ArticlePDF Available

In vitro effects and ex vivo binding of an EGFR-specific immunotoxin on rhabdomyosarcoma cells

Authors:

Abstract and Figures

Rhabdomyosarcoma (RMS) is a rare and aggressive soft tissue sarcoma with limited treatment options and a high failure rate during standard therapy. New therapeutic strategies based on targeted immunotherapy are therefore much in demand. The epidermal growth factor receptor (EGFR) has all the characteristics of an ideal target. It is overexpressed in up to 80 % of embryonal RMS and up to 50 % of alveolar RMS tumors. We therefore tested the activity of the EGFR-specific recombinant immunotoxin (IT) 425(scFv)-ETA' against EGFR(+) RMS cells in vitro and ex vivo. We tested the specific binding and internalization behavior of 425(scFv)-ETA' in RMS cell lines in vitro by flow cytometry, compared to the corresponding imaging probe 425(scFv)-SNAP monitored by live cell imaging. The cytotoxic activity of 425(scFv)-ETA' was tested using cell viability and apoptosis assays. Specific binding of the IT was confirmed on formalin-fixed paraffin-embedded tissue samples from two RMS patients. We confirmed the specific binding of 425(scFv)-ETA' to RMS cells in vitro and ex vivo. Both the IT and the corresponding imaging probe were rapidly internalized. The IT killed EGFR(+) RMS cells in a dose-dependent manner, while showing no effect against control cells. It showed specific apoptotic activity against one selected RMS cell line. This is the first study showing the promising therapeutic potential of a recombinant, EGFR-targeting, ETA'-based IT on RMS cells. We confirmed the selective killing with IC50 values of up to 50 pM, and immunohistochemical staining confirmed the specific ex vivo binding to primary RMS material.
No caption available
… 
Content may be subject to copyright.
1 3
J Cancer Res Clin Oncol
DOI 10.1007/s00432-014-1884-z
ORIGINAL ARTICLE – CANCER RESEARCH
In vitro effects and ex vivo binding of an EGFR‑specific
immunotoxin on rhabdomyosarcoma cells
Judith Niesen · Hannes Brehm · Christoph Stein · Nina Berges · Alessa Pardo ·
Rainer Fischer · Andre ten Haaf · Stefan Gattenlöhner · Mehmet K. Tur ·
Stefan Barth
Received: 30 October 2014 / Accepted: 19 November 2014
© Springer-Verlag Berlin Heidelberg 2014
IT was confirmed on formalin-fixed paraffin-embedded tis-
sue samples from two RMS patients.
Results We confirmed the specific binding of 425(scFv)-
ETA to RMS cells in vitro and ex vivo. Both the IT and
the corresponding imaging probe were rapidly internal-
ized. The IT killed EGFR+ RMS cells in a dose-depend-
ent manner, while showing no effect against control cells.
It showed specific apoptotic activity against one selected
RMS cell line.
Conclusions This is the first study showing the promising
therapeutic potential of a recombinant, EGFR-targeting,
ETA-based IT on RMS cells. We confirmed the selective
killing with IC50 values of up to 50 pM, and immunohisto-
chemical staining confirmed the specific ex vivo binding to
primary RMS material.
Keywords Immunotoxin · EGFR · scFv ·
Rhabdomyosarcoma · Pseudomonas exotoxin A
Introduction
Rhabdomyosarcoma (RMS) is the most common pediatric
soft tissue sarcoma with about 4.5 cases per million chil-
dren per year (Ray and Huh 2012). There are three main
subtypes: embryonal (ERMS), the more aggressive alveo-
lar (ARMS) and the rare pleomorphic (PRMS). ERMS is
the most common subtype and accounts for approximately
70 % of all cases and occurs most often in children under
10 years. It has a better prognosis than ARMS and shows a
heterogeneous mutation pattern, with diverse gain-of-func-
tion and loss-of-function mutations, but the most prominent
mutation is loss of heterozygosity at 11p15.5 which leads to
the overexpression of several other genes (Davicioni et al.
2009). ARMS, which occurs more often in young adults,
Abstract
Purpose Rhabdomyosarcoma (RMS) is a rare and aggres-
sive soft tissue sarcoma with limited treatment options and
a high failure rate during standard therapy. New therapeu-
tic strategies based on targeted immunotherapy are there-
fore much in demand. The epidermal growth factor recep-
tor (EGFR) has all the characteristics of an ideal target. It
is overexpressed in up to 80 % of embryonal RMS and up
to 50 % of alveolar RMS tumors. We therefore tested the
activity of the EGFR-specific recombinant immunotoxin
(IT) 425(scFv)-ETA against EGFR+ RMS cells in vitro
and ex vivo.
Methods We tested the specific binding and internaliza-
tion behavior of 425(scFv)-ETA in RMS cell lines in vitro
by flow cytometry, compared to the corresponding imag-
ing probe 425(scFv)-SNAP monitored by live cell imaging.
The cytotoxic activity of 425(scFv)-ETA was tested using
cell viability and apoptosis assays. Specific binding of the
Judith Niesen and Hannes Brehm shared first authorship.
J. Niesen · C. Stein · R. Fischer · S. Barth (*)
Fraunhofer Institute for Molecular Biology and Applied Ecology
IME, 52074 Aachen, Germany
e-mail: stefan.barth@ime.fraunhofer.de
H. Brehm · C. Stein · N. Berges · A. Pardo · S. Barth
Department of Experimental Medicine and Immunotherapy,
Institute of Applied Medical Engineering, University Hospital
RWTH Aachen, Aachen, Germany
R. Fischer
Institute of Molecular Biotechnology (Biology VII), RWTH
Aachen University, Aachen, Germany
A. ten Haaf · S. Gattenlöhner · M. K. Tur
Department of Pathology, Justus-Liebig University, Giessen,
Germany
J Cancer Res Clin Oncol
1 3
involves a recurrent chromosomal translocation t(2;13)
(q35;q14) generating the strongly expressed oncogene
PAX3-FKHR. Less frequent translocations include PAX7-
FHKR, PAX3-NCOA1 and PAX3-NCOA2 (Sumegi et al.
2010; Zanola et al. 2012). PRMS is rare, adult-specific and
has a complex mutation pattern (Jain et al. 2010). RMS is
still diagnosed and classified using histological methods,
but several studies have shown that gene expression pro-
files should also be considered for risk stratification and
treatment (Missiaglia et al. 2012; Williamson et al. 2010).
Despite recent improvements that have increased the 5-year
overall survival rate from 20 to 70 %, the clinical outcome
for adolescent ARMS patients and patients with relapsed
and metastatic RMS is still poor, with 20–30 % survival
(Ray and Huh 2012; Simon-Keller et al. 2013). Typical
treatment involves multi-agent chemotherapy, which may
be combined with radiotherapy and resection of the tumor
depending on the condition of the patient and site of the
tumor (Hawkins et al. 2014). Therefore, more effective and
targeted treatment options would be beneficial especially
for high-risk patients.
An ideal immunotherapeutic target, such as the epider-
mal growth factor receptor (EGFR), combines overexpres-
sion at the tumor site with little or no expression in healthy
tissue (Azemar et al. 2000; Bruell et al. 2003). The EGFR
or ErbB1 is a member of the ErbB family, which includes
HER-2/ErbB2, HER-3/ErbB3 and HER-4/ErbB4 (Koefoed
et al. 2011). EGFR is one of the best-defined target anti-
gens for cancer treatment, and therefore, a variety of thera-
peutic approaches and drug-development platforms are
based on this receptor (Tebbutt et al. 2013). EGFR signal-
ing is activated by the binding of ligands such as epidermal
growth factor (EGF) and transforming growth factor alpha
(TGFα). These regulate cell growth, proliferation, differ-
entiation and survival. The EGFR is a receptor tyrosine
kinase comprising an extracellular domain, a hydrophobic
transmembrane domain and an intracellular domain. The
latter is phosphorylated after ligand binding and receptor
dimerization, activating the downstream signaling cascade
(Yewale et al. 2013). EGFR signaling is often hyperac-
tive in tumor cells due to mutations that increase ligand
or receptor expression, confer constitutive receptor activ-
ity or allow cross-talk with other receptors. This causes
uncontrolled proliferation and is associated with resistance
to chemotherapy, poor prognosis and a greater likelihood
of metastasis (Bruell et al. 2003; Koefoed et al. 2011; Ped-
ersen et al. 2010). Like many other tumors, RMS is char-
acterized by the overexpression of EGFR, HER-2 and/or
HER-3 (De Giovanni et al. 1996; Ricci et al. 2002), and
studies have shown that EGFR is expressed and active in
31–76 % of ERMS and 16–50 % of ARMS tumors (Armi-
stead et al. 2007; Cen et al. 2007; Ganti et al. 2006; Wachtel
et al. 2006). Because EGFR is a tumor-associated surface
antigen that is rarely expressed in normal tissues, it repre-
sents an ideal target for RMS immunotherapy (Mendelsohn
2002; Yewale et al. 2013).
Targeted cancer therapeutics, such as antibody-based
immunotoxins (ITs), are representing an important field of
increasing interest, comprise a targeting component such as
a full-size monoclonal antibody (mAb) or fragment thereof,
and a cytotoxic agent based on small-molecule drugs, radi-
oisotopes or toxins derived from fungi, plants or bacteria
(Wayne et al. 2014; Weidle et al. 2014). Although mAbs
are specific, their size (~150 kDa) can limit tumor penetra-
tion; therefore, single-chain fragment variable (scFv) deriv-
atives (~30 kDa) comprising only the VH VL sequences
of the parent mAb are more versatile (Ahmad et al. 2012;
Bruell et al. 2005). Recombinant ITs are fusion proteins
that usually combine scFvs with truncated toxins (Bruell
et al. 2003; Singh et al. 2007).
Here, we used a truncated version of Pseudomonas aer-
uginosa exotoxin A (ETA) which has been used success-
fully as part of ITs against hematologic and solid tumors
(Becker and Benhar 2012). The natural cell-binding domain
of ETA is replaced with a ligand or scFv that binds to a
tumor surface antigen, so that the toxin is internalized and
can kill the cell (Antignani and Fitzgerald 2013). We used
the EGFR-specific IT 425(scFv)-ETA, which has shown
cytotoxicity against human pancreatic cancer cells in vitro
and in vivo in a xenograft tumor model (Bruell et al. 2003,
2005; Pardo et al. 2012). To our knowledge, only Ricci
et al. (2002) have reported the targeting of EGFR+ RMS
cells using an IT, although this was an indirect approach
in which a primary specific murine mAb binding to EGFR
and a secondary F(ab)2 anti-mouse Ig was linked to
saporin-S6 via a disulfide bond. Treatment with this indi-
rect IT induced apoptosis and inhibited protein synthesis in
the RMS cell line RD, with an IC50 of 950 pM (Ricci et al.
2002).
We compared the 425(scFv)-ETA IT with the corre-
sponding imaging probe 425(scFv)-SNAP which was cre-
ated using SNAP-tag technology. The SNAP-tag derived
from the human DNA-repair enzyme alkylguanine-DNA
alkyltransferase (hAGT) binds specifically, rapidly and
covalently to substrates that contain O(6)-benzylguanine
(BG). The self-labeling reaction involves the irreversible
transfer of an alkyl group to a cysteine residue. The advan-
tage of the SNAP-tag technology is the quantitative and
specific labeling of the antibody fragments without affect-
ing their antigen-binding properties. Any scFv can thus be
combined with a variety of BG-labeled fluorophores using
this simple and rapid covalent reaction (Amoury et al.
