, 22 and 25) with 1 mg of Nanobody per mouse
phorylation was then measured by means of Western and per injection, independent of body weight.
blotting as described earlier. Tumour growth was monitored twice a week, starting
at day 4 by measuring the two perpendicular diameters
Inhibition of EGF-induced cell proliferation of the tumour with a vernier caliper. When tumour
by anti-EGFR Nanobodies mass exceeded 10% of body weight, mice were sacri-
Wced. Tumour volume (in cm3) was calculated as
Her14 cells were seeded at 1,000 cells/well in 96 wells (/6)ab2, where a and b are length and width (in cm) of
tissue culture clusters (Costar, Corning, NY, USA) the tumour, respectively, and a ¸ b. The mean relative
and grown for 2 days to mid-log phase. Medium was tumour volume (RTV) for each group of mice was fol-
then replaced by medium containing 0.1% FCS and lowed for 37 days.

123
308 Cancer Immunol Immunother (2007) 56:303–317

Results and EGFR-expressing A431 or Her14 cells. Approxi-


mately 30% of the clones tested were found to react
Induction of a humoral anti-EGFR response with EGFR-positive cells and not to show reactivity
in Llama glama towards EGFR-negative cells (data not shown). DNA
Wngerprinting using the restriction enzyme Hinf1 was
To obtain antagonistic Nanobodies speciWc for the then used to identify possibly diVerent Nanobodies and
EGF receptor, we used phage antibody display with these were grouped according to their restriction
competitive elution [37] using the ligand EGF. As an proWle. Representative clones from each group were
optimally rich source of such antibodies, ‘immune’ re-tested in ELISA using puriWed phage. A strong
repertoires [7] of phage-displayed Nanobodies were reactivity of Nanobody Ia1 was found towards EGFR-
constructed. Llama glama were immunised with the positive cell line Her14, whereas the signal on EGFR-
human epidermoid carcinoma cell line A431 [19], con- negative cell line 3T3 clone 2.2 was negligible (Fig. 2a).
taining an ampliWcation of the EGFR gene [36] and No reaction was observed for control phage that did
thus a high expression of the protein (roughly 106 not express any Nanobody. These results provide proof
receptor molecules per cell), or with A431-derived of the successful isolation of speciWc anti-EGFR Nano-
membrane vesicles, made according to the method bodies. Because in this ELISA assay the cells were
described by Cohen et al. [8]. The induction of a Wxed prior to antibody incubation, the selected Nano-
humoral immune response was followed by testing sera bodies were also tested for their reactivity with surface-
of the animals before and after immunisation by exposed EGFR on non-Wxed, living cells by means of
(whole cell-) ELISA. Both immunisation strategies FACS staining. FITC was directly conjugated to puri-
resulted in the induction of a speciWc response towards Wed Nanobodies and labelled proteins were tested in
the EGFR (Fig. 1). When whole, live A431 cells were FACS staining of EGFR-positive and -negative cells.
used for immunisation, the induced antibody response As was found in whole cell-ELISA, selected antibody
was also directed towards other cell surface-exposed fragments were reactive with the EGFR-expressing
epitopes than the EGFR, witnessed by the reactivity of cell lines, A431 and Her14, and not with the EGFR-
immune sera with the EGFR-negative cell line 3T3, negative cell line 3T3 2.2 (shown for Nanobody Ia1 in
clone 2.2 (Fig. 1a, b). Immunisation with A431-derived Fig. 2b). Finally, to prove the EGFR-speciWcity of the
membrane vesicles resulted in a more speciWc response selected Nanobodies by a diVerent method, immuno-
towards the EGFR (Fig. 1c, d), although the titer of precipitations were performed using site-speciWcally
anti-EGFR antibodies in these immune sera seemed to biotinylated Nanobodies and streptavidin-coated aga-
be slightly lower (compare Fig. 1a with c). In addition, rose beads. The cell lysates prepared from Hela cells
immune sera were also reactive with puriWed EGF that were used for the diVerent immuno-precipitations
receptor, which is shown for immune sera taken from all contained the same quantity of EGFR (Fig. 2c, left
animals that were immunised with whole cells panel). Nanobody Ia1 eYciently precipitated the
(Fig. 1e). These data clearly demonstrate the successful EGFR, resulting in a strong enrichment of the receptor
induction of a humoral immune response towards the (Fig. 2c, right panel). Empty streptavidin beads, or
EGFR. beads loaded with a control (anti-GST) Nanobody
failed to precipitate the EGFR (Fig. 2c). Results for
Selection and characterisation of antagonistic the diVerent anti-EGFR Nanobodies isolated were
Nanobodies similar (data not shown).
Since the obtained Nanobodies were speciWcally
Phage Nanobody repertoires were then synthesised by selected for their ability to compete for EGF binding to
RT-PCR from two diVerent lymphoid sources obtained the EGFR, their potency in inhibiting binding of EGF
from immunised animals: peripheral blood lympho- to the receptor was tested. A431-derived membrane
cytes (PBLs) and a biopsy from a lymph node draining vesicles were immobilised and the binding of biotinyla-
the site of immunisation. This resulted in libraries of ted EGF to immobilised EGFR was detected in the
approximately 107 transformants each. These were presence of an increasing amount of puriWed, monova-
then panned to immobilised, A431-derived membrane lent Nanobody. The three Nanobodies that most
vesicles (coated to microtiter plates) and Nanobodies eYciently blocked EGF binding to the EGFR (named
competing with EGF binding were speciWcally eluted Ia1, IIIa3 and L2–3.40; shown in Fig. 3a) were selected
with a pulse of human EGF. After two rounds of selec- for further characterisation. These selected Nanobod-
tion, single clones were screened for EGFR reactivity ies inhibited binding of EGF to the receptor with IC50
by whole cell-ELISA on EGFR-negative 3T3 2.2 cells values in the low nanomolar range (approximately

