ArticlePDF Available

Abstract and Figures

Monoclonal antibody-based therapies have made an important contribution to current treatment strategies for cancer and autoimmune disease. However, the cost for these new drugs puts a significant strain on the health-care economy, resulting in limited availability for patients. Therapeutic vaccination, defined as induction of immunity against a disease-related self-molecule, is therefore an attractive alternative. To analyze the potential of such an approach, we have developed a vaccine against the extra domain-B (ED-B) of fibronectin. This 91-aa domain, inserted by alternative splicing, is expressed during vasculogenesis in the embryo, but essentially undetectable under normal conditions in the adult. However, ED-B is highly expressed around angiogenic vasculature, such as in tumorigenesis. Here, we demonstrate that it is possible to break self-tolerance and induce a strong antibody response against ED-B by vaccination. Nineteen of 20 vaccinated mice responded with production of anti-ED-B antibodies and displayed a 70% reduction in tumor size compared to those lacking anti-ED-B antibodies. Analysis of the tumor tissue revealed that immunization against ED-B induced several changes, consistent with an attack by the immune system. These data show that tumor vascular antigens are highly interesting candidates for development of therapeutic vaccines targeting solid tumors.
Content may be subject to copyright.
The FASEB Journal Research Communication
Vaccination against the extra domain-B of fibronectin
as a novel tumor therapy
Elisabeth J. M. Huijbers,* Maria Ringvall,* Julia Femel,* Sebastian Kalamajski,*
,1
Agneta Lukinius,
Magnus Åbrink,* Lars Hellman,
and Anna-Karin Olsson*
,2
*Department of Medical Biochemistry and Microbiology and
Department of Cell and Molecular
Biology, Biomedical Center, and
Department of Genetics and Pathology, Rudbeck Laboratory,
Uppsala University, Uppsala, Sweden
ABSTRACT Monoclonal antibody-based therapies have
made an important contribution to current treatment strate-
gies for cancer and autoimmune disease. However, the cost
for these new drugs puts a significant strain on the health-
care economy, resulting in limited availability for patients.
Therapeutic vaccination, defined as induction of immu-
nity against a disease-related self-molecule, is therefore
an attractive alternative. To analyze the potential of
such an approach, we have developed a vaccine against
the extra domain-B (ED-B) of fibronectin. This 91-aa
domain, inserted by alternative splicing, is expressed
during vasculogenesis in the embryo, but essentially
undetectable under normal conditions in the adult.
However, ED-B is highly expressed around angiogenic
vasculature, such as in tumorigenesis. Here, we demon-
strate that it is possible to break self-tolerance and
induce a strong antibody response against ED-B by
vaccination. Nineteen of 20 vaccinated mice responded
with production of anti-ED-B antibodies and displayed
a 70% reduction in tumor size compared to those
lacking anti-ED-B antibodies. Analysis of the tumor
tissue revealed that immunization against ED-B induced
several changes, consistent with an attack by the im-
mune system. These data show that tumor vascular
antigens are highly interesting candidates for develop-
ment of therapeutic vaccines targeting solid tumors.—
Huijbers, E. J. M., Ringvall, M., Femel, J., Kalamajski,
S., Lukinius, A., Åbrink, M., Hellman, L., Olsson, A.-K.
Vaccination against the extra domain-B of fibronectin
as a novel tumor therapy. FASEB J. 24, 4535–4544
(2010). www.fasebj.org
Key Words: therapeutic immunization neovascularization
extracellular matrix angiogenesis
Vaccination programs have served to virtually erad-
icate disabling and life-threatening diseases such as
polio, diphtheria, and smallpox. The possibility of
using vaccination as a treatment strategy also for can-
cer, allergy, and other inflammatory conditions, has for
the past years been the focus of intense research.
However, no vaccination that targets a self-molecule is
yet in clinical use. There are several reasons why the
development of vaccines for noninfectious disease is
more difficult. First, the antigen to be targeted is a
self-molecule. In contrast to vaccination against foreign
substances, such as virus or bacterial antigens, there is a
requirement to break self-tolerance of the immune
system toward the self-antigen (1). Vaccination against
tumor cell antigens is further complicated by the fact
that tumor cells have developed mechanisms to evade
recognition by the immune system, for example, by
decreased presentation of endogenous peptides on
major histocompatibility complex (MHC) (2). We have
carefully addressed these issues with the aim of increas-
ing the chances of clinical development of a cancer
vaccine. The immunization technique that we use is
highly efficient in breaking self-tolerance compared to
traditional methods, since it does not depend on MHC
presentation of the self-antigen (3, 4). To circumvent
the immune escape by the tumor cells, we have focused
on a tumor vascular antigen as a vaccine target, instead
of a tumor cell antigen. Tumor vessels are significantly
different from normal vessels, which can be exploited
for development of new cancer therapies (5). Not only
are the tumor vessels less functional, they also express
or overexpress distinct molecules, which can be used
for targeted cancer therapies. One well-characterized
marker in this category is the extra domain-B of fi-
bronectin (ED-B). ED-B is a 91-aa domain inserted into
fibronectin by alternative splicing (6, 7) (Fig. 1A). The
ED-B-containing splice variant of fibronectin is highly
expressed by developing blood vessels during embryo-
genesis. In the adult, ED-B is absent under normal
conditions and only expressed in situations involving
neovascularization, such as wound healing and tumor
growth (8, 9). In addition, ED-B is not only produced
by the vasculature, but it can also be expressed by the
tumor cells themselves in certain situations (10). Tar-
geting of ED-B by administration of antibodies coupled
1
Present address: Department of Orthopaedic Surgery and
Genetics, Children’s Hospital and Harvard Medical School,
Boston, MA 02115, USA.