2013; Hussain et al. 2013; Kampmeier et al. 2009).
Here, we show for the first time the successful and prom-
ising in vitro and ex vivo effect of a recombinant EGFR-
specific IT and the corresponding preclinical imaging
J Cancer Res Clin Oncol
1 3
probe against RMS cell lines and RMS tumor samples from
patients.
Materials and methods
Bacterial strains and plasmids
The expression plasmids were prepared using E. coli Dh5α
(F endA1 glnV44 thi-1 recA1 relA1 gyrA96 deoR nupG
Φ80dlacZΔM15 Δ(lacZYA-argF)U169, hsdR17(rK
mK
+),
λ–). The molecular cloning steps were carried out as pre-
viously described (Green and Sambrook 2012). The IT
425(scFv)-ETA was generated previously (Bruell et al.
2003, 2005) and was assembled in the vector pMT, derived
from pET27b (Novagen, Wisconsin, USA) for bacte-
rial expression (Bruell et al. 2003; Tur et al. 2003). The
425(scFv)-SNAP construct was cloned in pMS, a modi-
fied version of pSecTag2/HygroB vector for mammalian
expression (Stocker et al. 2003).
Cell lines and culture conditions
Cell lines were obtained from the American Type Culture
Collection (RMS: TE-671, RD; human embryonic kidney
cells: HEK293T), German Collection of Microorganisms
and Cell Cultures (human histiocytic lymphoma: U937) or
were kindly provided by Ewa Koscielniak (Olgahospital,
Stuttgart, Germany; RMS: FL-OH1). The cell lines were
cultured in RPMI 1640 (Gibco Invitrogen, Carlsbad, USA)
supplemented with 10 % heat-inactivated fetal bovine
serum (Biochrom AG, Berlin, Germany), 2 mM l-glu-
tamine, 100 U/ml penicillin and 0.1 mg/ml streptomycin
(Invitrogen, San Diego, USA).
HEK293T cells were transfected with Roti®-Fect
according to the manufacturer’s protocol (Carl Roth
GmbH, Karlsruhe, Germany). Transfected HEK293T cells
were cultivated in RPMI 1640 medium as described above,
supplemented with 100 µg/ml Zeocin™ (Invitrogen, San
Diego, USA). All cell lines were cultivated at 37 °C in a
humidified environment at 5 % CO2 in a 95 % air incubator
using the recommended media.
Production and purification of the IT and the SNAP-tag
imaging probe
The IT was cloned into a pMT vector as described above.
It was expressed under stress conditions in E. coli BL21
(DE3) cells (F ompT gal dcm lon hsdSB(rB
mB
) λ(DE3
[lacI lacUV5-T7 gene 1 ind1 sam7 nin5])) purchased from
Novagen (Wisconsin, USA) as described elsewhere (Barth
2002; Barth et al. 2000).
The scFv-SNAP construct was cloned in the pMS vector
and expressed as described earlier (Kampmeier et al. 2009;
Stocker et al. 2003). Both proteins were purified using
IMAC as previously described (Hristodorov et al. 2013).
The protein samples were separated by SDS-PAGE (12 %
acrylamide) and detected by staining with Coomassie bril-
liant blue. The separated IT was transferred to a nitrocellu-
lose membrane and specifically detected using the in-house
produced murine ETA-specific mAb TC-1 and an alkaline
phosphatase (AP)-labeled mouse-specific mAb (Green and
Sambrook 2012; Laemmli 1970). The SNAP-tagged pro-
tein was labeled with a fluorophore prior to SDS-PAGE
(Kampmeier et al. 2009, 2010), and the protein was visual-
ized using the CRi Maestro imaging system (Perkin Elmer,
Waltham, MA, USA) and the appropriate filter sets. Protein
concentrations were determined by densitometry after gel
staining compared to bovine serum albumin (BSA) stand-
ards using AIDA Image Analyzer Software (v.4.27.039).
Flow cytometry and internalization assays
Specific cell binding was evaluated by flow cytometry. We
incubated 4 × 105 cells with 1 µg of 425(scFv)-ETA in
100 µl PBS for 30 min on ice. The cells were then washed
and incubated for further 30 min with TC-1, diluted 1:100
in 100 µl PBS. As a positive control, we used a commer-
cially available human EGFR-specific rabbit mAb (Sino
Biological Inc., Beijing, China) diluted 1:10 in 100 µl PBS.
The IT was detected by incubating cells for 30 min on ice
with a FITC-labeled goat anti-mouse mAb (Dianova, Ham-
burg, Germany) diluted 1:100 in 50 µl PBS. For detection
of the control, antibody cells were incubated with a FITC-
labeled goat-anti-rabbit polyclonal antibody diluted 1:50
in 50 µl PBS (Sigma-Aldrich, Taufkirchen, Germany).
The cells were analyzed using a BD FACSVerse (Becton–
Dickinson, Heidelberg, Germany) and the corresponding
software.
The rate of IT internalization was measured as described
by Cizeau et al. (2009). We incubated 2 × 105 cells/ml
with 500 nM 425(scFv)-ETA for 30 min at 4 °C. After
washing, the cells were incubated for different periods of
time (15, 30, 60, 120 and 240 min) in cell culture medium
under normal culture conditions. We used the TC-1 mAb,
as described above, and a PE-labeled goat anti-mouse mAb
diluted 1:100 in 50 µl PBS for detection. The results are
presented as a percentage of the mean fluorescence inten-
sity. The mean fluorescence intensity of the non-internaliz-
ing 4 °C control was set to 100 %. The number of expressed
cell surface antigens was determined using the Qifikit® kit
(Dako, Hamburg, Germany). Flow cytometry was carried
out according to the manufacturer’s protocol using the anti-
human EGFR mAb.
J Cancer Res Clin Oncol
1 3
Confocal microscopy
Internalization of the 425(scFv)-SNAP probe was visual-
ized by confocal microscopy. The 425(scFv)-SNAP con-
struct was coupled to BG-SNAP-Surface® Alexa Fluor®-
488 (New England BioLabs, Schwalbach, Germany) as
described elsewhere (Kampmeier et al. 2009, 2010). We
added 100 nM of the labeled protein to 2 × 105 cells/ml
seeded 24 h earlier in 8-well chamber slides (Thermo Sci-
entific, Waltham, MA, USA). The cells were incubated at
37 °C for 120 min. After a washing step in PBS (only for
the RD cell line), the nucleus was counterstained with bis-
benzimide Hoechst 33342 (Sigma-Aldrich, Taufkirchen,
Germany) diluted 1:1,000 in PBS. Internalization was ana-
lyzed using a LEICA TCS SP8 confocal microscope and
the corresponding software (Leica Microsystems GmbH,
Wetzlar, Germany).
Colorimetric XTT cell proliferation assay
The cytotoxic activity of 425(scFv)-ETA against three
different RMS cell lines (RD, FL-OH1 and TE-671) was
determined using a cell proliferation assay based on XTT
as previously described (Schiffer et al. 2013). Briefly,
5 × 103 cells were plated in 96-well plates and incubated
with different concentrations of the IT for 72 h under nor-
mal culture conditions as described above. Untreated cells,
the non-toxic 425(scFv)-SNAP and a non-binding Mock-
ETAʹ construct were used as controls. After 72 h, we added
50 µl XTT/phenazine methosulfate (Serva and Sigma,
Steinheim, Germany) to the cells, and the samples were
incubated for additional 2–4 h under the same conditions.
The absorbance was measured at 450 and 630 nm as a ref-
erence wavelength using an Epoch Microplate Spectropho-
tometer (Biotek, Bad Friedrichshall, Germany). All experi-
ments were carried out in duplicates or triplicates.
The required IT concentration to achieve a 50 % reduc-
tion in protein synthesis (IC50) relative to the untreated con-
trol cells was calculated using GraphPad Prism 5 software
(GraphPad Software, La Jolla, CA, USA).
AnnexinV/PI Apoptosis assay
The induction of apoptosis was measured by AnnexinV/
PI staining (Stahnke et al. 2008). We incubated 1 × 105
RD cells/well with 30 nM 425(scFv)-ETA in a 24-well
plate for 48 h under the conditions described above. An
equimolar amount of the non-binding ETA-based IT
(Mock-ETA) was used as a negative control. Cells were
also incubated with PBS and camptothecin as negative
and positive controls, respectively. The EGFR cell line
U937 was used as control cell line. After incubation, the
cells were harvested (RD cells were detached by washing
in PBS followed by trypsin/EDTA treatment), washed
with 1 × AnnexinV binding buffer (15 mM NaCl, 1 mM
HEPES, 0.5 mM KCl, 0.2 mM CaCl2, pH 7.4) and stained
with 450 µl cell culture supernatant from AnnexinV-
EGFP-expressing HEK293T cells and 50 µl 10 × Annex-
inV binding buffer for 10 min at room temperature. The
cells were re-suspended in 1 × AnnexinV binding buffer
containing 1 µg/ml propidium iodide (PI) and analyzed by
flow cytometry using BD FACSVerse (Becton–Dickinson,
Heidelberg, Germany).
Ex vivo binding to human tumor tissue
Formalin-fixed and paraffin-embedded (FFPE) tis-
sue sections from RMS tumor samples representing one
ARMS patients and one ERMS patient were equilibrated
in xylene (2 × 5 min), 100 % isopropanol (2 × 5 min),
96 % isopropanol (1 × 5 min) and 70 % isopropanol
(1 × 5 min) and finally washed in PBS for 5 min. After-
ward, the slides were heated in a 600 W microwave for
5 min in citrate buffer (10 mM sodium citrate, 0.05 %
Tween 20, pH 6.0) and allowed to cool for 30 min at
room temperature before washing in PBS. The slides
were incubated for 1 h at room temperature in blocking
solution (PBS + 1 % (v/v) goat serum), washed once in
PBS and incubated for 24 h at 4 °C with either 1 µg of
recombinant 425(scFv)-ETA or an EGFR-specific mAb
(cetuximab/Erbitux®, Merck). Slides previously incu-
bated with the IT were also incubated for further 24 h at
4 °C with TC-1 (diluted 1:40 in PBS + 1 % (v/v) goat
serum) followed by a washing step. The secondary anti-
bodies, either an AP-labeled goat anti-mouse mAb, called
GAMAP, (Southern Biotech, Germany) for the detection
of the IT or an AP-labeled goat-anti-human mAb (Sigma-
Aldrich, Taufkirchen, Germany) for the detection of the
EGFR-specific mAb (Cetuximab/Erbitux®, Merck, Darm-
stadt, Germany), both diluted 1:50 in PBS + 1 % (v/v)
goat serum, were added to the slides, which were then
incubated for 24 h at 4 °C and washed twice in PBS and
once in 0.1 M Tris–HCl (pH 8.5). To generate a red stain-
ing, the AP activity was detected using naphthol AS-BI
phosphate (sodium salt, 50 mg/100 ml; Sigma-Aldrich,
Taufkirchen, Germany) as substrate and new fuchsin
(10 mg/100 ml; Sigma-Aldrich, Taufkirchen, Germany)
as chromogen dissolved in 0.1 M Tris–HCl (pH 8.5).