123
Cancer Immunol Immunother (2007) 56:303–317 309

A 1.4 day 0 B 1.4 day 0


A431 3T3 2.2
1.2 day 28 1.2 day 28
1 day 42 1 day 42

OD 490nm
OD 490nm
0.8 0.8
0.6 0.6
0.4 0.4
0.2 0.2
0 0
100 1000 10000 100000 1000000 100 1000 10000 100000 1000000
serum dilution serum dilution

C 0.6 day 0
A431 D 0.6
3T3 2.2 day 0
day 28
0.5 0.5 day 28
day 42
day 42
0.4
OD 490nm

0.4

OD 490nm
0.3 0.3
0.2 0.2
0.1 0.1

0 0
100 1000 10000 100000 1000000 100 1000 10000 100000 1000000
serum dilution serum dilution

E 0.7 Purified EGFR day 0


0.6 day 42
0.5
OD 490nm

0.4
0.3
0.2
0.1
0
100 1000 10000 100000 1000000
serum dilution

Fig. 1 Immunisation of Llama glama with EGFR-containing cell derived membrane vesicles (c, d) towards A431 cells (a, c), to-
preparations induces a strong anti-EGFR humoral immune wards EGFR-negative 3T3 2.2 cells (b, d) and towards puriWed
response. The reactivity of pre-immune (day 0: diamonds) and EGF receptor (e) was determined by whole cell ELISA (a–d) or
immune sera (day 28: squares and day 42: triangles) of animals conventional ELISA (e) as described in Materials and methods
immunised with whole, intact A431 cells (a, b, e), or with A431-

5 nM for the Ia1 and L2–3.40 Nanobody and 20 nM for sequence: the N-terminal part of the long hinge region
the IIIa3 Nanobody: Fig. 3a). When tested for their found in IgG3 HcAb’s (EPKTPKPQP). PuriWed,
epitope-speciWcity, none of these three Nanobodies multi-valent proteins (Fig. 3c) were then tested for
competed with the whole monoclonal anti-EGFR anti- their potency to block EGF binding to the receptor.
body Erbitux for binding to the EGFR, but they recog- The addition of a second antigen-binding site to the
nised overlapping epitopes on the receptor, as judged same molecule decreased the IC50 value with an order
by competition ELISA (data not shown). of magnitude, as is shown for the Ia1 Nanobody in
To investigate the eVect of increased apparent aYn- Fig. 3b (an IC50 value of approximately 5 nM for
ity (through avidity) on the capacity of the selected monovalent Ia1 and 0.5 nM for the bivalent molecule).
Nanobodies to block EGF binding to the receptor, However, addition of a third Nanobody to the mole-
bi- and trivalent molecules were synthesised. Two, or cule did not signiWcantly further decrease the IC50
three Nanobody-encoding genes were re-cloned in- value for inhibition of EGF binding. Similar results
frame and expressed as a single molecule (Table 1). were obtained for the IIIa3 and L2–3.40 Nanobodies
The diVerent Nanobodies were separated by a Xexible (data not shown).

123
310 Cancer Immunol Immunother (2007) 56:303–317

A 1.6
1.4
1.2

OD 490nm
1
0.8
0.6
0.4
0.2
0
Control Ia1
3T3 2.2 Her14

B 3T3 2.2 Her14 A431

C Lysates Immunoprecipitation

MW (kDa)

170
Blot:
130
anti-EGFR
beads control Ia1 beads control Ia1
only only

Fig. 2 Selected Nanobodies speciWcally recognise the EGFR. Nanobody Ia1 (black line) or control Nanobody (grey Wll). c Im-
a The binding of anti-EGFR Nanobody Ia1 displayed on phage muno-precipitation (IP) of EGFR from Hela cell lysates using
and of control phage (not expressing any antibody fragment) to biotinylated anti-EGFR Nanobody Ia1. Lanes 1–3 total cell ly-
EGFR-negative cell line 3T3 2.2 and EGFR-expressing cell line sates used for IP. Lanes 4–6 IP with streptavidin beads only, with
Her14 was determined by cell-ELISA. Error bars indicate the an anti-glutathion S transferase (control) Nanobody and with the
standard deviation of three independent results. b FACS staining anti-EGFR Nanobody Ia1, respectively. Blots were stained for
of cell lines expressing EGFR (A431 and Her14) and of the EGFR as described in Materials and methods
EGFR-negative cell line 3T3 2.2 with FITC-coupled anti-EGFR

Re-formatting of Nanobodies for in vivo use of bivalent anti-EGFR Nanobodies, trivalent, bispec-
and in vitro characterisation of these formats iWc Nanobody constructs were designed. In these
molecules, two antigen-binding sites are directed
Monovalent Nanobodies have an in vivo half-life towards the EGFR and one Nanobody is speciWc
(T½) of approximately 1.5 h in blood [10]. Therefore, for mouse serum albumin (MSA21; Table 1). These
this Nanobody format (being a 15 kDa protein) is not 45–50 kDa proteins were Wrst shown to be able to simul-
optimal for the in vivo use in cancer treatment. Even taneously bind to immobilised EGFR and biotinylated
bi- or trivalent molecules (of approximately 30 and MSA in a sandwich-ELISA (Fig. 3d). In addition,
45 kDa, respectively) are below the renal threshold their serum half-life was determined and found to be
for Wrst-pass clearance and will therefore have a very signiWcantly longer than that of a control trimeric
short in vivo half-life. To increase the in vivo half-life Nanobody of the same size (T½ of approximately