2
Correspondence: Department of Medical Biochemistry
and Microbiology, Uppsala University, Biomedical Center,
Husargatan 3, Box 582, SE-751 23 Uppsala, Sweden. E-mail:
anna-karin.olsson@imbim.uu.se
doi: 10.1096/fj.10-163022
45350892-6638/10/0024-4535 © FASEB
to radioactive or cytotoxic agents has been developed
by the research groups of Luciano Zardi and Dario
Neri, and these strategies have given promising results
in mouse models of cancer (11–14) and also in a
clinical study (15). Instead of using ED-B as a target for
the delivery of cytotoxic compounds to the tumor
tissue, we have developed a vaccine that induces pro-
duction of antibodies directed against ED-B, which
elicits an immune response against the tumor tissue
where ED-B is expressed. A major advantage of this
approach is that the manufacturing cost will only be a
fraction of that required for repeated and long-term
administration of a GMP-produced monoclonal anti-
body.
MATERIALS AND METHODS
Expression vectors for recombinant proteins
TRX-EDB
The region encoding the ED-B domain and a N-terminal
His-tag (6xhistidine) (309 bp) was inserted in frame, down-
stream the bacterial TRX sequence (354 bp), in a pET-21a
vector (Novagen; EMD Chemicals, Gibbstown, NJ, USA). The
resulting expression vector was named pET21a TRX-EDB.
GST-EDB
The TRX sequence in the pET21a TRX-EDB vector was
replaced with a PCR-amplified GST sequence (678 bp). The
resulting vector construct was named pET21a GST-EDB.
Expression and purification of recombinant fusion-proteins
The vectors pET21a TRX-EDB and pET21a GST-EDB were
transformed into Escherichia coli Rosetta gami (DE3) (Nova-
gen; EMD Chemicals) for expression of the fusion proteins.
Rosetta gami (DE3) is an E. coli strain that is optimized for
production of eukaryotic proteins. Overnight cultures were
diluted 1:10 and grown until OD
600
0.5. Protein expression
was induced with 1 mM isopropyl -D-1-thiogalactopyrano-
side (IPTG) (VWR International AB, Stockholm, Sweden).
TRX-EDB protein expression was induced at 22°C for 16 h,
and expression of GST-EDB was induced at 37°C for 4 h. The
lower temperature was used to increase the soluble fraction of
TRX-EDB.
Fusion proteins were released by sonication of the bacteria
on ice for 4 30 s (repeated 3 times). Bacterial debris was
pelleted by centrifugation at 2500 gand the supernatant was
collected. Thirty milliliters of bacterial supernatant (originat-
ing from a 1.2-L bacterial culture) was mixed with 1 ml
Ni-NTA agarose slurry (Qiagen, Hilden, Germany) and incu-
bated “end-over-end” at 4°C for 3 h. The Ni-NTA-agarose was
pelleted by centrifugation, washed 5 times with PBS pH 7.0/1
M NaCl/0.1% Tween-20, transferred to a column with a glass
filter (Sartorius AG, Go¨ttingen, Germany) and washed again.
The protein was eluted with 100 mM imidazole (Sigma-
Aldrich GmbH, Steinheim, Germany) in 20 mM Tris (pH
8.0)/0.1 M NaCl in 500-l fractions. Purification of the
GST-EDB protein was performed as above, with the exception
that the Ni-NTA-agarose column with bound GST-EDB was
treated with 50 mM imidazole before elution, to reduce
background. Protein-containing fractions were pooled and
dialyzed against PBS (pH 7.0) [Spectra/Por CE (cellulose
ester) membrane, 6- to 8-kDa M
W
cutoff; Spectrum Medical
Industries, Los Angeles, CA, USA]. Final protein concentra-
tion was estimated by comparison with a BSA fraction V
(Roche Diagnostics, Mannheim, Germany) standard on SDS-
PAGE and by a protein quantification assay (BCA Protein
Assay; Pierce, Rockford, IL, USA). Purified fractions of both
TRX-EDB and GST-EDB protein were analyzed by mass-
spectrometry to confirm their identity.
Cell culture
T241 fibrosarcoma cells (American Type Culture Collection,
Manassas, VA, USA) were cultured in DMEM Glutamax
(31966; Invitrogen AB, Lidingo¨, Sweden) supplemented with
10% FCS (ECS 0180L; Euroclone Ltd, Devon, UK).
Animal studies
Animal work was approved by the Uppsala University board of
animal experimentation (reg. no. C207/7) and thus per-
formed according to the UK Coordinating Committee on
Cancer Research (UKCCCR) guidelines for the welfare of
animals in experimental neoplasia (16). The mice were
anesthesized with isoflurane (Forene; Abbott Scandinavia,
Solna, Sweden) (induction 4.5%; maintenance 2.5–3%) dur-
ing all manipulations. Four- to 8-wk-old female C57BL/6 mice
(Taconic, Lille Skensved, Denmark), were immunized in the
groin with 100 l of an emulsion containing 100 g recom-
binant TRX-EDB protein in PBS, mixed 50:50 with Freund’s
complete adjuvant (FCA) (263810; Difco Laboratories, De-
troit, MI, USA) atd1oftheexperiment. Booster injections
containing the same amount of recombinant protein but in
Freund’s incomplete adjuvant (FIA) (F5506; Sigma-Aldrich)
were given in the opposite groin at d 14 and 28. Control
animals received PBS in FCA or FIA. Ten mice/group were
included in both experiments. At d 35, all mice were inocu-
lated subcutaneously in the left flank with 0.5 10
6
T241
fibrosarcoma cells in a total volume of 100 l in PBS. The
tumors were allowed to grow for 21 d (first experiment) or
26 d (second experiment), until the maximum allowed size
was reached. One mouse was lost in the first experiment and
2 mice in the second experiment, due to ruptured tumors. At
the last day of the experiment, blood samples were taken
from all mice before euthanization. Tumors were removed
and measured with a caliper before cryopreservation in
isopentane/dry ice. Tumor volume was calculated using the
formula width
2
length ⫻␲/6. The frozen tissue was stored
at 80°C until further processing.