Endogenous AP activity was inhibited by adding 0.35 mg/
ml levamisole (Sigma-Aldrich, Taufkirchen, Germany)
to the reaction mixture. Finally, we counterstained the
slides with ready-to-use hematoxylin and eosin solutions
according to the manufacturer’s protocol (Sigma-Aldrich,
Taufkirchen, Germany). The slides were analyzed under
a Leica DMR-HC light microscope (Leica, Wetzlar, Ger-
many) using the corresponding Leica QWin software.
J Cancer Res Clin Oncol
1 3
Primary tissue samples were obtained during routine clin-
ical practice at the University Hospital Giessen, in accord-
ance with the principles of the Declaration of Helsinki.
Results
Expression and purification of 425(scFv)-ETA
The IT 425(scFv)-ETA was expressed in E. coli BL-21
(DE3) under osmotic stress in the presence of compatible
solutes (Barth et al. 2000). The imaging probe 425(scFv)-
SNAP was expressed in HEK293T cells. Both proteins
were purified by Ni–NTA affinity chromatography using
the His-tag. The proteins were separated by SDS-PAGE
and detected by immunoblotting using a specific mAb
for the IT (Fig. 1, lane 2) or by coupling the fluorophore
SNAP-Surface® Alexa Fluor®-488 to 425(scFv)-SNAP and
visualizing the conjugate using the Maestro CRi-Imaging
system (Fig. 1, lane 4). Both proteins showed the expected
electrophoretic mobility, corresponding to ~70 kDa for
425(scFv)-ETA and ~48 kDa for 425(scFv)-SNAP (Fig. 1,
lanes 1 and 3). The yield of 425(scFv)-ETA was ~1.5 mg
per liter of bacterial culture, and the yield of 425(scFv)-
SNAP was ~21 mg per liter of cell culture supernatant.
Binding of 425(scFv)-ETA to RMS cell lines
The binding of 425(scFv)-ETA to RMS cell lines was eval-
uated by flow cytometry. The purified IT 425(scFv)-ETA
bound to all three RMS cell lines but not to the control cell
line U937 (Fig. 2). An EGFR-specific mAb was used as a
positive control and showed specific binding to all EGFR+
RMS cell lines but not to the EGFR cell line U937
(Fig. 2). The amount of EGFR was measured using the
Qifikit® kit. The expression profile was similar for all three
RMS cell lines with ~9,413–13,741 receptors expressed on
the surface (Table 1).
Internalization of the 425(scFv) constructs by RMS cell
lines
ITs must be internalized rapidly for greatest efficacy, so we
investigated the internalization behavior of both 425(scFv)
constructs by flow cytometry for the IT (Cizeau et al. 2009)
and live cell imaging for the SNAP-tag construct. Flow
cytometry showed that ~50 % of 425(scFv)-ETA was
internalized within 20 min by RD and FL-OH1 cells and
within 40 min by TE-671 cells. After ~240 min, nearly all
of the IT was internalized by all three RMS cell lines. The
4 °C non-internalization control was set as 100 % in the
evaluation (Fig. 3a).
For the corresponding SNAP-Tag construct, internali-
zation behavior was visualized by confocal microscopy
using SNAP-surface® Alexa Fluor®-488 coupled to the
SNAP-tagged 425(scFv) protein. The fluorophore-labeled
425(scFv)-SNAP construct was efficiently internalized
within 120 min by the RMS cell line RD, resulting in the
intracellular accumulation of the fluorescence signal at
37 °C but not at 4 °C (Fig. 3b). The EGFR cell line U937
is demonstrated in Fig. 3c. No signal could be detected after
incubation of the 425(scFv)-SNAP construct; the counter-
staining of the nucleus was done as for the RD cell line.
The viability of RMS cells is reduced by 425(scFv)-ETA
The ability of 425(scFv)-ETA to inhibit the growth of
EGFR+ cell lines in vitro has been demonstrated with other
tumor cell lines (Bruell et al. 2003, 2005). Here, we used an
XTT assay to confirm that 425(scFv)-ETA also inhibited
the growth of all three RMS cell lines in a dose-dependent
manner. The IT was added in decreasing concentrations to
the target cells, and the proliferation was measured in com-
parison with untreated cells after 72 h of incubation. As
shown in Fig. 4, 425(scFv)-ETA showed specific toxicity
toward the EGFR+ RMS cell lines but not to the EGFR
cell line U937. The non-binding ETA-construct (Mock-
ETA) inhibited the cell proliferation only at a 100x higher
concentration as used for the 425(scFv)-ETA construct.
The 425(scFv)-SNAP imaging probe does not inhibit the
proliferation of the RMS cell line RD, demonstrated exem-
plarily. The IC50 values were in the picomolar range and
are summarized in Table 1.
Fig. 1 The enrichment of the IT 425(scFv)-ETA was determined by
sodium dodecyl sulfate–polyacrylamide gel electrophoresis (SDS-
PAGE) followed by Coomassie brilliant blue staining (lane 1). West-
ern blot analysis of 425(scFv)-ETA was carried out to verify protein
identity (lane 2) using a mouse anti-ETA mAb (TC-1) and a goat
anti-mouse AP-labeled secondary mAb. A pre-stained protein marker
was used. Lanes 3 and 4 show the SDS-PAGE of the 425(scFv)-
SNAP construct. Approximately 500 ng of SNAP-Surface® Alexa
Fluor®-488 labeled scFv-SNAP-tag fusion protein was separated
by SDS-PAGE, visualized by a CRi Maestro Imaging System using
Maestro software and the blue filter set (500–720 nm) to prove the
functionality of the SNAP-tag (lane 4) and stained with Coomassie
brilliant blue (lane 3). Expected size of 425(scFv)-SNAP is ~48 kDa
and of 425(scFv)-ETA ~70 kDa
J Cancer Res Clin Oncol
1 3
The apoptotic impact of 425(scFv)-ETA in one selected
RMS cell line
An AnnexinV/PI assay was used to determine whether the
observed inhibitory effect of 425(scFv)-ETA reflects its
pro-apoptotic activity (Fig. 5). The concentration of 30 nM
IT and the incubation time of 48 h were chosen to capture
the RMS cell line RD within the early- and late-apoptotic
stages. Both populations (early apoptosis in the lower right
corner and late apoptosis in the upper right corner) increased
Fig. 2 Specific cell-binding
activity of the recombinant IT
425(scFv)-ETA and a com-
mercially available rabbit anti-
EGFR monoclonal antibody
(mAb-EGFR) was analyzed by
flow cytometry, using the TC-1
anti-ETA antibody and a FITC-
labeled goat anti-mouse second-
ary mAb for the detection of
the IT and a FITC-labeled goat
anti-rabbit Ab for the mAb-
EGFR (FL-1 fluorescence
channel/FITC). The solid curve
illustrates the binding of mAb-
EGFR, and the dashed curve
represents the binding of the IT
with a twice as high concentra-
tion. Both curves are shown
against the background control
(filled gray curve). U937 was
used as EGFR cell line
Table 1 The EGFR expression level and the IC50 values of 425(scFv)-ETA on different RMS cell lines
The IC50 values indicate the concentrations of 425(scFv)-ETA required to achieve the reduction of protein synthesis by 50 % in comparison
with the untreated control. The values are derived from the colorimetric XTT assays shown in Fig. 4. The EGFR expression on the RMS cell
lines was determined using the Qifikit® kit. The data represent three independent experiments and are presented as mean ± SD. An unspecific
cytotoxic effect was measured by Mock-ETA on all cell lines tested 100 times higher protein concentrations. For 425(scFv)-SNAP no cytotoxic
effect was determined on the cell line RD. Its unspecific cytotoxicity was not determined (n.d.) on other cell lines
Cell line RD FL-OH1 TE-671 U937
EGF-R expression 10,410 (±2,655 SD) 9,413 (±2,626 SD) 13,741 (±4,377 SD)
IC50 [nM]
425(scFv)-ETA
0.63 0.05 0.68 –
IC50 [nM]
Mock-ETA
279.9 16.99 60.13 No effect
425(scFv)-SNAP No effect n.d. n.d. n.d.
J Cancer Res Clin Oncol
1 3
significantly compared to the controls, i.e., the buffer only
and Mock-ETA samples (Fig. 5b). Mock-ETA did not
induce apoptosis compared to the buffer control, whereas
the positive control camptothecin induced apoptosis in both
the EGFR+ cell line RD and the EGFR cell line U937. The
ITs did not induce apoptosis in the control cell line U937.
Fig. 3 Internalization behavior of 425(scFv)-ETA and the cor-
responding 425(scFv)-SNAP into RMS cell lines. In A the three
RMS cell lines RD, FL-OH1 and TE-671 were incubated with 500
nM of the IT 425(scFv)-ETA for 30 min on ice, followed by incu-
bations steps at 37 °C for different points of time (15, 30, 60, 120
and 240 min). One control tube was incubated constantly at 4 °C.
For detection, the murine ETA-specific mAb TC-1 and a PE-labeled
goat anti-mouse mAb were used. For evaluation, the 4 °C control
was set as 100 %. Datasets were normalized using GraphPad Prism
5 software (GraphPad Software). Additionally the internalization of
425(scFv)-SNAP was visualized via live cell imaging using a confo-
cal microscope shown in (b) and (c). In (a), RD cells were incubated
with 100 nM of 425(scFv)-SNAP coupled to SNAP-Surface® Alexa
Fluor®-488. Internalization was monitored after 120 min at 37 °C.
The non-internalization control was incubated at 4 °C. In Fig. 3 C,
the control cell line U937 is demonstrated after incubation with the
425(scFv)-SNAP coupled to SNAP-Surface® Alexa Fluor®-488 at
37 °C as before. No internalization signal could be measured (last
picture); the nucleus was counterstained as for the RD cells. The
first picture in C demonstrates the white light picture of the cell, the
second one the overlay with the counterstained nucleus, the third the
counterstained nucleus in the appropriated fluorescence channel and
the fourth one demonstrated the cell in the fluorescence channel for
the SNAP-Surface® Alexa Fluor®-488-dye
J Cancer Res Clin Oncol
1 3
Ex vivo targeting of EGFR in primary material from RMS
patients
We also tested the binding of 425(scFv)-ETA to FFPE
primary material from RMS patients. Successful binding
was shown for the IT (Fig. 6c, g) and the EGFR-specific
positive control mAb Cetuximab/Erbitux® (Fig. 6i) to sec-
tions representing two different RMS patients. The sections
in Fig. 6a–d are classified as ARMS and those in Fig. 6e–i
as ERMS. Specific binding was confirmed by new fuchsin
and hematoxylin staining. No signal was detected in the
negative control stained only with the detection antibodies
(Fig. 6b, f). The specific binding of the IT and the mAb is
shown in J-L with a higher magnification (objective: 40×).
Discussion
The limited therapeutic options available for RMS consist
of multi-agent chemotherapy, radiotherapy and surgical
resection if appropriate given the tumor site and condi-
tion of the patient (Bisogno et al. 2012; Weiss et al. 2014).