123
Cancer Immunol Immunother (2007) 56:303–317 311

MW (kDa)

C 55

44
A 0.5

Ia1 33
0.4
IIIa3
L2-3.40
OD 490nm

0.3 control
background
24
0.2

0.1 17

0
0.001 0.01 0.1 1 10 100 1000
Nanobody concentration (nM) Mono- Bi- Trivalent

D 0.4
B 0.9 Ia1 monovalent
Ia1 bivalent 0.35
0.8
Ia1 trivalent
0.7 background 0.3
0.6
OD 490nm

0.25
OD 490nm

0.5
0.2
0.4
0.15
0.3
0.1
0.2
0.1 0.05

0 0
0.001 0.01 0.1 1 10 100 1000 Ia1-Ia1- IIIa3-IIIa3- L2-3.40-L2- Ia1-Ia1 biv. no Nanobody
MSA21 MSA21 3.40-MSA21
Nanobody concentration (nM)

Fig. 3 Anti-EGFR Nanobodies compete for EGF binding to the Nanobody Ia1 were tested for their ability to block EGF binding
EGFR and trivalent bispeciWc Nanobodies react simultaneously to the EGFR in ELISA. c 3 g of puriWed mono-, bi- and trivalent
with EGFR and MSA. a By means of ELISA, the binding of bio- Ia1 Nanobody was size-separated on a 15% poly-acrylamide gel
tinylated EGF to immobilised EGFR was measured in the pres- and the gel was stained with coomassie brilliant blue to visualise
ence of increasing amounts of a control Nanobody (stripes) or of the proteins. d Trivalent, bispeciWc Nanobodies (of Ia1, IIIa3 and
the anti-EGFR Nanobodies Ia1 (diamonds), IIIa3 (squares) or L2–3.40), or bivalent Ia1 were tested for simultaneous binding to
L2-3.40 (triangles). Background staining was deWned as no biotin- EGFR (coated to the ELISA plate) and biotinylated MSA (used
ylated EGF being added (rounds). b Monovalent (rounds), biva- to detect bound Nanobody with alkaline-phosphatase coupled
lent (diamonds) and trivalent (triangles) variants of anti-EGFR extravidin)

44 h, compared to 1 h: G. van Dongen, personal com- the EGF-induced phosphorylation of Y-1068 of the
munication). EGFR (Fig. 4a). This eVect was already signiWcant by
The ability of these trivalent, bispeciWc anti-EGFR addition of an equimolar quantity of antibody fragment.
Nanobodies to inhibit EGF-mediated signalling was sub- Furthermore, when the antibody fragments themselves
sequently tested: EGF-induced EGFR phosphorylation were used as ligand at high concentration (1 M), only a
was measured in the presence or absence of the diVerent marginal phosphorylation of the EGFR Y-1068 could be
Nanobodies. As a measure of mitogenic signalling, the observed (Fig. 4b), showing that the fragments them-
phosphorylation status of tyrosine (Y-)1068 of the selves did not act as receptor agonists.
EGFR was determined, as this tyrosine is the docking The next test performed in the development of these
site for Grb2 and its phosphorylation is the initiation of antibody fragments for cancer therapy was to measure
signalling towards Ras [5]. Addition of an increasing their eVect on EGF-induced cell proliferation. Increas-
amount of the trivalent, bispeciWc Nanobodies to a con- ing quantities of the three selected anti-EGFR trivalent
stant quantity (8 nM) of EGF could completely block Nanobodies were mixed with a constant quantity of