ELISA
For analysis of total anti-ED-B antibody levels, blood samples
were left to coagulate overnight at 4°C and centrifuged twice
at 13,000 rpm for 5 min in a microcentrifuge. Supernatants
(serum) were collected and stored at 20°C until use. ELISA
plates (Costar MaxiSorp surface; Corning Incorporated,
Corning, NY, USA) were coated with GST-EDB (10 g/ml
protein in PBS) and blocked with centrifuged horse serum
(Life Technologies; Biocult, Glasgow, UK). Plates were incu-
bated with serum from control or TRX-EDB vaccinated mice
(diluted 1:25 in horse serum), followed by biotinylated goat
anti-mouse IgG (HL) (BA-9200; Vector Laboratories, Burl-
ington, ON, Canada) diluted 1:500 and streptavidin-horserad-
ish peroxidase (SA-HRP) (SA-5004; Vector Laboratories)
diluted 1:500. All incubations were performed at 37°C. HRP
activity was detected by incubation with TMB substrate
4536 Vol. 24 November 2010 HUIJBERS ET AL.The FASEB Journal www.fasebj.org
(T8665; Sigma-Aldrich). The absorbance was measured at
405 nm.
For analysis of the levels of anti-ED-B antibodies of differ-
ent subclasses serum from control and TRX-EDB vaccinated
mice without tumors, was analyzed by ELISA. The assay was
performed as described above with the exceptions that bio-
tinylated goat anti-mouse antibodies specific against IgG
subclasses 1, 2a, 2b, and 3 (1070-08, 1080-08, 1090-08, 1110-
08; Southern Biotech, Birmingham, AL, USA) were used as
secondary reagent (diluted 1:2500 in PBS) and that the
absorbance was read at 650 nm.
Analysis of tumor necrosis
Estimation of the necrotic area fraction (%) was performed
on tumor sections stained with Mayer’s hematoxylin and
eosin (HistoLab products AB, Go¨teborg, Sweden) and CD31,
using an ocular grid. Necrosis was defined as avascular areas
lacking nuclear structures. Four tumors of relatively equal size
from control and TRX-EDB vaccinated mice from experi-
ment 1 and 2, respectively (in total n8/group), were
selected for the analysis, which was performed in a masked
fashion. Pictures of necrotic areas were taken in a Nikon
Eclipse 90i microscope with NIS Elements 3.06 software
(Nikon Instruments B.V. Europe, Amstelveen, The Nether-
lands), with the 40 objective.
IHC of glioma tissue using serum from control and
TRX-EDB immunized mice
Cryosections of normal brain tissue and grade III glioma from
Gtv-a Arf
/
mice (17) were used for IHC with serum from
control or TRX-EDB immunized mice. The transgenic Gtv-a
Arf
/
glioma model expresses the avian leukemia virus
receptor tv-a in glial progenitor cells. Injection of the avian
virus RCAS, engineered to express platelet-derived growth
factor B (PDGFB), in the brain of newborn Gtv-a mice
induces development of glioma, with a similar histology to
humans. The tissue used for the current IHC analysis was
derived from RCAS-PDGFB virus injected Gtv-a mice that
were also deficient for the tumor suppressor gene Arf. The
sections were blocked with FCS, incubated with mouse-serum
(diluted 1:10) from control or TRX-EDB-immunized animals,
and treated with 1% hydrogen peroxide (Merck KGaA,
Darmstadt, Germany). Detection of primary antibody was
performed with biotinylated anti-mouse IgG (BA-9200; Vector
Laboratories) diluted 1:500, followed by incubation with
SA-HRP diluted 1:500 and AEC substrate (SK-4200; Vector
Laboratories). Counterstaining was performed with Mayer’s
hematoxylin (HistoLab). For negative controls, the primary
antibody was omitted.
Electron microscopy
T241 fibrosarcoma tumor tissue from 4 control- and 4 TRX-
EDB-immunized mice were fixed in 0.15 M sodium cacody-
late-buffered 2.5% glutaraldehyde and postfixed in 0.1 M
s-collidine-buffered 2% osmium tetroxide, dehydrated, and
embedded in epoxy resin. Ultrathin sections were analyzed by
electron microscope (Philips CM-10; FEJ; Philips AB, Stock-
holm, Sweden). Approximately 15–20 vessels from each indi-
vidual were analyzed.
Fibrinogen staining and quantification
T241 fibrosarcoma cryosections from control- and TRX-EDB-
immunized mice (n4/group) were blocked in 3% BSA for
1 h at room temperature (RT) and double stained overnight
at 4°C with purified rat anti-mouse CD31 (PECAM-1) mono-
clonal antibody (cat. no. 553370; BD Pharmingen, BD Bio-
sciences, Stockholm, Sweden) diluted 1:1000 and polyclonal
rabbit anti-fibrinogen (cat. no. A0080; DakoCytomation,
Glostrup, Denmark) diluted 1:500 in blocking solution. De-
tection antibodies used were Alexa555 donkey anti-rabbit
(cat. no. A31572; Invitrogen) and Alexa488 rabbit anti-rat IgG
(HL) (cat. no. A21210; Invitrogen), at a concentration of 2
g/ml in blocking solution. Incubation time was 45 min at
RT. Nuclei were visualized with Hoechst 33342 (VWR Inter-
national) at 1 g/ml. Sections were washed, mounted with
Fluoromount-G (Southern Biotech), and stored at 4°C until
use. For negative controls, the primary antibody was omitted.