However, the treatment failure rate is high, and new thera-
peutic options are needed urgently especially for relapsed
or metastatic RMS. Immunotherapy using antibody-based
ITs is a promising approach now that third-generation ITs
based on scFv-toxin fusion proteins have demonstrated
success in clinical trials. These are made by recombinant
DNA techniques, combining the scFv of a mAb and tox-
ins lacking their cell-binding domain (Becker and Benhar
2012). Pseudomonas exotoxin A is often used as a com-
ponent of ITs (Weldon and Pastan 2011) because it can be
produced rapidly and efficiently in E. coli, and mutations
that improve its stability and reduce its immunogenicity do
not affect its cytotoxicity (Allen 2002; Becker and Benhar
2012). ITs incorporating scFv-based targeting components
are less immunogenic than full-length antibodies because
most human anti-mouse mAbs are directed against the Fc
domain of used therapeutic mAbs. In addition, the lack of
an Fc domain prevents the binding of the IT to Fc-receptor-
expressing cells. Therefore, it is not rapidly cleared from
the bloodstream or causes undesired cytotoxic effects.
(Thorpe et al. 2003). Furthermore, the smaller size of scFv-
based ITs (~70 kDa) allow more efficient tumor penetra-
tion, which is beneficial for the treatment of solid tumors
such as RMS (Colcher et al. 1998; Yokota et al. 1992).
ITs can also be combined with conventional chemo-
therapy, immunosuppression or radioimmunoconjugates. A
combination of different agents can limit the immunogenic-
ity of each component and increase the overall cytotoxic-
ity (Singh et al. 2012; Wayne et al. 2014). For example,
BL22 is an IT comprising a scFv directed against CD22
Fig. 4 The in vitro cellular cytotoxicity of recombinant 425(scFv)-
ETA to RMS cell lines is shown. The three RMS cell lines RD,
FL-OH1 and TE-671 were incubated with serial dilutions of sterile
425(scFv)-ETA in RPMI medium. As control, the cells were incu-
bated with serial dilutions of a non-specific Mock-ETA as well as
425(scFv)-SNAP as a scFv-control without any toxic compound
(425(scFv)-SNAP was shown exemplarily on RD cells). The cyto-
toxicity assays were performed in duplicates/triplicates at least two
times. Nonlinear regression was performed using GraphPad Prism
5 software (GraphPad Software). The error bars represent standard
error of means. No unspecific effects of the IT or the Mock-ETA
could be demonstrated by using the control cell line U937
J Cancer Res Clin Oncol
1 3
and a version of Pseudomonas exotoxin A known as PE38.
This has been combined with the macrolide lactone bry-
ostatin 1 for the treatment of low-grade and high-grade B
cell lymphoma, with a more potent effect than each agent
alone (Biberacher et al. 2012). Wei et al. (2000) demon-
strated that pre-treatment with a radioimmunoconjugate
improved the effect of an IT in vivo against human B-cell
lymphoma. ITs combined with radioimmunoconjugates are
particularly useful for advanced metastatic cancer and large
tumors because ITs kill individual tumor cells following
internalization, whereas radioimmunoconjugates can kill
also surrounding tumor cells and can penetrate several cell
diameters thus helping to eliminate minimal residual dis-
ease (Ghetie et al. 1994; Wei et al. 2000).
Until recently, RMS has rarely been targeted using
ITs. We lately show the successful killing by an αMCSP-
ETA IT which specifically recognizes CSPG4 on RMS
cell lines (Brehm et al. 2014). Gattenlohner et al. (2010)
Fig. 5 Apoptotic effects of 425(scFv)-ETA on one selected RMS
cell line are shown. RD and U937 (control cell line, EGFR) cells
were incubated with 30 nM 425(scFv)-ETA for 48 h in three inde-
pendent experiments. Dot plots of RD and U937 cells demonstrate
the population of early- and late-apoptotic cells (lower and upper
right corner). The bar chart in B illustrates the sum of early-apop-
totic and late-apoptotic cells. Error bars represent the standard devia-
tion. Differences in cell death relative to the untreated control reach-
ing statistical significance (***p 0.001) are indicated by asterisks.
camptothecin was used as an apoptosis inducing positive control
J Cancer Res Clin Oncol
1 3
achieved promising results using a recombinant scFv-ETA
IT (scFv35-ETA) targeting the fetal acetylcholine receptor,
and Ricci et al. (2002) have targeted RMS indirectly via
EGFR, but to our knowledge, there are no previous reports
of EGFR-specific recombinant single polypeptide chain
ITs for RMS. Ricci et al. (2002) showed that an indirect IT
approach (the application of a murine mAb recognizing the
EGFR followed by a secondary F(ab)2 anti-mouse immu-
noglobulin chemically linked via an artificial disulfide bond
to saporin-S6) can inhibit the growth of RMS cells with an
IC50 of ~950 pM. The IC50 ranges we achieved with our
direct IT approach are better (50–680 pM depending on the
cell line, Table 1) and the single recombinant polypeptide
would be safer and more practicable in the clinic. Another
approach is the conjugation of saporin to cetuximab using
streptavidin, which can be enhanced by photochemical
internalization (Yip et al. 2007). Likewise, a combination
treatment with cetuximab and the cytotoxic agent actino-
mycin D shows antitumor activity in different RMS cell
lines (Yamamoto et al. 2013). There are different EGFR-
specific ITs published, but not specifically for RMS (Wels
et al. 2004; Chandramohan and Bigner 2013).
The purpose of this study was to evaluate the EGFR-
targeting IT 425(scFv)-ETA (Bruell et al. 2003, 2005;
Pardo et al. 2012) for its effect against RMS cells in vitro
and ex vivo. The cell-binding domain of ETA was removed
to generate the truncated version ETA, which was com-
bined with an EGFR-specific scFv to allow the direct tar-
geting of EGFR+ tumor cells and cell killing by receptor-
mediated internalization (Pastan et al. 2007; Weldon and
Pastan 2011). We confirmed specific binding of the IT to
EGFR+ RMS cells, as expected because the homodimeric
425(scFv) has the same binding characteristics as the
parental mAb as measured using a BIAcore instrument
(Muller et al. 1998). The 425 mAb is presumed to bind to
the external domain of the human EGFR, recognizing an
epitope containing amino acid residues G460/S461 which
is close to the EGF-binding site (Kamat et al. 2008; Murthy
Fig. 6 Immunohistochemical staining of primary tumor tissue with
the IT 425(scFv)-ETA and an EGFR-specific mAb is demonstrated.
Tumor biopsies were taken from two different RMS patients. One
patient (ad) was classified as ARMS and the other (e–i) as ERMS.
h, e Staining of tumor specimens demonstrating the presence of
tumor cells in the biopsies of both patients (a, e). b, f Demonstrate
a representative micrograph for the negative control of new fuch-
sin staining with the detection antibodies only (TC-1/GAMAP). c, g
Represent the micrograph of tumor section stained with new fuchsin
substrate using the 425(scFv)-ETA IT detected with TC-1/GAMAP.
d, h Demonstrate the processed samples (background subtracted),
examples of red-stained EGFR+ clusters of tumor cells are indi-
cated by arrows. The positive binding of the EGFR-specific anti-
body cetuximab (Erbitux®) is shown for one selected micrograph in
(i) (size 100 µm, objective: ×10). The micrographs jl were taken
with a higher magnification (size 50 µm, objective: ×40). [j ARMS
425(scFv)-ETA, k ERMS 425(scFv)-ETA and l ERMS EGFR-spe-
cific antibody cetuximab (Erbitux®)]
J Cancer Res Clin Oncol
1 3
et al. 1987). The binding and subsequent internalization of
ITs is necessary for cytotoxic activity. We confirmed the
specific binding and internalization of 425(scFv)-ETA in
three different RMS cell lines, with 50 % of the IT inter-
nalized within 20–40 min depending on the cell line. Spe-
cific internalization was confirmed for the cell line RD
after 120 min, using the corresponding imaging probe
425(scFv)-SNAP labeled with SNAP-Surface® Alexa
Fluor®-488. These internalization results support the data
reported for other scFv-based ITs (Cizeau et al. 2009).
ITs are promising anti-cancer therapeutics because only a
few molecules need to be taken up by cancer cells in order
to kill the cell, because the toxin acts in a catalytic man-
ner (Allen 2002). We tested the cytotoxicity of 425(scFv)-
ETA against RMS cells and calculated IC50 values in the
picomolar range, which is 2.5-fold better for the cell lines
FL-OH1 and RD than the values reported for scFv35-ETA
(Gattenlohner et al. 2010). This reflects the two- to threefold
higher level of EGFR compared to the fetal acetylcholine
receptor, the target antigen of scFv35-ETA. Bachran et al.
(2010) showed a correlation between EGFR expression and
the sensitivity of cervical carcinoma cells toward EGFR-
specific immunotoxins. Our data likewise show that RD and
FL-OH1 cells express EGFR at two- to threefold the level of
the fetal acetylcholine receptor and have a two- to threefold
lower IC50 value for 425(scFv)-ETA compared to scFv35-
ETA. The cell line TE-671 did not fit this profile because the
EGFR was expressed at threefold the level of the fetal ace-
tylcholine receptor, but the IC50 value was 50 times lower.
Gattenlohner et al. (2010) have shown with the ETA-based
IT (scFv35-ETA) also a higher IC50 on TE-671 compared
to RD and FL-OH1, which is comparable to our observa-
tion with an EGFR-specific ETA-based IT. Furthermore,
we were able to demonstrate a reduction in cell viability due
to the induction of apoptosis, which is the common mecha-
nism of ETA-based ITs (Kreitman 2006; Pastan et al. 1992;
Pastan and FitzGerald 1991). The effect of the Mock-ETA at
a about 100× higher concentration as used for the specific IT
was also shown by others (Gattenlohner et al. 2010).
Our immunohistochemistry results confirmed the spe-
cific binding of 425(scFv)-ETA to primary material from
human RMS patients. These findings correlate with other
studies dealing with EGFR expression on RMS primary
material (Ganti et al. 2006; Wachtel et al. 2006).
EGFR expression correlates with tumor treatment resist-
ance (Chong and Janne 2013). Different clinically validated
resistance mechanisms to EGFR-targeted mAbs or small-
molecule inhibitors are known in patients with various can-
cer types, e.g., lung adenocarcinoma. These mechanisms
could involve EGFR mutations that promote drug resist-
ance or the activation of bypass signaling pathways such as
KRAS (Chong and Janne 2013). Furthermore, many differ-
ent tumors are treated with EGFR-specific therapeutics, but
such approaches are often inefficient in monotherapy (Camp
et al. 2005; Sequist and Lynch 2008). For example, EGFR
promotes resistance to radiotherapy due to nuclear shut-
tling and activation of the DNA-repair enzyme DNA pro-
tein kinase (DNA-PK) in certain types of cancer (Dittmann
et al. 2010). Mutations of the intracellular kinase domain
can also reduce the efficacy of tyrosine kinase inhibitors
such as erlotinib and gefitinib (Yun et al. 2008). Ganti et al.
(2006) did not detect mutations of the EGFR in the tested
RMS biopsies, but erlotinib does not inhibit tumor growth
in a genetically engineered ARMS mouse model (Abraham
et al. 2011). The efficacy of ITs is independent of intracel-
lular tyrosine kinase mutations as long as the extracellular
binding region is unaffected (Ratti and Tomasello 2014).