123
312 Cancer Immunol Immunother (2007) 56:303–317

Table 1 Schematic illustration of the diVerent Nanobody- treated with high doses of antibody (1 mg per mouse per
derived constructs injection, twice a week for 4 weeks). This treatment
schedule, in combination with the long in vivo half-life
of the antibody fragments, was designed to ensure a con-
tinuous high blood level of Nanobody. Treatment with
all three Nanobodies resulted in a signiWcant delay of
tumour outgrowth (Fig. 6d). Since Nanobody-treatment
was abrogated after 4 weeks, most tumours started
growing at control rate from that time onwards. How-
ever, this was surprisingly not the case for tumours
treated with the IIIa3 Nanobody (Fig. 6d). During treat-
ment, there was no statistically signiWcant diVerence
between the diVerent Nanobodies tested. In conclusion,
Bio biotinylation tag; Hinge N-terminal part of the long hinge the selected Nanobodies were eVective both in vitro and
region found in IgG3 HcAb’s; His6 hexahistidine tag; MSA21 in vivo in inhibiting tumour (cell) growth.
cDNA encoding the anti-mouse serum albumin (MSA) Nano-
body clone 21; Myc cMyc-derived epitope tag; pLacZ LacZ pro-
moter; B BstEII site; N Not1 site; P Pst1 site; S SW1 site
Discussion
EGF and the mitogenic eVect of these mixtures was
then tested on serum-starved Her14 cells. Both the Ia1 In this paper, we report on the successful isolation and
and L2–3.40 Nanobodies inhibited proliferation for the characterisation of a panel of antagonistic anti-
more than 80% at high doses (Fig. 5). The IIIa3 Nano- EGFR Nanobodies. These antibody fragments were
body was less potent, giving a reduction in proliferation shown to potently inhibit EGF binding to the receptor,
of approximately 40% at the highest dose tested (1 M; to block EGF-induced EGFR signalling and to inhibit
Fig. 5b). The observed growth eVect was dependent on EGF-dependent cell proliferation. Importantly, to our
EGF, since control cells not receiving any EGF stimula- knowledge, this is the Wrst report describing the suc-
tion did not signiWcantly proliferate in the time span of cessful in vivo use of unconjugated Nanobodies to
the assay (4 days; Fig. 5). Crucial for the application of inhibit solid tumour outgrowth. These antibody frag-
Nanobodies in cancer therapy is their eVect on the ments may prove to be useful building blocks for fur-
growth of tumour cells. This eVect was measured using ther rational design of anti-cancer therapeutics.
the A431 cell line, both in vitro and in vivo in a murine Several reports have already described the isolation
xenograft model. The eVect of the selected Nanobodies of recombinant antibody fragments speciWc for the
on A431 cell proliferation was assessed by measuring EGFR [23, 51]. However, this is the Wrst report
[3H]-thymidine incorporation. In the presence of a low describing EGFR-speciWc Nanobodies. Furthermore,
concentration of EGF (170 pM), all three trivalent anti- by employing a functional selection approach, EGFR-
body fragments inhibited the growth of A431 cells speciWc Nanobodies could readily be identiWed that
(Fig. 6a–c). When added in excess, all three Nanobodies indeed blocked EGFR signalling by competing for
could completely block the EGF-induced proliferation EGF binding. This obviated the need to screen EGFR-
of A431 cells, whereas a large excess of a control Nano- speciWc Nanobodies for their capacity to block EGF
body had no signiWcant eVect (data not shown). Having binding to the receptor. However, true EGFR antago-
the in vitro characteristics of true EGFR antagonistic nism still had to be conWrmed (Fig. 4), since the
molecules with suitable in vivo half-lives, these triva- selected Nanobodies could have been able to stimulate
lent, bispeciWc Nanobodies were then tested for their the tyrosine kinase activity of the receptor.
in vivo eYcacy in tumour treatment. To synthesise ‘immune’ Nanobody repertoires for
the EGFR, Llama glama were Wrst immunised with
In vivo eYcacy of bispeciWc, trivalent anti-EGFR EGFR-containing cell extracts. The successful induc-
Nanobodies tion of a humoral immune response in the animals was
then demonstrated by whole cell-ELISA (Fig. 1).
To test the eVect of the selected anti-EGFR Nanobodies However, this assay detects conventional, as well as
on in vivo tumour development, an eYcacy study was HcAb’s. Earlier reports describe signiWcant contribu-
performed to determine the inhibition of tumour out- tions of HcAb’s to the total antibody response induced
growth in athymic (nude) mice. Mice were subcutane- by immunisation of camelids, especially for protein
ously injected with A431 tumour cells and subsequently antigens [32]. For hapten antigens, however, conXicting

123
Cancer Immunol Immunother (2007) 56:303–317 313

A MW (kDa)
Blot:
170 anti-EGFR
P-Y 1068

Blot:
170 anti-EGFR

Blot:
40 anti-actin

EGF (8nM) - + + + + + + + + + + + + +

Ia1(nM) 1 10 100 1000

IIIa3 (nM) 1 10 100 1000

L2-3.40 1 10 100 1000

B MW (kDa)

170 Blot:
anti-EGFR
130 P-Y 1068

170
Blot:
130 anti-EGFR

40 Blot:
anti-actin
No EGF Ia1 IIIa3 L2-3.40
ligand

Fig. 4 Anti-EGFR Nanobodies block EGF-induced EGFR staining for phosphorylated tyrosine 1068 of EGFR; middle panel
phosphorylation but do not act as receptor agonists. a EGF staining for total quantity of EGFR; lower panel staining for total
(50 ng/ml, corresponding to approximately 8 nM) was mixed with amount of actin. b EGF (8 nM) or trivalent, bispeciWc Nanobod-
increasing amounts of puriWed, trivalent Nanobodies and added ies (1 M) were added to serum-starved Her14 cells for 15 min
to serum-starved Her14 cells for 15 min. EGFR phosphorylation and EGFR phosphorylation was measured in total cell lysates by
was then measured in cell lysates by Western blotting. Upper panel Western blotting. Blots were stained as described in a

data have been reported, ranging from the successful To improve the aYnity of the selected anti-EGFR
induction of HcAb titers [18] to a complete failure to Nanobodies, multi-valent formats were synthesised.
detect hapten-speciWc HcAb’s in the serum of immun- When these molecules were tested for their potency
ised animals [31]. Therefore, the serum titers shown in to block EGF binding to the receptor, bivalent and
Fig. 1 are merely a strong indication of the successful trivalent molecules seemed almost equally eVective,
induction of an HcAb-response. However, when poly- whereas bivalent molecules were signiWcantly better
clonal phage, prepared from one of the immune phage than their monovalent counterparts (Fig. 3). ConXict-
Nanobody libraries, was tested in an ELISA on puri- ing data have been obtained when measuring the
Wed EGFR, speciWc binding was observed (data not eVect of increased apparent aYnity of antibody frag-
shown). This means that there probably was a rela- ments through the addition of a second antigen-
tively high percentage of anti-EGFR Nanobodies pres- binding site to the same molecule [44, 53]. One
ent in the ‘immune’ repertoires, indicative of a strong explanation for this is that this eVect heavily depends
humoral immune response directed towards the on the format of the assay used to determine the
EGFR. Such a strong response is remarkable in view of apparent aYnity. In the assay used to determine the
the low quantity of antigen used per immunisation (108 capacity of multi-valent Nanobodies to block EGF
A431 cells contain roughly 60 g of EGFR). binding to the receptor, the density of coated EGFR