For quantification of the fibrinogen-stained area, three pic-
tures of each tumor section (n12 areas) were taken at
random in a Nikon Eclipse 90i microscope with NIS Elements
3.06 software, with the 20 objective and exposure time 400
ms. Analysis of the pictures was performed with Image J 1.42
software (National Institutes of Health, Bethesda, MD, USA).
Neutrophil staining and quantification
T241 fibrosarcoma cryosections from control- and TRX-EDB-
immunized mice (n4/group) were blocked with FCS (ECS
0180L; Euroclone, Devon, UK) for1hatRTandstained with
purified rat anti-mouse Ly6G and Ly-6C (Gr-1) monoclonal
antibody (cat. no. 553123; BD Pharmingen, BD Biosciences,
Stockholm, Sweden) at a concentration of 0.5 g/ml in
blocking solution overnight at 4°C. Detection of primary
antibody binding sites was done with an Alexa555 goat anti-rat
IgG (HL) (Invitrogen) at a concentration of 2 g/ml in
blocking solution for 45 min at RT. Nuclei were visualized
with Hoechst 33342 (VWR International) at 1 g/ml. Sec-
tions were washed, mounted with Fluoromount-G (Southern
Biotech), and stored at 4°C until use. For negative controls,
the primary antibody was omitted. For quantification of the
Gr1-stained area, 3 pictures of each tumor section (n12
areas) were taken at random in a Nikon Eclipse 90i micro-
scope with NIS Elements 3.06 software, with the 20 objective
and exposure time 300 ms. Analysis of the pictures was
performed with the Image J 1.42 software.
Wound-healing assay
Animal work was approved by the Uppsala University board of
animal experimentation (reg. no. C63/9). Immunocompe-
tent C57BL/6 mice were immunized as described previously.
One week after the second boost, mice were anesthesized with
isoflurane (Forene; Abbott Scandinavia) (induction 4.5%;
maintenance 2.5–3%) and a full-thickness wound, 8 mm in
diameter, was placed on the back using a disposable biopsy
punch (Pfm Medical, Carlsbad, CA, USA). Buprenorphine sc
(Temgesic; Schering-Plough AB, Stockholm, Sweden) 0.05–
0.1 mg/kg was given 2/dforthefirst2daspain medica-
tion. In addition, Bimotrim (trimethoprim: sulfadoxine 1:5;
CEVA Vetpharma, Lund, Sweden) 0.2% (w/v) was adminis-
tered to the drinking water during the first 5 d of the
experiment. Wounds were protected with Cavilon no-sting
barrier spray (3M, Sollentuna, Sweden). Mice were observed
every day for the first7doftheexperiment. Pictures of the
wounds were taken on d 0, 7, and 14 after wounding with a
Nikon D70 camera. At d 20, the experiment was terminated,
and all animals were sacrificed, and blood was drawn for
analysis of anti-ED-B antibody titers with ELISA.
Arthritis analysis
Control- and EDB-immunized mice were examined for signs
of arthritis, both at the macroscopic and the cellular level by
4537TARGETING TUMOR VESSELS BY THERAPEUTIC VACCINATION
a person with experience from mouse models of rheumatoid
arthritis.
Macroscopic analysis
The examined mice (n6/group) had anti-ED-B antibodies
in their circulation for 8 mo. Signs of arthritis were analyzed
as described previously (18) in a masked fashion.
Cellular analysis
Control- and TRX-EDB-immunized mice were sacrificed 2
mo after immunization; front and hind paws were incubated
in 10% buffered formalin solution, extensively washed in tap
water, and transferred to Parengy solution (BIE & Berntsen
A/S, Roedovre, Denmark) for decalcification. The solution
was exchanged to fresh Parengy solution after 30 h, and the
tissue was further incubated for 4 d, washed in tap water for
24 h, dehydrated, and paraffin embedded. The paws were
sectioned sagitally, and stained with hematoxylin and eosin.
Sections of 3 animals in each group were examined in a
masked analysis for histopathological changes, such as cellu-
lar infiltration and cartilage destruction.
Statistical analysis
For statistical analysis a Mann-Whitney Utest was performed,
and values of P0.05 were considered significant. The
Mann-Whitney Utest was used because of the small sample
size (n10) and because normal distribution could not be
assumed.
RESULTS
The basis of the vaccination technique is to produce
the self-antigen of choice as a fusion protein with a
foreign (i.e., nonself) part, derived, for example,
from bacteria (1). The mechanism by which autore-
active B cells are activated to become antibody-
producing plasma cells is described in Fig. 1B. The
polyclonal anti-self antibodies bind to the target pro-
tein, and the resulting immune complexes will be
cleared by phagocytosis if the target protein is soluble.
When the antigen is membrane- or matrix-bound (as in
the case of ED-B), “frustrated phagocytosis” will com-
mence, since the immune complexes cannot be phago-
cytosed. This results in recruitment of monocytes/
macrophages, neutrophils, and natural killer cells,
which leads to tissue damage where the antigen is
expressed.