Furthermore, the existence of cancer stem cells (CSCs) with
the capacity for self-renewal and tumor bulk progression
has been reported for RMS (Walter et al. 2011). Mazzo-
leni et al. (2010) proved that EGFR is required to maintain
the CSC phenotype in glioblastomas. If the EGFR is also
required for CSC maintenance in RMS, efficient CSC-tar-
geting therapies would be likely to achieve complete remis-
sion and reduce the likelihood of a relapse. Therefore, the
application of an IT in RMS therapy could be an interesting
and promising approach.
In summary, we are the first to demonstrate the in vitro
efficacy of a recombinant IT targeting the EGFR on dif-
ferent RMS cell lines. The IC50 values were much better
than those reported for an indirect IT approach (Ricci et al.
2002). In preparation for in vivo experiments, we have
shown that the IT can also bind to primary tumor mate-
rial. Although our data need further preclinical in vivo
confirmation, 425(scFv)-ETA appears to be a promising
candidate for clinical development and eventual deploy-
ment as an RMS treatment option to complement current
approaches.
Acknowledgments Christoph Stein was supported by the INTER-
REG IV A project Microbiomed. We would like to thank Rado-
slav Mladenov for his help with the tissue sections. We thank Dr.
Agnieszka Weinandy (University Hospital Aachen, Neurosurgery
Clinic, Aachen, Germany) for providing cetuximab, and we also thank
Dr. Richard M. Twyman for the critical reading of the manuscript.
Conflict of interest None.
Ethical standard Primary tissue samples were obtained during rou-
tine clinical practice at the University Hospital Giessen approved by
the appropriate ethics committee, in accordance with the principles
and the ethical standards of the Declaration of Helsinki.
References
Abraham J et al (2011) Preclinical testing of erlotinib in a trans-
genic alveolar rhabdomyosarcoma mouse model. Sarcoma
2011:130484. doi:10.1155/2011/130484
J Cancer Res Clin Oncol
1 3
Ahmad ZA, Yeap SK, Ali AM, Ho WY, Alitheen NB, Hamid M
(2012) scFv antibody: principles and clinical application. Clin
Dev Immunol 2012:980250. doi:10.1155/2012/980250
Allen TM (2002) Ligand-targeted therapeutics in anticancer therapy.
Nat Rev Cancer 2:750–763. doi:10.1038/nrc903
Amoury M et al (2013) SNAP-tag based agents for preclinical in vitro
imaging in malignant diseases. Curr Pharm Des 19:5429–5436
Antignani A, Fitzgerald D (2013) Immunotoxins: the role of the toxin.
Toxins 5:1486–1502. doi:10.3390/toxins5081486
Armistead PM et al (2007) Expression of receptor tyrosine kinases
and apoptotic molecules in rhabdomyosarcoma: correlation with
overall survival in 105 patients. Cancer 110:2293–2303. doi:10.1
002/cncr.23038
Azemar M et al (2000) Recombinant antibody toxins specific for
ErbB2 and EGF receptor inhibit the in vitro growth of human
head and neck cancer cells and cause rapid tumor regression in
vivo International journal of cancer. J Int Cancer 86:269–275
Bachran D et al (2010) Epidermal growth factor receptor expression
affects the efficacy of the combined application of saponin and
a targeted toxin on human cervical carcinoma cells. Int J Cancer
127:1453–1461. doi:10.1002/ijc.25123
Barth S (2002) Technology evaluation: BL22. NCI current opinion in
molecular therapeutics 4:72–75
Barth S, Huhn M, Matthey B, Klimka A, Galinski EA, Engert A
(2000) Compatible-solute-supported periplasmic expression of
functional recombinant proteins under stress conditions. Appl
Environ Microbiol 66:1572–1579
Becker N, Benhar I (2012) Antibody-based immunotoxins for the treat-
ment of cancer. Antibodies 1:39–69. doi:10.3390/antib1010039
Biberacher V et al (2012) The cytotoxicity of anti-CD22 immuno-
toxin is enhanced by bryostatin 1 in B-cell lymphomas through
CD22 upregulation and PKC-betaII depletion. Haematologica
97:771–779. doi:10.3324/haematol.2011.049155
Bisogno G et al (2012) Rhabdomyosarcoma in adolescents: a report
from the AIEOP soft tissue sarcoma committee. Cancer 118:821–
827. doi:10.1002/cncr.26355
Brehm H et al (2014) A CSPG4-specific immunotoxin kills rhabdo-
myosarcoma cells and binds to primary tumor tissues Cancer
Lett. doi:10.1016/j.canlet.2014.07.006
Bruell D et al (2003) The recombinant anti-EGF receptor immuno-
toxin 425(scFv)-ETA suppresses growth of a highly metastatic
pancreatic carcinoma cell line. Int J Oncol 23:1179–1186
Bruell D et al (2005) Recombinant anti-EGFR immunotoxin 425(scFv)-
ETA demonstrates anti-tumor activity against disseminated human
pancreatic cancer in nude mice. Int J Mol Med 15:305–313
Camp ER, Summy J, Bauer TW, Liu W, Gallick GE, Ellis LM (2005)
Molecular mechanisms of resistance to therapies targeting the
epidermal growth factor receptor. Clin Cancer Res 11:397–405
Cen L et al (2007) Phosphorylation profiles of protein kinases in
alveolar and embryonal rhabdomyosarcoma. Mod Pathol 20:936–
946. doi:10.1038/modpathol.3800834
Chandramohan V, Bigner D (2013) A novel recombinant immu-
notoxin-based therapy targeting wild-type and mutant EGFR
improves survival in murine models of glioblastoma. OncoImmu-
nology 2(12):e26852
Chong CR, Janne PA (2013) The quest to overcome resistance to
EGFR-targeted therapies in cancer. Nat Med 19:1389–1400.
doi:10.1038/nm.3388
Cizeau J, Grenkow DM, Brown JG, Entwistle J, MacDonald GC
(2009) Engineering and biological characterization of VB6-
845, an anti-EpCAM immunotoxin containing a T-cell epitope-
depleted variant of the plant toxin bouganin. J Immunother
32:574–584. doi:10.1097/CJI.0b013e3181a6981c
Colcher D, Pavlinkova G, Beresford G, Booth BJ, Choudhury A,
Batra SK (1998) Pharmacokinetics and biodistribution of genet-
ically-engineered antibodies. Q J Nucl Med 42:225–241
Davicioni E et al (2009) Molecular classification of rhabdomyo-
sarcoma–genotypic and phenotypic determinants of diagno-
sis: a report from the Children’s Oncology Group. Am J Pathol
174:550–564. doi:10.2353/ajpath.2009.080631
De Giovanni C et al (1996) Antisense epidermal growth factor recep-
tor transfection impairs the proliferative ability of human rhabdo-
myosarcoma cells. Cancer Res 56:3898–3901
Dittmann K, Mayer C, Fehrenbacher B, Schaller M, Kehlbach R,
Rodemann HP (2010) Nuclear EGFR shuttling induced by ion-
izing radiation is regulated by phosphorylation at residue Thr654.
FEBS Lett 584:3878–3884. doi:10.1016/j.febslet.2010.08.005
Ganti R et al (2006) Expression and genomic status of EGFR and
ErbB-2 in alveolar and embryonal rhabdomyosarcoma. Mod
Pathol 19:1213–1220. doi:10.1038/modpathol.3800636
Gattenlohner S et al (2010) A human recombinant autoantibody-
based immunotoxin specific for the fetal acetylcholine recep-
tor inhibits rhabdomyosarcoma growth in vitro and in a murine
transplantation model. J Biomed Biotechnol 2010:187621.
doi:10.1155/2010/187621
Ghetie MA, Tucker K, Richardson J, Uhr JW, Vitetta ES (1994) Erad-
ication of minimal disease in severe combined immunodeficient
mice with disseminated Daudi lymphoma using chemotherapy
and an immunotoxin cocktail. Blood 84:702–707
Green MR, Sambrook J (2012) Molecular cloning: a laboratory man-
ual. Cold Spring Harbor Laboratory Press, New York
Hawkins DS, Gupta AA, Rudzinski ER (2014) What is new in the
biology and treatment of pediatric rhabdomyosarcoma? Curr
Opin Pediatr 26:50–56. doi:10.1097/MOP.0000000000000041
Hristodorov D et al (2013) Microtubule-associated protein tau facili-
tates the targeted killing of proliferating cancer cells in vitro
and in a xenograft mouse tumour model in vivo. Br J Cancer
109:1570–1578. doi:10.1038/bjc.2013.457
Hussain AF, Amoury M, Barth S (2013) SNAP-tag technology: a
powerful tool for site specific conjugation of therapeutic and
imaging agents. Curr Pharm Des 19:5437–5442
Jain S, Xu R, Prieto VG, Lee P (2010) Molecular classification of soft
tissue sarcomas and its clinical applications Int J. Clin Exp Pathol
3:416–428
Kamat V et al (2008) Enhanced EGFR inhibition and distinct epitope
recognition by EGFR antagonistic mAbs C225 and 425. Cancer
Biol Ther 7:726–733
Kampmeier F, Ribbert M, Nachreiner T, Dembski S, Beaufils F,
Brecht A, Barth S (2009) Site-specific, covalent labeling of
recombinant antibody fragments via fusion to an engineered ver-
sion of 6-O-alkylguanine DNA alkyltransferase. Bioconjug Chem
20:1010–1015. doi:10.1021/bc9000257
Kampmeier F et al (2010) Rapid optical imaging of EGF receptor
expression with a single-chain antibody SNAP-tag fusion pro-
tein. Eur J Nucl Med Mol Imaging 37:1926–1934. doi:10.1007/
s00259-010-1482-5
Koefoed K et al (2011) Rational identification of an optimal antibody
mixture for targeting the epidermal growth factor receptor. mAbs
3:584–595. doi:10.4161/mabs.3.6.17955
Kreitman RJ (2006) Immunotoxins for targeted cancer therapy. AAPS
j 8:E532–E551. doi:10.1208/aapsj080363
Laemmli UK (1970) Cleavage of structural proteins during the assem-
bly of the head of bacteriophage T4. Nature 227:680–685
Mazzoleni S et al (2010) Epidermal growth factor receptor expression
identifies functionally and molecularly distinct tumor-initiating
cells in human glioblastoma multiforme and is required for glio-
magenesis. Cancer Res 70:7500–7513. doi:10.1158/0008-5472.
can-10-2353
Mendelsohn J (2002) Targeting the epidermal growth factor receptor
for cancer therapy. J Clin Oncol 20:1s–13s
Missiaglia E et al (2012) PAX3/FOXO1 fusion gene status is the
key prognostic molecular marker in rhabdomyosarcoma and
J Cancer Res Clin Oncol
1 3
significantly improves current risk stratification. J Clin Oncol
30:1670–1677. doi:10.1200/jco.2011.38.5591
Muller KM, Arndt KM, Strittmatter W, Pluckthun A (1998) The first
constant domain (C(H)1 and C(L)) of an antibody used as het-
erodimerization domain for bispecific miniantibodies. FEBS Lett
422:259–264
Murthy U, Basu A, Rodeck U, Herlyn M, Ross AH, Das M (1987)
Binding of an antagonistic monoclonal antibody to an intact and
fragmented EGF-receptor polypeptide. Arch Biochem Biophys
252:549–560
Pardo A, Stocker M, Kampmeier F, Melmer G, Fischer R, Thepen
T, Barth S (2012) In vivo imaging of immunotoxin treatment
using Katushka-transfected A-431 cells in a murine xenograft
tumour model. Cancer Immunol Immunother 61:1617–1626.
doi:10.1007/s00262-012-1219-3
Pastan I, FitzGerald D (1991) Recombinant toxins for cancer treat-
ment. Science 254:1173–1177
Pastan I, Chaudhary V, FitzGerald DJ (1992) Recombinant toxins
as novel therapeutic agents. Annu Rev Biochem 61:331–354.
doi:10.1146/annurev.bi.61.070192.001555
Pastan I, Hassan R, FitzGerald DJ, Kreitman RJ (2007) Immu-
notoxin treatment of cancer. Annu Rev Med 58:221–237.
doi:10.1146/annurev.med.58.070605.115320
Pedersen MW, Jacobsen HJ, Koefoed K, Hey A, Pyke C, Haurum
JS, Kragh M (2010) Sym004: a novel synergistic anti-epidermal
growth factor receptor antibody mixture with superior antican-
cer efficacy. Cancer Res 70:588–597. doi:10.1158/0008-5472.