123
314 Cancer Immunol Immunother (2007) 56:303–317

A 3.5 Ia1-Ia1-MSA21 Nanobody molecules may positively aVect their thera-


peutic capacity.
3

Relative cell number


Nanobodies are small proteins and will therefore
2.5
have a very short half-life in vivo that is not compatible
2 with their use in tumour therapy. To increase the in
1.5 vivo half-life of small proteins that are below the renal
1 threshold for Wrst-pass clearance, addition of polyethyl-
ene glycol (PEG; [48]) has often been used. However,
0.5
since Nanobodies are very small, there is a consider-
0
able danger that such chemical modiWcation may
EGF (8nM) - + + + + + diminish the immuno-reactivity of the molecule [13].
Nanobody (nM) - - 1 10 100 1000 Therefore, a mouse serum albumin-speciWc Nanobody
was linked to the bivalent anti-EGFR Nanobodies to
B 4 IIIa3-IIIa3-MSA21
prolong their in vivo half-life. The three ‘heads’ in such
3.5
a trivalent molecule function independently of each
Relative cell number

3 other. This is exempliWed by the fact that the IC50 val-


2.5 ues for EGF competition did not diVer between biva-
2 lent Nanobodies and their trivalent counterparts
1.5 containing the anti-MSA Nanobody (data not shown).
1 Indeed, the half-life of these trivalent Nanobodies was
0.5 signiWcantly longer and much more adequate for treat-
ment purposes. However, this half-life is still less than
0
that of a whole IgG (being 10–14 days).
EGF (8nM) - + + + + + The immunogenicity of Nanobodies is something
Nanobody (nM) - - 1 10 100 1000
that still needs to be thoroughly investigated. This is
L2-3.40-L2-3.40-MSA21 also important for the pharmacokinetic behaviour of
C 3 these molecules, as immunogenicity will cause the for-
2.5 mation of immune complexes and a diminished half-
Relative cell number

life in vivo. However, the large sequence homology


2
between Nanobodies and human VH genes of the VH
1.5 III family [40] indicates that this may not be a major
1
problem. In addition, no B- or T-cell responses have
been detected in mice treated with Nanobodies, giving
0.5 further support to the notion that these molecules may
0 ultimately be safely administered to patients.
EGF (8nM) - + + + + +
An important advantage of this phage display
Nanobody (nM) - - 1 10 100 1000 approach is the large number of diVerent anti-EGFR
Nanobodies selected. Subsequent selection criteria,
Fig. 5 Trivalent, bispeciWc Nanobodies inhibit EGF-dependent based on the in vitro, but also the in vivo characteris-
growth of Her14 cells in vitro. Increasing quantities (0. 1, 10, 100
and 1,000 nM) of puriWed trivalent Nanobodies were mixed with
tics of the molecules are then very important to
EGF (8 nM) and added to serum-starved Her14 cells for 4 days. determine the candidate having the greatest thera-
Total cellular protein was then precipitated with tri-chloro acetic peutic potential. For example, it is not always possi-
acid (TCA) and stained with sulpho-rhodamine B (SRB) as a ble to extrapolate in vitro data, e.g. blocking of
measure of total cell number. Background proliferation was
determined in the absence of EGF. a trivalent Ia1; b trivalent
ligand binding and inhibition of (EGF-dependent)
IIIa3 and c trivalent L2–3.40 cell growth, to in vivo performance (i.e. inhibition of
tumour growth). This is exempliWed by the fact that
the Nanobody displaying the weakest capacity to
molecules used might be so low that trivalent binding block EGF binding to the EGFR (IIIa3; Fig. 3)
was spatially impossible. Therefore, trivalency for the seemed most potent in inhibition of in vivo tumour
EGFR may still prove to be advantageous for in vivo growth (Fig. 6d). Whether the observed reduced
therapy. In addition, the introduction of more Xexibil- tumour growth after treatment with Nanobody IIIa3
ity in the trivalent molecules by changing the length had been stopped (Fig. 6d) is signiWcant remains to
and composition of the linker sequence joining two be determined. It is possible that treatment with

123
Cancer Immunol Immunother (2007) 56:303–317 315

A 60000 Ia1-Ia1-M SA21

50000 Fig. 6 Trivalent, bispeciWc Nanobodies inhibit A431 tumour cell


growth in vitro and growth of A431 xenografts in athymic mice.
40000 a–c A431 epidermoid carcinoma cells were grown in serum-free
medium supplemented with insulin, transferrin and selenium.
cpm

30000
After 24 h, medium was refreshed and dilutions (100, 10, 1 and
20000 0 nM) of trivalent Nanobodies (a Ia1; b IIIa3; c L2–3.40) were
added, together with 1 ng/ml (approximately 170 pM) of human
10000 EGF. After 2 days of culture, cells were pulsed with [3H]- thymi-
dine and assayed 24 h later for the incorporation of radioactivity
0 in genomic DNA. d Groups of 8 mice were subcutaneously inject-
ed with A431 cells (107 cells per mouse). One day later, treatment
EGF - + + + + + was started by intra-peritoneal injection of 1 mg of puriWed tri-
NB (nM) - - 1 10 100 1000 meric Nanobody. Mice were treated twice weekly with the same
quantity of Nanobody for up to 4 weeks (indicated by arrows).
IIIa3-IIIa3-MSA21 Relative tumour volume (RTV) was measured as function of
B 60000 time. Squares solvent control (PBS); triangles IaI; inverse triangles
50000 IIIa3 and diamonds L2–3.40. Error bars indicate the standard
error of the mean of 8 mice
40000
cpm