Two essentially pure and soluble recombinant fusion
proteins containing ED-B as the self-antigen part were
produced by expression in bacteria (see Fig. 1Cfor a
schematic illustration of the expression vector). ED-B
was either fused to the E. coli-derived protein thiore-
doxin (TRX) or glutathione-S-transferase (GST; de-
rived from the parasite worm Schistozoma japonicum),
generating the fusion proteins TRX-EDB and GST-EDB
(Fig. 1D). C57BL/6 mice were immunized with TRX-
EDB or vehicle in Freund’s adjuvant. After 5 wk,
including 2 booster immunizations, all mice received
subcutaneous injections of T241 fibrosarcoma cells.
Before the experiment was initiated, expression of
ED-B in this particular tumor type was confirmed using
a synthetic anti-ED-B antibody, kindly provided by Prof.
Dario Neri (ETH Zurich, Zurich, Switzerland; data not
shown). After 21 d, blood was collected from all mice,
tumors were removed, measured with a caliper and
cryopreserved. The tumor volume in TRX-EDB vacci-
Figure 1. Mechanism for breaking self-tolerance (adapted from ref. 1). A) Illustration of a fibronectin monomer with the
alternatively spliced ED-B indicated. B)1) Antigen presenting cells (APC) internalize the fusion protein (TRX-EDB) and
present nonself (TRX) and self (ED-B) peptides via MHC class II. 2) Nonself peptides are recognized by the T-cell receptor
(TCR) on T-helper cells (TH) that become activated. Self-peptides are not recognized, since autoreactive T cells are deleted in
the thymus during development. 3) Autoreactive B cells are, however, present in the circulation and recognize the fusion
protein’s self-part (ED-B) via their B-cell receptor (BCR). These autoreactive B-cells take up the fusion protein and present
peptides from both the self- and the nonself part on MHC class II. TH cells previously activated by the foreign part of the fusion
protein now activate these autoreactive B cells, since they present the same foreign peptides. 4) Autoreactive B-cells undergo
clonal expansion and produce anti-ED-B antibodies. C) Schematic illustration of the pET21a bacterial expression vector
encoding TRX-EDB under the control of the IPTG-inducible T7lac promoter. His-tag is included for the purpose of purification.
D) Schematic representation of the fusion proteins TRX-EDB (20 kDa) and GST-EDB (33 kDa) and their appearance on
reducing SDS-PAGE after purification.
4538 Vol. 24 November 2010 HUIJBERS ET AL.The FASEB Journal www.fasebj.org
nated mice was significantly reduced compared to the
control group (Fig. 2A; 1st experiment). An attenuated
tumor growth in TRX-EDB vaccinated mice was con-
firmed in a second independent experiment (Fig. 2A;
2nd experiment). Antibodies against ED-B were present
in the TRX-EDB-immunized mice in both experi-
ments (Fig. 3A). Interestingly, the only mouse that
for unknown reasons did not generate any significant
amount of anti-ED-B antibodies above background
(Fig. 3A, 2nd experiment; pound sign) developed the
largest tumor in this group (Fig. 2A, 2nd experiment;
pound sign). The average reduction of tumor volume
in mice immunized against ED-B, compared to those
lacking anti-ED-B antibodies, was 68.5% (1st experi-
ment, 71%; 2nd experiment, 66%; Fig. 2B). Histolog-
ical analysis of hematoxylin-and-eosin-stained tumor
tissue from both experiments showed larger necrotic
areas in the TRX-EDB group compared to controls
(Fig. 2C). The pictures in Fig. 2Dare examples of a
viable and a necrotic area of the tumor tissue.
To analyze the antibody response against ED-B in
more detail, we determined which IgG subclasses that
Figure 2. Reduced tumor size in TRX-EDB-vaccinated mice.
A) TRX-EDB (EDB)-vaccinated mice displayed a significantly
reduced tumor size compared to the control group (Ctrl) in 2
independent experiments: 1st experiment, P0.0007; 2nd
experiment, P0.0188. Each dot represents the tumor
volume of 1 individual. One animal with no or very low
amounts of anti-ED-B antibodies is denoted by pound sign (#;
see Fig. 3A, 2nd experiment). B) Mice from the 1st and 2nd
experiment grouped according to the presence of anti-ED-B
antibodies (EDB ab
and EDB ab
;P0.0001). C) Necrotic
area fraction (%) in 8 tumors from control and TRX-EDB-vaccinated mice, respectively, from the 1st and 2nd experiment
(P0.0148). D) Photos illustrating a viable (left) and a necrotic (right) tumor area. Viable tissue has clearly distinguishable
nuclei and is vascularized, as illustrated by the CD31-positive blood vessel (red). Necrotic tissue contains no clear structures.
Scale bar 50 m.
Figure 3. Functional anti-ED-B antibodies are produced in TRX-EDB
vaccinated mice. A) Serum from control (Ctrl) and TRX-EDB (EDB)-
vaccinated mice was analyzed for the presence of anti-ED-B antibodies
using ELISA. Each bar represents an individual animal. Animal lacking
anti-ED-B antibodies is indicated by pound sign (#). B) Serum from
control and TRX-EDB-vaccinated mice without tumors was analyzed for
anti-ED-B IgG subclasses; Ig1, IgG2a, IgG2b, and IgG3. C) Immunohis-
tochemical staining of normal brain tissue (top panels) and grade III
experimental glioma from mouse (bottom panels) using serum from
control (left panels) and TRX-EDB (right panels)-immunized animals.
Scale bar 20 m.
4539TARGETING TUMOR VESSELS BY THERAPEUTIC VACCINATION
were generated in immunized mice (Fig. 3B). As ex-
pected, the major subclass was found to be IgG1,
characteristic of a TH2-response, meaning an antibody-
mediated immune response.