CAN-09-1417
Ratti M, Tomasello G (2014) Emerging combination therapies to
overcome resistance in EGFR-driven tumors. Anticancer Drugs
25:127–139. doi:10.1097/cad.0000000000000035
Ray A, Huh WW (2012) Current state-of-the-art systemic therapy
for pediatric soft tissue sarcomas. Curr Oncol Rep 14:311–319.
doi:10.1007/s11912-012-0243-y
Ricci C et al (2002) HER/erbB receptors as therapeutic targets of
immunotoxins in human rhabdomyosarcoma cells. J Immunother
25:314–323
Schiffer S et al (2013) Species-dependent functionality of the human
cytolytic fusion proteins granzyme B-H22(scFv) and H22(scFv)-
angiogenin in macrophages. Antibodies 2:9–18. doi:10.3390/an
tib2010009
Sequist LV, Lynch TJ (2008) EGFR tyrosine kinase inhibitors in
lung cancer: an evolving story. Annu Rev Med 59:429–442.
doi:10.1146/annurev.med.59.090506.202405
Simon-Keller K, Barth S, Vincent A, Marx A (2013) Targeting the
fetal acetylcholine receptor in rhabdomyosarcoma. Expert Opin
Ther Targets 17:127–138. doi:10.1517/14728222.2013.734500
Singh R, Samant U, Hyland S, Chaudhari PR, Wels WS, Bandyo-
padhyay D (2007) Target-specific cytotoxic activity of recom-
binant immunotoxin scFv(MUC1)-ETA on breast carcinoma
cells and primary breast tumors. Mol Cancer Ther 6:562–569.
doi:10.1158/1535-7163.MCT-06-0604
Singh R, Zhang Y, Pastan I, Kreitman RJ (2012) Synergistic anti-
tumor activity of anti-CD25 recombinant immunotoxin
LMB-2 with chemotherapy. Clin Cancer Res 18:152–160.
doi:10.1158/1078-0432.ccr-11-1839
Stahnke B et al (2008) Granzyme B-H22(scFv), a human
immunotoxin targeting CD64 in acute myeloid leukemia
of monocytic subtypes. Mol Cancer Ther 7:2924–2932.
doi:10.1158/1535-7163.MCT-08-0554
Stocker M, Tur MK, Sasse S, Krussmann A, Barth S, Engert A (2003)
Secretion of functional anti-CD30-angiogenin immunotoxins
into the supernatant of transfected 293T-cells. Protein Expr Purif
28:211–219
Sumegi J, Streblow R, Frayer RW, Dal Cin P, Rosenberg A, Mel-
oni-Ehrig A, Bridge JA (2010) Recurrent t(2;2) and t(2;8)
translocations in rhabdomyosarcoma without the canonical PAX-
FOXO1 fuse PAX3 to members of the nuclear receptor transcrip-
tional coactivator family. Genes Chromosom Cancer 49:224–236.
doi:10.1002/gcc.20731
Tebbutt N, Pedersen MW, Johns TG (2013) Targeting the ERBB
family in cancer: couples therapy. Nat Rev Cancer 13:663–673.
doi:10.1038/nrc3559
Thorpe SJ, Turner C, Heath A, Feavers I, Vatn I, Natvig JB, Thomp-
son KM (2003) Clonal analysis of a human antimouse antibody
(HAMA) response. Scand J Immunol 57:85–92
Tur MK et al (2003) Recombinant CD64-specific single chain immu-
notoxin exhibits specific cytotoxicity against acute myeloid leu-
kemia cells. Cancer Res 63:8414–8419
Wachtel M et al (2006) Subtype and prognostic classification of rhab-
domyosarcoma by immunohistochemistry. J Clin Oncol 24:816–
822. doi:10.1200/jco.2005.03.4934
Walter D et al (2011) CD133 positive embryonal rhabdomyosarcoma
stem-like cell population is enriched in rhabdospheres. PLoS
ONE 6:e19506. doi:10.1371/journal.pone.0019506
Wayne AS, Fitzgerald DJ, Kreitman RJ, Pastan I (2014) Immu-
notoxins for leukemia. Blood 123:2470–2477. doi:10.1182/
blood-2014-01-492256
Wei BR, Ghetie MA, Vitetta ES (2000) The combined use of an
immunotoxin and a radioimmunoconjugate to treat disseminated
human B-cell lymphoma in immunodeficient mice. Clin Cancer
Res 6:631–642
Weidle UH, Tiefenthaler G, Schiller C, Weiss EH, Georges G, Brink-
mann U (2014) Prospects of bacterial and plant protein-based
immunotoxins for treatment of cancer. Cancer Genomics Prot-
eomics 11:25–38
Weiss A, Gill J, Goldberg J, Lagmay J, Spraker-Perlman H, Ven-
katramani R, Reed D (2014) Advances in therapy for pedi-
atric sarcomas. Curr Oncol Rep 16:395. doi:10.1007/
s11912-014-0395-z
Weldon JE, Pastan I (2011) A guide to taming a toxin–recom-
binant immunotoxins constructed from Pseudomonas exo-
toxin A for the treatment of cancer. FEBS J 278:4683–4700.
doi:10.1111/j.1742-4658.2011.08182.x
Wels W et al (2004) Recombinant immunotoxins and retargeted
killer cells: employing engineered antibody fragments for tumor-
specific targeting of cytotoxic effectors. Cancer Immunol Immu-
nother 53:217–226. doi:10.1007/s00262-003-0482-8
Williamson D et al (2010) Fusion gene-negative alveolar rhabdo-
myosarcoma is clinically and molecularly indistinguishable from
embryonal rhabdomyosarcoma. J Clin Oncol 28:2151–2158. doi:
10.1200/jco.2009.26.3814
Yamamoto Y et al (2013) Cetuximab promotes anticancer drug tox-
icity in rhabdomyosarcomas with EGFR amplification in vitro.
Oncol Rep 30:1081–1086. doi:10.3892/or.2013.2588
Yewale C, Baradia D, Vhora I, Patil S, Misra A (2013) Epidermal growth
factor receptor targeting in cancer: a review of trends and strategies.
Biomaterials 34:8690–8707. doi:10.1016/j.biomaterials.2013.07.100
Yip WL, Weyergang A, Berg K, Tonnesen HH, Selbo PK (2007) Tar-
geted delivery and enhanced cytotoxicity of cetuximab-saporin
by photochemical internalization in EGFR-positive cancer cells.
Mol Pharm 4:241–251. doi:10.1021/mp060105u
Yokota T, Milenic DE, Whitlow M, Schlom J (1992) Rapid tumor
penetration of a single-chain Fv and comparison with other
immunoglobulin forms. Cancer Res 52:3402–3408
Yun CH et al (2008) The T790 M mutation in EGFR kinase causes
drug resistance by increasing the affinity for ATP. Proc Natl Acad
Sci USA 105:2070–2075. doi:10.1073/pnas.0709662105
Zanola A, Rossi S, Faggi F, Monti E, Fanzani A (2012) Rhabdomyo-
sarcomas: an overview on the experimental animal models. J Cell
Mol Med 16:1377–1391. doi:10.1111/j.1582-4934.2011.01518.x
... 18,19 Recently, EGFR-specific immunotoxins and a human cytolytic fusion protein have shown promising results in vitro against ERMS cell lines among others. [20][21][22] Here, we describe the development of next-generation ADCs in which a single-chain fragment variable (scFv) instead of a full-length monoclonal antibody (mAb) is used, and the scFv is combined with a SNAP-tag to facilitate covalent coupling to auristatin F (AURIF) via benzylguanine (BG). AURIF is a BG-modified version of MMAF suitable for SNAP-tag coupling. ...
... Paraffin was removed from formalin-fixed paraffinembedded (FFPE) tumor biopsy samples as previously described. 20 After drying, circles were drawn around the tissue sections on the slides using a Dako Pen (Dako Denmark A/S, Glostrup, Denmark). After blocking in PBS supplemented with 1% (v/v) bovine serum albumin (BSA), the slides were incubated at 4°C overnight with the following constructs in PBS: 425(scFv)-SNAP, 1711(scFv)-SNAP, and mock-SNAP, all conjugated with AURIF. ...
... After washing with PBS, AP activity was detected as previously described. 20,21 All slides were counterstained using hematoxylin and eosin (Sigma-Aldrich, St Louis, MI, USA). Images were captured using a Leica DMR-HC light microscope with the Leica QWin software (Leica Microsystems, Wetzlar, Germany). ...
Article
Full-text available
Abstract: Antibody–drug conjugates (ADCs) can deliver toxins to specific targets such as tumor cells. They have shown promise in preclinical/clinical development but feature stoichiometrically undefined chemical linkages, and those based on full-size antibodies achieve only limited tumor penetration. SNAP-tag technology can overcome these challenges by conjugating benzylguanine-modified toxins to single-chain fragment variables (scFvs) with 1:1 stoichio­metry while preserving antigen binding. Two (human and mouse) scFv-SNAP fusion proteins recognizing the epidermal growth factor receptor (EGFR) were expressed in HEK 293T cells. The purified fusion proteins were conjugated to auristatin F (AURIF). Binding activity was confirmed by flow cytometry/immunohistochemistry, and cytotoxic activity was confirmed by cell viability/apoptosis and cell cycle arrest assays, and a novel microtubule dynamics disassembly assay was performed. Both ADCs bound specifically to their target cells in vitro and ex vivo, indicating that the binding activity of the scFv-SNAP fusions was unaffected by conjugation to AURIF. Cytotoxic assays confirmed that the ADCs induced apoptosis and cell cycle arrest at nanomolar concentrations and microtubule disassembly. The SNAP-tag technology provides a platform for the development of novel ADCs with defined conjugation sites and stoichiometry. We achieved the stable and efficient linkage of AURIF to human or murine scFvs using the SNAP-tag technology, offering a strategy to improve the development of personalized medicines.