30000

20000
Nanobody IIIa3 caused an immune response generat-
10000
ing immunological memory (e.g. through the induc-
0 tion of an anti-idiotype (Id) cascade). A recent
report [2] describes anti-tumour eVects of an anti-
EGF - + + + + +
NB (nM) - 1 10 100 1000 EGFR whole human monoclonal antibody that are
-
probably mediated by Fc receptor-bearing immune
eVector cells (ADCC). However, whether these immune
C 60000 L2-3.40-L2-3.40-MSA21
eVector functions will have important beneWcial
50000 eVects in patients remains to be determined. Indeed,
an antibody devoid of eVector functions (ABX-EGF,
40000
IgG2) has shown promising pre-clinical results [17]
cpm

30000 giving further support to the notion that blocking


20000 EGFR signalling on itself can have potent anti-
tumour eVects.
10000 Recombinant anti-EGFR antibody fragments have
0 also been used to generate fusion proteins designed
to re-direct immune eVector cells to tumours [22] or
EGF - + + + + +
to deliver cytotoxic agents [3, 4, 33] or genes [20] to
NB (nM) - - 1 10 100 1000
tumour cells. The observed anti-tumour eVects of the
D 40 Nanobodies described in this report can be solely
attributed to their antagonistic mode of action. In
PBS
addition, they are internalised in target cells over-
30 Ia1-Ia1-MSA21
IIIa3-IIIA3-MSA21
expressing the receptor (data not shown). Therefore,
L2-3.40-L2-3.40-MSA21 the re-formatting of these fragments, e.g. as immuno-
RTV

20 toxin, may signiWcantly enhance their anti-tumour


eVect. Because the antibody fragments described here
are already eVective in delaying tumour outgrowth,
10 we are conWdent that they will prove to be useful
building blocks for the rational design of anti-cancer
0 therapeutics. An especially promising treatment
0 10 20 30 40 modality may be the combination of chemo- or radio-
therapeutic regimens in combination with antibody
Time [Days] treatment [25, 41].