To confirm that the anti-ED-B antibodies in immu-
nized mice were functional and recognized native ED-B
in tissue, we performed immunohistochemical staining
of murine grade III glioma tissue (17), using serum
from control or TRX-EDB vaccinated mice. Gliomas are
known to express high levels of ED-B around the tumor
vasculature (19), in contrast to normal brain tissue,
where ED-B is undetectable. As expected, no staining
was detected when normal brain tissue was incubated
with serum from control or TRX-EDB vaccinated mice
(Fig. 3C, top panels). However, when glioma tissue was
incubated with serum from TRX-EDB vaccinated mice,
a typical ED-B expression pattern was visualized along
the vasculature, which was not seen with serum from
the control animals (Fig. 3C, bottom panels). This
clearly demonstrates that functional anti-ED-B antibod-
ies, recognizing native ED-B in tissue, are produced in
mice immunized with TRX-EDB.
Stereological analysis of tumor vascularization (20)
using CD31 as a marker, showed no obvious differences
between the control and ED-B-vaccinated group (data
not shown). However, electron microscope analysis of
the tumor tissue revealed an altered morphology of the
vasculature in TRX-EDB immunized mice (Fig. 4). The
vessel wall displayed invaginations and protrusions
(Fig. 4B2, arrow) not seen in the control group (Fig.
4A1, arrow). Furthermore, transport vesicles present
in endothelium from controls (Fig. 4A1, arrowheads)
were less frequent in TRX-EDB immunized mice.
Instead, a large number of free ribosomes was seen in
tumor endothelial cells from mice carrying anti-ED-B
antibodies, indicating an acute cellular response
(Fig. 4B3, arrowheads). In addition, macrophages
actively trying to phagocyte the endothelium were
seen in tumors from mice vaccinated against ED-B.
These macrophages displayed a dilated endoplasmic
reticulum (Fig. 4B4, asterisk), indicating production
of proteolytic enzymes for lysosomal degradation.
Collectively, these observations are consistent with an
immunological response against the tumor blood
vessels in TRX-EDB-immunized mice and indicate a
decreased functionality. To address vessel function,
Figure 4. Altered vessel morphology and fibrinogen extravasation in tumors from TRX-EDB-vaccinated mice. Representative electron
micrographs of blood vessels from ultrathin sections of tumors from control (Ctrl; A) and TRX-EDB (EDB; B) vaccinated mice. Panels
A1 and B2B4 are magnifications of the corresponding area in Aand B.A1) Arrowheads indicate transport vesicles; asterisk indicates
pericyte. B2) Arrow indicates invaginations and protrusions of the vessel wall, not seen in the control group (compare with A1; arrow).
B3) Arrowhead indicates free ribosomes. B4) Asterisk indicates dilated endoplasmic reticulum. C) Top panels: representative
immunohistochemical stainings for CD31 (green) and fibrinogen (red) in tumor tissue from control and TRX-EDB-vaccinated
animals. Bottom panel: tumors from animals with anti-ED-B antibodies showed a significantly increased area positive for fibrinogen
staining (P0.0351). Data are derived from n4 tumors/group; each dot represents 1 quantified field, 3 fields/tumor. Similar
symbols are used to denote individual tumors in the 2 groups. Scale bars 2m(A,B); 200 m(C).
4540 Vol. 24 November 2010 HUIJBERS ET AL.The FASEB Journal www.fasebj.org
we performed immunohistochemical staining for fi-
brinogen in tumor tissue derived from control- or
EDB-immunized mice, as a measurement of vascular
leakage. The stainings revealed significantly in-
creased amounts of extravasated fibrinogen in the
tumor tissue derived from mice with anti-ED-B anti-
bodies (Fig. 4C).
To address whether an ongoing immune response in
the tumor tissue could be detected at a cellular level, we
performed immunohistochemical staining for neutro-
phils using the marker Gr1. The area with Gr1-positive
staining was quantified using an image analysis program.
A significantly increased amount of infiltrating neutro-
phils was detected in TRX-EDB-immunized mice (Fig. 5).
As previously mentioned, ED-B is expressed during
neovascularization in wound healing. From a safety
perspective, it is, therefore, important to address
whether wound healing is affected by the presence of
anti-ED-B antibodies. A wound-healing assay was, there-
fore, performed in control- and EDB-immunized mice.
A full-thickness wound of 1 cm in diameter was
placed on the back of the mice using a biopsy punch.
We followed the mice closely during the healing period
but could not detect any differences between the
groups. After 14 d, the wounds of all mice were
completely healed (Fig. 6A). As can be seen in Fig. 6B,
all TRX-EDB-immunized mice in the wound healing
assay carried anti-ED-B antibodies.
In addition to situations with neovascularization,
there are also some reports on ED-B expression in
cartilage (21, 22). One concern was, therefore, whether
TRX-EDB immunized mice could develop arthritis. We
have carefully analyzed the TRX-EDB immunized mice,
both at a macroscopic and cellular level, as described in
Materials and Methods, but failed to detect any signs of
arthritis (data not shown).
DISCUSSION
Vaccination against ED-B induced high levels of anti-
ED-B antibodies, which was previously believed to be
very difficult, or even impossible, due to the high grade
of conservation of this protein domain (23, 24). The
amino acid sequence of ED-B is, in fact, identical in
mouse, rat, rabbit dog, monkey, and human (5). Our
Figure 5. Increased neutrophil infiltration in tumors from TRX-EDB-immunized mice. Left and center panels: representative
immunohistochemical stainings for neutrophils (red), using the marker Gr1, in tumor tissue from control (Ctrl) and TRX-EDB
(EDB)-vaccinated animals. Right panel: Gr1-positive area (%) was significantly increased in tumors from TRX-EDB-vaccinated
mice (P0.0061). Data are derived from n4 tumors/group; each dot represents 1 quantified field, 3 fields/tumor. Similar
symbols are used to denote individual tumors in the two groups. Scale bars 200 m.