... Similar to IGF1R and VEGF(R), combined therapy of the anti-EGFR antibody cetuximab and the chemotherapeutic dactinomycin was superior to dactinomycin treatment alone in EGFRexpressing ARMS and ERMS cell lines (22). Despite the fact that EGFR overexpression is present in 37%-76% of ERMS and 16%-50% of ARMS (23), no in vivo or clinical trials have been described for EGFR-based combination treatment in rhabdomyosarcoma; hence, no solid conclusions can be drawn regarding the potential of this combination in the clinic. A novel way of targeting EGFR in combination with cytotoxic compounds was recently introduced, by creating antibody-drug conjugations. ...
... Further analysis showed binding of the IT to EGFR-expressing rhabdomyosarcoma cell lines and reduced cell viability with induction of apoptosis. Moreover, the IT was shown to bind to EGFR-expressing rhabdomyosarcoma tumor tissue (23). Niesen and colleagues more recently developed fully human cytolytic fusion proteins (hCFP) by conjugating serine protease granzyme B to EGFR scFvs. ...
Article
Full-text available
Targeted therapies have revolutionized cancer treatment; however, progress lags behind in alveolar (ARMS) and embryonal rhabdomyosarcoma (ERMS), a soft-tissue sarcoma mainly occurring at pediatric and young adult age. Insulin-like growth factor 1 receptor (IGF1R)-directed targeted therapy is one of the few single-agent treatments with clinical activity in these diseases. However, clinical effects only occur in a small subset of patients and are often of short duration due to treatment resistance. Rational selection of combination treatments of either multiple targeted therapies or targeted therapies with chemotherapy could hypothetically circumvent treatment resistance mechanisms and enhance clinical efficacy. Simultaneous targeting of distinct mechanisms might be of particular interest in this regard, as this affects multiple hallmarks of cancer at once. To determine the most promising and clinically relevant targeted therapy-based combination treatments for ARMS and ERMS, we provide an extensive overview of preclinical and (early) clinical data concerning a variety of targeted therapy-based combination treatments. We concentrated on the most common classes of targeted therapies investigated in rhabdomyosarcoma to date, including those directed against receptor tyrosine kinases and associated downstream signaling pathways, the Hedgehog signaling pathway, apoptosis pathway, DNA damage response, cell-cycle regulators, oncogenic fusion proteins, and epigenetic modifiers. Mol Cancer Ther; 17(7); 1365-80. ©2018 AACR.
... PCa biopsy specimen of four patients with early-to-intermediate stage (Gleason scores 6-7) is moderately differentiated, whereas prostatic chips obtained by transurethral resection of the prostate (TURP) of one patient (Gleason score 9) represent poorly differentiated advanced PCa. The slides were rehydrated and dewaxed as described elsewhere (Niesen et al. 2015a). Sections were circled using a Dako-pen (Dako). ...
... After washing twice in PBS/Tween and once in Tris-HCl (0.1 M, pH 8.5), AP activity was detected using New Fuchsin staining. Subsequent counterstaining with hematoxylin and eosin (H&E) was performed as described elsewhere (Niesen et al. 2015a). Tissue sections were analyzed using an Olympus BX 41 light microscope (Olympus, Hamburg, Germany) and ProgRes ® CapturePro Version 2.8.8 software (Jenoptik Optical Systems, Jena). ...
Article
Full-text available
Purpose: Despite great progress in the diagnosis and treatment of localized prostate cancer (PCa), there remains a need for new diagnostic markers that can accurately distinguish indolent and aggressive variants. One promising approach is the antibody-based targeting of prostate stem cell antigen (PSCA), which is frequently overexpressed in PCa. Here, we show the construction of a molecular imaging probe comprising a humanized scFv fragment recognizing PSCA genetically fused to an engineered version of the human DNA repair enzyme O6-alkylguanine-DNA alkyltransferase (AGT), the SNAP-tag, enabling specific covalent coupling to various fluorophores for diagnosis of PCa. Furthermore, the recombinant immunotoxin (IT) PSCA(scFv)-ETA' comprising the PSCA(scFv) and a truncated version of Pseudomonas exotoxin A (PE, ETA') was generated. Methods: We analyzed the specific binding and internalization behavior of the molecular imaging probe PSCA(scFv)-SNAP in vitro by flow cytometry and live cell imaging, compared to the corresponding IT PSCA(scFv)-ETA'. The cytotoxic activity of PSCA(scFv)-ETA' was tested using cell viability assays. Specific binding was confirmed on formalin-fixed paraffin-embedded tissue specimen of early and advanced PCa. Results: Alexa Fluor(®) 647 labeling of PSCA(scFv)-SNAP confirmed selective binding to PSCA, leading to rapid internalization into the target cells. The recombinant IT PSCA(scFv)-ETA' showed selective binding leading to internalization and efficient elimination of target cells. Conclusions: Our data demonstrate, for the first time, the specific binding, internalization, and cytotoxicity of a scFv-based fusion protein targeting PSCA. Immunohistochemical staining confirmed the specific ex vivo binding to primary PCa material.
... The cytotoxicity observed in this study is comparable to that of scFv1711-ETA, another immunotoxin consisting of the scFv of panitumumab linked to domains II and III of PE (PE40/ETA). ScFv1711-ETA demonstrated specific cytotoxicity against EGFR with an IC50 of 32 pM on the MDA-MB-468 cell line (Niesen et al., 2015), indicating its superior potency compared to RTA-scFv immunotoxin. ...
Article
The Epidermal Growth Factor Receptor (EGFR) has been of high importance as it is over expressed in a wide diversity of epithelial cancers, promoting cell proliferation and survival pathways. Recombinant immunotoxins (ITs) have emerged as a promising targeted therapy for cancer treatment. In this study, we aimed to investigate the antitumor activity of a novel recombinant immunotoxin designed against EGFR. Using an in silico approach, we confirmed the stability of the RTA-scFv fusion protein. The immunotoxin was successfully cloned and expressed in the pET32a vector, and the purified protein was analyzed by electrophoresis and western blotting. In vitro evaluations were conducted to assess the biological activities of the recombinant proteins (RTA-scFv, RTA, scFv). The novel immunotoxin demonstrated significant anti-proliferative and pro-apoptotic effects against cancer cell lines. The MTT cytotoxicity assay revealed a decrease in cell viability in the treated cancer cell lines. Additionally, Annexin V/Propidium iodide staining followed by flow cytometry analysis showed a significant induction of apoptosis in the cancer cell lines, with half maximal inhibitory concentration (IC50) values of 81.71 nM for MDA-MB-468 and 145.2 nM for HCT116 cells (P < 0.05). Furthermore, the EGFR-specific immunotoxin exhibited non-allergenic properties. The recombinant protein demonstrated high affinity binding to EGFR. Overall, this study presents a promising strategy for the development of recombinant immunotoxins as potential candidates for the treatment of EGFR-expressing cancers.
... Moreover, 425(scFv)-ETA was able to reduce cell viability (IC 50 values in picomolar range) and to strongly activate apoptotic pathway. The EGFR + cell binding activity of the IT 425(scFv)-ETA was also demonstrated ex vivo on two patient-derived formalin-fixed paraffin-embedded RMS specimens [86]. EGFR was also used as target of an indirect IT, consisting of a primary EGFR specific mAb followed by a secondary F(ab') 2 anti-mouse Ig linked to saporin-S6. ...
Article
Full-text available
Sarcomas are one of the most difficult type of cancer to manage and treat because of their extremely heterogeneous molecular and morphological features. Despite the progress made over the years in the establishment of standard protocols for high and low grading/staging sarcoma patients, mostly with chemotherapy and/or radiotherapy, 50% of treated patients experience relapse episodes. Because of this, in the last 20 years, new therapeutic approaches for sarcoma treatment have been evaluated in preclinical and clinical studies. Among them, antibody-based therapies have been the most studied. Immunoconjugates consist of a carrier portion, frequently represented by an antibody, linked to a toxic moiety, i.e., a drug, toxin, or radionuclide. While the efficacy of immunoconjugates is well demonstrated in the therapy of hematological tumors and more recently also of epithelial ones, their potential as therapeutic agents against sarcomas is still not completely explored. In this paper, we summarize the results obtained with immunoconjugates targeting sarcoma surface antigens, considering both preclinical and clinical studies. To date, the encouraging results obtained in preclinical studies allowed nine immunoconjugates to enter clinical trials, demonstrating the validity of immunotherapy as a promising pharmacological tool also for sarcoma therapy.
... The formalin-fixed paraffin-embedded (FFPE) ovarian biopsies were prepared as previously described, 48 blocked with 1% (v/v) bovine serum albumin in PBS for 1 h and incubated with either CLIP-scFv-425-SNAP or scFv-425-SNAP at 4°C overnight. The slides were washed with PBS and incubated with the M2D11 anti-SNAP antibody 49 for 3 h at room temperature. ...
Article
Antibody-based diagnostic and therapeutic reagents armed with effector molecules such as dyes and drugs offer hope in the battle against cancer. Several site-specific conjugation methods have been developed to equip antibodies with such effector molecules, but they tend to be expensive and involve multiple reaction steps. The conjugation of two different effector molecules to a single antibody also remains a major challenge. Here we describe a simple, controlled and robust method for the dual site-specific conjugation of an antibody with two effector molecules in a single-pot reaction using the self-labeling SNAP and CLIP protein tags. We verified the principle of the method by labeling an epidermal growth factor receptor (EGFR)-specific single-chain antibody fragment (scFv-425) simultaneously with IRDye®700 and Alexa-Fluor®647. This dual-labeled antibody bound to EGFR+ ovarian cancer cell lines and tissue samples with high specificity and its phototherapeutic efficacy was confirmed by the selective killing of EGFR+ cells in vitro.
... For example, bacterial and plant toxins, enzymes and cytotoxic factors of human origin (apoptosis-inducing factor AIF-1, granzymes, RNAse), as well as protein photosensitizers, among others. Several targeted toxins based on recombinant antibodies (immunotoxins) specific to the receptors of the EGFR family undergo preclinical [63][64][65][66][67][68][69][70][71][72][73][74][75][76][77] and clinical [78][79][80] trials. In addition, the use of alternative scaffold proteins, in particular designed ankyrin repeat proteins In vitro models of tumors expressing EGFR family receptors: 2D monolayer culture (a), tumor speroids of one-cell and multi-cell types (b), tumor models based on natural and synthetic matrices, including organotypic models (c), and microfluidics-based models (d). ...
Article
Carcinomas overexpressing EGFR family receptors are of high clinical importance, because the receptors have prognostic value and are used as molecular targets for anticancer therapy. Insufficient drug efficacy necessitates further in-depth research of the receptor biology and improvement in preclinical stages of drug evaluation. Here, we review the currently used advanced 3D in vitro models of tumors, including tumor spheroids, models in natural and synthetic matrices, tumor organoids and microfluidic-based models, as a potent tool for studying EGFR biology and targeted drug development. We are especially focused on factors that affect the biology of tumor cells, causing modification in the expression and basic phosphorylation of the receptors, crosstalk with other signaling pathways and switch between downstream cascades, resulting ultimately in the resistance to antitumor agents.
... We therefore used the established pMS vector system [53] to prepare fusion constructs combining two human effector proteins with TTC as a model antigen, resulting in two novel TTC-based fusion proteins: EGrB(R201K)-TTC and TTC-MAPTau. These proteins were expressed successfully following the transfection of HEK 293T cells, and the expression yield of the protein EGrB(R201K)-TTC we achieved (4.5 mg/l) is comparable to the yields of other granzyme B fusion proteins produced in HEK 293T cells, ranging from 1 to 30 mg/l [30,32,62]. The yield of TTC-MAPTau was 1.6 mg/l, which is comparable to other MAPTau fusion proteins such as Ki4scFv-MAP and anti-EpCAMscFv-MAP produced in E. coli with yields of up to 1 mg/l [60,63,64]. ...