123
316 Cancer Immunol Immunother (2007) 56:303–317

In conclusion, the data presented here demonstrate 12. De Haard HJ, Bezemer S, Ledeboer AM, Muller WH,
that the selection of Nanobodies for a particular func- Boender PJ, Moineau S, Coppelmans MC, Verkleij AJ, Fren-
ken LG, Verrips CT (2005) Llama antibodies against a lacto-
tion using antibody phage display is a very powerful coccal protein located at the tip of the phage tail prevent
tool to quickly generate biologically active antibody- phage infection. J Bacteriol 187:4531
based cancer therapeutics. In addition, the favourable 13. Delgado C, Pedley RB, Herraez A, Boden R, Boden JA,
characteristics of Nanobodies for cancer therapy [42] Keep PA, Chester KA, Fisher D, Begent RH, Francis GE
(1996) Enhanced tumour speciWcity of an anti-carcinoembr-
make these molecules ideal candidates for the develop- ionic antigen Fab’ fragment by poly(ethylene glycol) (PEG)
ment of antibody-based immuno-therapeutics. modiWcation. Br J Cancer 73:175
14. Dolk E, van Vliet C, Perez JM, Vriend G, Darbon H, Ferrat
Acknowledgments We thank prof. Dr. Guus van Dongen G, Cambillau C, Frenken LG, Verrips T (2005) Induced
(VUMC, Amsterdam, The Netherlands) for pharmacokinetic refolding of a temperature denatured llama heavy-chain anti-
data of trivalent, bispeciWc Nanobody fragments and for critically body fragment by its antigen. Proteins 59:555
reading the manuscript. The term Nanobody™ was used with the 15. Els Conrath K, Lauwereys M, Wyns L, Muyldermans S
kind permission of Ablynx N.V. (2001) Camel single-domain antibodies as modular building
units in bispeciWc and bivalent antibody constructs. J Biol
Chem 276:7346
16. Ewert S, Cambillau C, Conrath K, Pluckthun A (2002) Bio-
References physical properties of camelid V(HH) domains compared to
those of human V(H)3 domains. Biochemistry 41:3628
1. Arbabi Ghahroudi M, Desmyter A, Wyns L, Hamers R, 17. Foon KA, Yang XD, Weiner LM, Belldegrun AS, Figlin RA,
Muyldermans S (1997) Selection and identiWcation of single Crawford J, Rowinsky EK, Dutcher JP, Vogelzang NJ, Gol-
domain antibody fragments from camel heavy-chain antibod- lub J, Thompson JA, Schwartz G, Bukowski RM, Roskos LK,
ies. FEBS Lett 414:521 Schwab GM (2004) Preclinical and clinical evaluations of
2. Bleeker WK, Lammerts van Bueren JJ, van Ojik HH, Gerrit- ABX-EGF, a fully human anti-epidermal growth factor
sen AF, Pluyter M, Houtkamp M, Halk E, Goldstein J, Schu- receptor antibody. Int J Radiat Oncol Biol Phys 58:984
urman J, van Dijk MA, van de Winkel JG, Parren PW (2004) 18. Frenken LG, van der Linden RH, Hermans PW, Bos JW,
Dual mode of action of a human anti-epidermal growth factor Ruuls RC, de Geus B, Verrips CT (2000) Isolation of antigen
receptor monoclonal antibody for cancer therapy. J Immunol speciWc llama VHH antibody fragments and their high level
173:4699 secretion by Saccharomyces cerevisiae. J Biotechnol 78:11
3. Bremer E, Samplonius DF, van Genne L, Dijkstra MH, 19. Giard DJ, Aaronson SA, Todaro GJ, Arnstein P, Kersey JH,
Kroesen BJ, de Leij LF, Helfrich W (2005) Simultaneous Dosik H, Parks WP (1973) In vitro cultivation of human
inhibition of EGFR signaling and enhanced activation of tumors: establishment of cell lines derived from a series of
TRAIL-R-mediated apoptosis induction by an scFv:sTRAIL solid tumors. J Natl Cancer Inst 51:1417
fusion protein with speciWcity for human EGFR. J Biol Chem 20. Haisma HJ, Grill J, Curiel DT, Hoogeland S, van Beusechem
280(11):10025–10033 VW, Pinedo HM, Gerritsen WR (2000) Targeting of adeno-
4. Bruell D, Stocker M, Huhn M, Redding N, Kupper M, Schum- viral vectors through a bispeciWc single-chain antibody. Can-
acher P, Paetz A, Bruns CJ, Haisma HJ, Fischer R, Finnern R, cer Gene Ther 7:901
Barth S (2003) The recombinant anti-EGF receptor immuno- 21. Hamers-Casterman C, Atarhouch T, Muyldermans S, Robin-
toxin 425(scFv)-ETA’ suppresses growth of a highly meta- son G, Hamers C, Songa EB, Bendahman N, Hamers R
static pancreatic carcinoma cell line. Int J Oncol 23:1179 (1993) Naturally occurring antibodies devoid of light chains.
5. Buday L, Downward J (1993) Epidermal growth factor regu- Nature 363:446
lates p21ras through the formation of a complex of receptor, 22. Hayashi H, Asano R, Tsumoto K, Katayose Y, Suzuki M,
Grb2 adapter protein, and Sos nucleotide exchange factor. Unno M, Kodama H, Takemura S, Yoshida H, Makabe K,
Cell 73:611 Imai K, Matsuno S, Kumagai I, Kudo T (2004) A highly eVec-
6. Chomczynski P, Sacchi N (1987) Single-step method of RNA tive and stable bispeciWc diabody for cancer immunotherapy:
isolation by acid guanidinium thiocyanate-phenol-chloro- cure of xenografted tumors by bispeciWc diabody and T-LAK
form extraction. Anal Biochem 162:156 cells. Cancer Immunol Immunother 53:497
7. Clackson T, Hoogenboom HR, GriYths AD, Winter G 23. Heitner T, Moor A, Garrison JL, Marks C, Hasan T, Marks
(1991) Making antibody fragments using phage display librar- JD (2001) Selection of cell binding and internalizing epider-
ies. Nature 352:624 mal growth factor receptor antibodies from a phage display
8. Cohen S, Ushiro H, Stoscheck C, Chinkers M (1982) A native library. J Immunol Methods 248:17
170,000 epidermal growth factor receptor-kinase complex 24. Herbst RS, Arquette M, Shin DM, Dicke K, Vokes EE, Azar-
from shed plasma membrane vesicles. J Biol Chem 257:1523 nia N, Hong WK, Kies MS (2005) Phase II multicenter study
9. Cortez-Retamozo V, Backmann N, Senter PD, Wernery U, of the epidermal growth factor receptor antibody Cetuximab
De Baetselier P, Muyldermans S, Revets H (2004) EYcient and Cisplatin for recurrent and refractory squamous cell car-
cancer therapy with a nanobody-based conjugate. Cancer Res cinoma of the head and neck. J Clin Oncol 23:5578
64:2853 25. Herbst RS, Langer CJ (2002) Epidermal growth factor recep-
10. Cortez-Retamozo V, Lauwereys M, Hassanzadeh Gh G, Go- tors as a target for cancer treatment: the emerging role of
bert M, Conrath K, Muyldermans S, De Baetselier P, Revets IMC-C225 in the treatment of lung and head and neck can-
H (2002) EYcient tumor targeting by single-domain antibody cers. Semin Oncol 29:27
fragments of camels. Int J Cancer 98:456 26. Holbro T, Civenni G, Hynes NE (2003) The ErbB receptors
11. Daub H, Weiss FU, Wallasch C, Ullrich A (1996) Role of and their role in cancer progression. Exp Cell Res 284:99
transactivation of the EGF receptor in signalling by G-pro- 27. Honegger AM, Dull TJ, Felder S, Van Obberghen E, Bellot
tein-coupled receptors. Nature 379:557 F, Szapary D, Schmidt A, Ullrich A, Schlessinger J (1987)