Figure 6. No impairment of wound healing in TRX-EDB-
vaccinated mice. A) Full-thickness wounds of control (Ctrl) and
TRX-EDB (EDB) vaccinated animals (n5/group) are depicted
at d 0, 7, and 14 after wounding. No difference in wound healing
between the two groups was observed. B) Anti-ED-B antibodies
were present in serum of all 5 TRX-EDB-immunized mice
included in the wound healing assay, but not in the control
mice. Bars represent individual animals.
4541TARGETING TUMOR VESSELS BY THERAPEUTIC VACCINATION
data now show that immunization against ED-B is,
indeed, possible and therefore also isolation of monoclo-
nal anti-ED-B antibodies using hybridoma technology.
The anti-ED-B antibodies generated by the vaccine
had a protective effect against tumor growth; 70%
reduction in tumor volume was achieved by vaccination
against this single tumor vascular antigen. Targeting a
combination of 2 or more such antigens may prove
even more effective with respect to suppression of
tumor growth. In this context, it is important to remem-
ber that the antibodies that are generated by immuni-
zation are polyclonal and therefore significantly more
efficient in immune complex formation and activation
of the immune system than a monoclonal antibody.
Electron microscope analysis, in combination with im-
munohistochemical staining for fibrinogen, revealed
an aberrant morphology and increased leakage of
tumor vessels in TRX-EDB immunized mice compared
to controls. Moreover, an increased infiltration of neu-
trophils in tumors from mice with anti-ED-B antibodies
support the conclusion of an ED-B targeted attack by
the immune system.
An important safety factor is that this type of vacci-
nation is reversible and not life-long, in contrast to
immunization against certain foreign antigens (4, 25,
26). By “reversible” we mean 2 things: first, the antibody
titers decrease to background levels at a faster rate
when you immunize against a self-molecule than is the
case for immunization against a foreign antigen; and
second, a new injection of the fusion protein TRX-EDB
is required to increase the antibody titers again. Using
the same therapeutic vaccination technique as in the
present study, others (4, 25) demonstrated the revers-
ibility of the immune response against IgE and IL-13.
After 3–4 mo, the majority of the therapeutic antibod-
ies were gone and after 11 mo, the antibody titers
were reduced by 89–99% (4). Ongoing studies in our
laboratory indicate that the same kinetics apply to
antibody production against ED-B (unpublished re-
sults). The main reason for this observation is that the
antigen is a self-molecule. When an autoreactive B cell
meets its antigen (for instance, ED-B) in the absence of
T-cell help (which is provided by the TRX-part of the
fusion protein TRX-EDB in the vaccine), the result is
inactivation of the B cell (anergy). This mechanism is
part of the peripheral tolerance of the immune system,
which is aimed to reduce the risk of autoimmune
responses. If instead a B cell has specificity for a nonself
antigen, the result will be activation of the B cell on
antigen encounter, since at the same time, it will
receive help from T cells directed against the same
nonself molecule. Even though repeated immuniza-
tions with the fusion protein may be required, the
reversibility is an advantage in case adverse effects are
observed. A relevant comparison is the time it takes to
clear an injected monoclonal antibody (MAb) from the
circulation. MAbs have a half-life of 3 wk, which
means that it takes 3 mo to clear 90% of the injected
antibody.
Using the immunization strategy with fusion proteins
presented in this work, we consider the risk for auto-
immune disease as very low for several reasons. It is
generally very difficult to break self-tolerance. Although
our approach works well for this purpose, 2 require-
ments have to be fulfilled at the same time: the pres-
ence of the fusion protein TRX-EDB, and concomitant
administration of an adjuvant. The presence of TRX-
EDB alone, without additional immunostimulatory
compounds, will not induce production of anti-ED-B
antibodies. Moreover, as described above, the presence
of endogenous ED-B will suppress, not enhance, activa-
tion of autoreactive B cells with specificity for ED-B.
Another safety issue that has been raised is whether
the described immunization strategy against ED-B
could potentially induce immune complex glomerulo-
nephritis. Immune complexes will, however, only de-
posit in the kidney if the targeted antigen is soluble. In
our case, the antigen is matrix bound. Moreover,
vaccination induces a slow increase in antibody titers,
and macrophages, therefore, have the capacity to clear
immune complexes as they form, even for a soluble
antigen. This is in sharp contrast to injection of hun-
dreds of milligrams of a monoclonal antibody at one
single occasion, which is usually the case when treating
patients with monoclonal antibodies.
In a recent report by Haller et al. (27), the re-
searchers clearly show that immunization against the
endothelial tip cell protein delta-like ligand 4
(DLL4) delays growth of mammary carcinomas in
mouse. In this study, a DNA vaccine approach was
used where the foreign epitopes were provided by
human DLL4. However, there are additional safety
issues to consider when targeting a self-molecule with
a DNA vaccine. There is always a risk for integration
of the vaccine DNA into the host genome. As long as
a foreign antigen, like a virus, is the target, the risk of
host integration may be acceptable from a clinical
perspective. But when it comes to self-molecules, it is
crucial that the immune response is reversible, espe-
cially if the expression pattern of the target antigen is
not completely known.