Article
Full-text available
The antigen-specific targeting of autoreactive B cells via their unique B cell receptors (BCRs) is a novel and promising alternative to the systemic suppression of humoral immunity. We generated and characterized cytolytic fusion proteins based on an existing immunotoxin comprising tetanus toxoid fragment C (TTC) as the targeting component and the modified Pseudomonas aeruginosa exotoxin A (ETA') as the cytotoxic component. The immunotoxin was reconfigured to replace ETA' with either the granzyme B mutant R201K or MAPTau as human effector domains. The novel cytolytic fusion proteins were characterized with a recombinant human lymphocytic cell line developed using Transpo-mAb™ technology. Genes encoding a chimeric TTC-reactive immunoglobulin G were successfully integrated into the genome of the precursor B cell line REH so that the cells could present TTCreactive BCRs on their surface. These cells were used to investigate the specific cytotoxicity of GrB(R201K)-TTC and TTC-MAPTau, revealing that the serpin proteinase inhibitor 9- resistant granzyme B R201K mutant induced apoptosis specifically in the lymphocytic cell line. Our data confirm that antigen-based fusion proteins containing granzyme B (R201K) are suitable candidates for the depletion of autoreactive B cells. © 2017 Klose et al. This is an open access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Article
Objectives Epithelial growth factor receptor (EGFR), as a malignancy marker, is overly expressed in multiple solid tumors including colorectal neoplasms, one of the most prevalent malignancies worldwide. The main objective of this study is to enhance the efficacy of anti-tumor therapy targeting EGFR by constructing a novel EGFR-specific immunotoxin (C-CUS245C) based on Cetuximab and recombinant Cucurmosin (CUS245C).MethodsE. coli BL21 (DE3) PlysS (E. coli) was used to express CUS245C with a cysteine residue inserting to the C-terminus of Cucurmosin. Then immobilized metal ion affinity chromatography (IMAC) was used to purify CUS245C. The chemical conjugation method was used for the preparation of C-CUS245C. Then dialysis and IMAC were used to purify C-CUS245C. Western blot as well as SDS-PAGE was carried out to characterize the formation of C-CUS245C. At last the anti-colorectal cancer activity of C-CUS245C was investigated in vitro and in vivo.ResultsCUS245C with high purity could be obtained from the prokaryotic system. C-CUS245C was successfully constructed and highly purified. The cytotoxicity assays in vitro showed a significant proliferation inhibition of C-CUS245C on EGFR-positive cells for 120 h with IC50 values less than 0.1 pM. Besides, the anti-tumor efficacy of C-CUS245C was remarkably more potent than that of Cetuximab, CUS245C, and C + CUS245C (P < 0.001). Whereas the cytotoxicity of C-CUS245C could hardly be detected on EGFR-null cell line. Our results also showed that C-CUS245C had efficacy of anti-colorectal cancer in mouse xenograft model, indicating the therapeutic potential of C-CUS245C for the targeted therapy of colorectal neoplasms.ConclusionsC-CUS245C exhibits potent and EGFR-specific cytotoxicity. Insertional mutagenesis technique is worthy to be adopted in the preparation of immunotoxin. Immunotoxin can be highly purified through dialysis followed by IMAC.
Article
Full-text available
In vitro and ex vivo development of novel therapeutic agents requires reliable and accurate analyses of the cell conditions they were preclinical tested for, such as apoptosis. The detection of apoptotic cells by annexin V (AV) coupled to fluorophores has often shown limitations in the choice of the dye due to interference with other fluorescent-labeled cell markers. The SNAP-tag technology is an easy, rapid and versatile method for functionalization of proteins and was therefore used for labeling AV with various fluorophores. We generated the fusion protein AV-SNAP and analyzed its capacity for the specific display of apoptotic cells in various assays with therapeutic agents. AV-SNAP showed an efficient coupling reaction with five different fluorescent dyes. Two selected fluorophores were tested with suspension, adherent and peripheral blood cells, treated by heat-shock or apoptosis-inducing therapeutic agents. Flow cytometry analysis of apoptotic cells revealed a strong visuali-zation using AV-SNAP coupled to these two fluorophores exemplary, which was comparable to a commercial AV-Assay-kit. The combination of the apoptosis-specific binding protein AV with the SNAP-tag provides a novel solid method to facilitate protein labeling using several, easy to change, fluorescent dyes at once. It avoids high costs and allows an ordinary exchange of dyes and easier use of other fluorescent-labeled cell markers, which is of high interest for the preclinical testing of therapeutic agents in e.g. cancer research.plos one
Article
Full-text available
Antibody-based immunotoxins comprise an important group in targeted cancer therapeutics. These chimeric proteins are a form of biological guided missiles that combine a targeting moiety with a potent effector molecule. The targeting moiety is mostly a monoclonal antibody (MAb) or a recombinant antibody-based fragment that confers target specificity to the immunotoxin. The effector domain is a potent protein toxin of bacterial or plant origin, which, following binding to the target cells, undergoes internalization and causes cell death. Over time and following research progression, immunotoxins become better fitted to their purpose, losing immunogenic fragments and non-specific targeting moieties. Many immunotoxins have gone through clinical evaluation. Some of these have been shown to be active and work is progressing with them in the form of further clinical trials. Others, mostly developed in the previous century, failed to generate a response in patients, or even caused undesired side effects. This article reviews the antibody and protein-toxin based immunotoxins that were clinically evaluated up to the present day.
Article
Full-text available
Pediatric sarcomas are relatively rare malignancies individually. As a group they are typically approached with combination chemotherapies in addition to local control. Fortunately, these malignancies have been approached through careful clinical trial collaboration to define risk groups and appropriately deliver local control measures and systemic therapies. Although local disease is typically approached with curative intent, therapy typically lasts over 6 months and has significant associated morbidities. It is more difficult to cure metastatic disease or induce sustained remissions. In this article, we discuss recent advances in the understanding of the disease process and highlight recent and future cooperative group trials in osteosarcoma, Ewing sarcoma, rhabdomyosarcoma, nonrhabdomyosarcoma soft tissue sarcomas, and desmoid tumor as well as discuss promising therapeutic approaches such as epigenetics and immunotherapy.
Article
Full-text available
Bacterial- and plant-derived immunotoxins have documented potential for treatment of cancer. We discuss Anthrax toxin, ribosome inactivating-toxins, such as saporin and ricin, and ADP-ribosylating toxins such as Diphtheria toxin and Pseudomonas exotoxin, with focus on the latter, which has been most thoroughly investigated. Regarding their potential as anticancer agents, critical issues such as immunogenicity and toxicity are outlined. We describe different generations of immunotoxins, the pathways for the delivery of the cytotoxic 'warheads', molecular parameters modulating efficacy, and combination therapy with other anticancer agents. Finally, we discuss deimmunization strategies based on the removal of B- and T-cell epitopes from the cytotoxic component, and highlight promising clinical proof-of-concept studies.
Article
Full-text available
Unconjugated monoclonal antibodies that target hematopoietic differentiation antigens have been developed to treat hematologic malignancies. Although some of these have activity against chronic lymphocytic leukemia and hairy cell leukemia, in general, monoclonal antibodies have limited efficacy as single agents in the treatment of leukemia. To increase their potency, the binding domains of monoclonal antibodies can be attached to protein toxins. Such compounds, termed immunotoxins, are delivered to the interior of leukemia cells based on antibody specificity for cell surface target antigens. Recombinant immunotoxins have been shown to be highly cytotoxic to leukemic blasts in vitro, in xenograft model systems and in early phase clinical trials in humans. These agents will likely play an increasing role in the treatment of leukemia.
Article
Severe combined immunodeficient (SCID) mice injected intravenously with a human Burkitt's lymphoma cell line (Daudi) develop disseminated lymphoma (SCID/Daudi), which is fatal in 100% of the mice. Early treatment of these mice with either an immunotoxin (IT) cocktail (consisting of anti-CD19-ricin A chain plus anti-CD22-ricin A chain) or chemotherapy significantly prolonged survival but was not curative. Combination therapy with the IT cocktail and any one of three chemotherapeutic drugs (doxorubicin, cytoxan, or camptothecin) cured the mice. Cure was demonstrated by both histopathologic examination of treated mice and, more importantly, by adoptive transfer of cells from organs of the cured mice to naive SCID mice where 100 tumor cells would have caused disease in the recipients. These results provide a strong rationale for combining IT therapy with conventional chemotherapy in the treatment of B-cell neoplasia.
Article
Overexpression of the ErbB2 and epidermal growth factor receptor (EGFR) tyrosine kinases is frequently observed in squamous cell carcinomas of the head and neck, and has been correlated with shorter overall survival. By immunoblot analysis, we have found EGFR and ErbB2 expression in 6 out of 6 established head and neck cancer cell lines. Elevated EGFR protein levels were noted in 3 and elevated ErbB2 levels in 5 of them. Significant expression of EGFR and ErbB2 was also detected in 17 of 47 and 26 of 45 primary tumor samples. Due to their enhanced expression on the tumor cell surface, these receptors can be regarded as suitable targets for directed cancer therapy. We have analyzed the antitumoral activity of recombinant single-chain antibody toxins specific for ErbB2 and EGFR against head and neck cancer cells in vitro and in vivo. The recombinant toxins consist of the variable domains of the heavy and light chains of monoclonal antibodies (MAbs) genetically fused to a truncated Pseudomonas exotoxin A (ETA). At low concentrations, the ErbB2-specific single-chain antibody (scFv) toxin scFv(FRP5)-ETA and the EGFR-specific toxins scFv(225)-ETA and scFv(14E1)-ETA inhibited the in vitro growth of established head and neck cancer cell lines and primary tumor cells. In a nude mouse tumor model, intratumoral injection of the antibody toxins resulted in the rapid regression of subcutaneously growing CAL 27 tumor xenografts, with scFv(FRP5)-ETA and scFv(14E1)-ETA treatment being most effective and leading to the cure of up to 50% of the animals. Our results suggest that EGFR and ErbB2-specific antibody toxins may become valuable therapeutic reagents for the treatment of squamous cell carcinomas of the head and neck. Int. J. Cancer 86:269–275, 2000. © 2000 Wiley-Liss, Inc.
Article
Immunotoxins are composed of a protein toxin connected to a binding ligand such as an antibody or growth factor. These molecules bind to surface antigens (which internalize) and kill cells by catalytic inhibition of protein synthesis within the cell cytosol. Immunotoxins have recently been tested clinically in hematologic malignancies and solid tumors and have demonstrated potent clinical efficacy in patients with malignant diseases that are refractory to surgery, radiation therapy and chemotherapy - the traditional modalities of cancer treatment. This therapy is thus evolving into a separate modality of cancer treatment, capable of rationally targeting cells on the basis of surface markers. Efforts are underway to obviate impediments to clinical efficacy, including immunogenicity and toxicity to normal tissues. Immunotoxins are now being developed to new antigens for the treatment of cancer.