123
Cancer Immunol Immunother (2007) 56:303–317 317

Point mutation at the ATP binding site of EGF receptor abol- 43. Roovers RC, Henderikx P, Helfrich W, van der Linden E,
ishes protein-tyrosine kinase activity and alters cellular rout- Reurs A, de Bruine AP, Arends JW, de Leij L, Hoogenboom
ing. Cell 51:199 HR (1998) High-aYnity recombinant phage antibodies to the
28. Hoogenboom HR (2005) Selecting and screening recombi- pan-carcinoma marker epithelial glycoprotein-2 for tumour
nant antibody libraries. Nat Biotechnol 23:1105 targeting. Br J Cancer 78:1407
29. Hoogenboom HR, de Bruine AP, Hufton SE, Hoet RM, 44. Roovers RC, van der Linden E, de Bruine AP, Arends JW,
Arends JW, Roovers RC (1998) Antibody phage display Hoogenboom HR (2001) In vitro characterisation of a mono-
technology and its applications. Immunotechnology 4:1 valent and bivalent form of a fully human anti Ep-CAM
30. Laemmli UK (1970) Cleavage of structural proteins during phage antibody. Cancer Immunol Immunother 50:51
the assembly of the head of bacteriophage T4. Nature 227:680 45. Rowinsky EK (2004) The erbB family: targets for therapeutic
31. Lange IG, Daxenberger A, Meyer HH (2001) Studies on the development against cancer and therapeutic strategies using
antibody response of Lama glama—evaluation of the binding monoclonal antibodies and tyrosine kinase inhibitors. Annu
capacity of diVerent IgG subtypes in ELISAs for clenbuterol Rev Med 55:433
and BSA. Vet Immunol Immunopathol 83:1 46. Rowinsky EK, Schwartz GH, Gollob JA, Thompson JA,
32. Lauwereys M, Arbabi Ghahroudi M, Desmyter A, Kinne J, Vogelzang NJ, Figlin R, Bukowski R, Haas N, Lockbaum P,
Holzer W, De Genst E, Wyns L, Muyldermans S (1998) Li YP, Arends R, Foon KA, Schwab G, Dutcher J (2004) Safety,
Potent enzyme inhibitors derived from dromedary heavy- pharmacokinetics, and activity of ABX-EGF, a fully human
chain antibodies. Embo J 17:3512 anti-epidermal growth factor receptor monoclonal antibody in
33. Mamot C, Drummond DC, Greiser U, Hong K, Kirpotin DB, patients with metastatic renal cell cancer. J Clin Oncol 22:3003
Marks JD, Park JW (2003) Epidermal growth factor receptor 47. Saltz LB, Meropol NJ, Loehrer PJ Sr, Needle MN, Kopit J,
(EGFR)-targeted immunoliposomes mediate speciWc and Mayer RJ (2004) Phase II trial of cetuximab in patients with
eYcient drug delivery to EGFR- and EGFRvIII-overexpress- refractory colorectal cancer that expresses the epidermal
ing tumor cells. Cancer Res 63:3154 growth factor receptor. J Clin Oncol 22:1201
34. Marks JD, Hoogenboom HR, Bonnert TP, McCaVerty J, 48. Savoca KV, Abuchowski A, van Es T, Davis FF, Palczuk NC
GriYths AD, Winter G (1991) By-passing immunization. (1979) Preparation of a non-immunogenic arginase by the
Human antibodies from V-gene libraries displayed on phage. covalent attachment of polyethylene glycol. Biochim Biophys
J Mol Biol 222:581 Acta 578:47
35. McCaVerty J, GriYths AD, Winter G, Chiswell DJ (1990) 49. Skehan P, Storeng R, Scudiero D, Monks A, McMahon J,
Phage antibodies: Wlamentous phage displaying antibody var- Vistica D, Warren JT, Bokesch H, Kenney S, Boyd MR
iable domains. Nature 348:552 (1990) New colorimetric cytotoxicity assay for anticancer-
36. Merlino GT, Xu YH, Ishii S, Clark AJ, Semba K, Toyoshima drug screening. J Natl Cancer Inst 82:1107
K, Yamamoto T, Pastan I (1984) AmpliWcation and enhanced 50. Smith GP (1985) Filamentous fusion phage: novel expression
expression of the epidermal growth factor receptor gene in vectors that display cloned antigens on the virion surface. Sci-
A431 human carcinoma cells. Science 224:417 ence 228:1315
37. Meulemans EV, Slobbe R, Wasterval P, Ramaekers FC, van 51. Souriau C, Rothacker J, Hoogenboom HR, Nice E (2004)
Eys GJ (1994) Selection of phage-displayed antibodies spe- Human antibody fragments speciWc for the epidermal growth
ciWc for a cytoskeletal antigen by competitive elution with a factor receptor selected from large non-immunised phage dis-
monoclonal antibody. J Mol Biol 244:353 play libraries. Growth Factors 22:185
38. Mosesson Y, Yarden Y (2004) Oncogenic growth factor 52. Spano JP, Lagorce C, Atlan D, Milano G, Domont J, Bena-
receptors: implications for signal transduction therapy. Semin mouzig R, Attar A, Benichou J, Martin A, Morere JF, Rapha-
Cancer Biol 14:262 el M, Penault-Llorca F, Breau JL, Fagard R, Khayat D, Wind
39. Muyldermans S (2001) Single domain camel antibodies: cur- P (2005) Impact of EGFR expression on colorectal cancer
rent status. J Biotechnol 74:277 patient prognosis and survival. Ann Oncol 16:102
40. Nguyen VK, Hamers R, Wyns L, Muyldermans S (2000) 53. Todorovska A, Roovers RC, Dolezal O, Kortt AA, Hoogen-
Camel heavy-chain antibodies: diverse germline V(H)H and boom HR, Hudson PJ (2001) Design and application of
speciWc mechanisms enlarge the antigen-binding repertoire. diabodies, triabodies and tetrabodies for cancer targeting.
Embo J 19:921 J Immunol Methods 248:47
41. Raben D, Helfrich B, Chan DC, Ciardiello F, Zhao L, Franklin 54. Verheesen P, ten Haaft MR, Lindner N, Verrips CT, de
W, Baron AE, Zeng C, Johnson TK, Bunn PA Jr (2005) The Haard JJ (2003) BeneWcial properties of single-domain anti-
eVects of cetuximab alone and in combination with radiation body fragments for application in immunoaYnity puriWcation
and/or chemotherapy in lung cancer. Clin Cancer Res 11:795 and immuno-perfusion chromatography. Biochim Biophys
42. Revets H, De Baetselier P, Muyldermans S (2005) Nanobod- Acta 1624:21
ies as novel agents for cancer therapy. Expert Opin Biol Ther 55. Yarden Y, Sliwkowski MX (2001) Untangling the ErbB
5:111 signalling network. Nat Rev Mol Cell Biol 2:127

123

Vous aimerez peut-être aussi