There are also previous studies reporting therapeutic
effects on tumor growth in mice following vaccination
against angiogenesis-related molecules, such as VEGF
and VEGFR2. However, the chances of clinical approval
of a vaccine against VEGFR2, present on all blood
vessels in the body, are minor. The important distinc-
tion between those vaccines and the ED-B vaccine we
present here is that immunological targeting of ED-B is
clinically feasible due to its restricted expression pat-
tern. The first clinical study targeting ED-B was recently
reported (15). In this study, Hodgkin lymphoma pa-
tients received an anti-ED-B antibody conjugated to
131
I
(
131
I-L19SIP). The treatment induced a sustained par-
tial remission in two out of three relapsed patients (15),
indicating that targeting of ED-B is interesting for
future development as a cancer treatment. This conclu-
sion is also supported by the well-documented tumor-
specific expression pattern of ED-B in a large number
of distinct tumor types, such as head and neck tumors
4542 Vol. 24 November 2010 HUIJBERS ET AL.The FASEB Journal www.fasebj.org
(28), high-grade astrocytomas (24), colorectal carcino-
mas (29), breast cancer (30), as well as lung and gastric
carcinoma and malignant melanoma (7).
The effect of the EDB vaccine on established tumors
is highly relevant and remains to be addressed. There
is, however, a major obstacle connected to this ap-
proach and that is that we need a slow-growing tumor
model in an immunocompetent mouse strain for this
purpose. The majority of subcutaneous tumor models
in immunocompetent mice are fast growing. From
when the tumor has become established and is palpa-
ble, it may take 2 to 3 wk or less until the mouse has to
be euthanized, either due to its health condition or
because the tumor has reached the maximum allowed
size according to ethical regulations. Antibody produc-
tion is not instantaneous after immunization, but re-
quires a few weeks (and repeated injection) to develop.
It is, therefore, not possible to perform this type of
experiment in the fast growing subcutaneous models
available. There are plenty of slow-growing tumor mod-
els in immunocompromised mice (SCID, Nude), but
for obvious reasons these mouse strains cannot be used
when studying effects of vaccines that require both B
and T cells. The situation in humans is of course very
different, where a tumor can develop over years.
We could not detect any adverse effects on wound
healing in mice carrying anti-ED-B antibodies, despite
expression of ED-B in this situation. The reason for this
is not known, but one possible explanation could be
that the expression of ED-B in a wound is transient
compared to the situation in tumors. Another possibil-
ity is that the quality of the vessels is different in tumors
and during wound healing (pathological vs. physiolog-
ical angiogenesis). Tumor vessels are constantly leaky,
while increased leakiness is a transient phenomenon in
vessels undergoing physiological angiogenesis. The an-
tibodies need to cross the endothelium to reach ED-B
in the extracellular matrix and targeting of ED-B may,
therefore, be more efficient in the tumor.
As mentioned previously, expression of ED-B in carti-
lage has also been reported. Unlike other connective
tissue, cartilage is not vascularized. The source of the
fibronectin splice variants is the chondrocytes themselves.
One possible explanation for the finding that cartilage
does not seem to be attacked by the immune system in
TRX-EDB-immunized mice, is that the lack of blood
vessels in cartilage may reduce the accessibility of the
anti-ED-B antibodies for their target.
Breaking self-tolerance is a central issue in the field
of therapeutic vaccines. To transfer the ED-B vaccine to
the clinic, it must be based on a less toxic, but equally
potent, adjuvant than Freund’s. On the basis of a large
screen, we have recently identified alternative adju-
vants, which are as efficient as Freund’s in evoking a
self-antigen response, but are biodegradable and non-
toxic (31). This finding will significantly aid clinical
development of therapeutic vaccines.
In summary, our data show that therapeutic vaccines
targeting tumor vascular antigens, single molecules or
combinations, are highly interesting for clinical devel-
opment and could provide potent and cost-efficient
new treatment strategies for cancer.
The authors thank Dr. Lene Uhrbom (Department of
Genetics and Pathology, Uppsala University) for providing
tissue sections of normal mouse brain and glioma and Prof.
Dario Neri (Institute of Pharmaceutical Sciences, ETH Zu-
rich, Zurich, Switzerland) for providing SIP anti-ED-B anti-
bodies. The authors also thank Prof. Wilhelm Jahnen-Dech-
ent and Dr. Daniela Dreymu¨ller (Institute for Biomedical
Engineering, RWTH Aachen, Aachen, Germany) for gener-
ously teaching us the wound-healing model. Financial sup-
port was provided by The Swedish Research Council, The
Swedish Cancer Society, The Swedish Society of Medicine,
Jeanssons Foundation, and the Åke Wiberg Foundation.
REFERENCES
1. Hellman, L. (2008) Therapeutic vaccines against IgE-mediated
allergies. Expert Rev. Vaccines 7, 193–208
2. Ridolfi, L., Petrini, M., Fiammenghi, L., Riccobon, A., and
Ridolfi, R. (2009) Human embryo immune escape mechanisms
rediscovered by the tumor. Immunobiology 214, 61–76
3. Hellman, L. (1994) Profound reduction in allergen sensitivity
following treatment with a novel allergy vaccine. Eur. J. Immunol.
24, 415–420
4. Vernersson, M., Ledin, A., Johansson, J., and Hellman, L.
(2002) Generation of therapeutic antibody responses against
IgE through vaccination. FASEB J. 16, 875–877
5. Neri, D., and Bicknell, R. (2005) Tumour vascular targeting.
Nat. Rev. Cancer 5, 436–446
6. Zardi, L., Carnemolla, B., Siri, A., Petersen, T. E., Paolella, G.,
Sebastio, G., and Baralle, F. E. (1987) Transformed human cells
produce a new fibronectin isoform by preferential alternative
splicing of a previously unobserved exon. EMBO J. 6, 2337–2342
7. Carnemolla, B., Balza, E., Siri, A., Zardi, L., Nicotra, M. R.,
Bigotti, A., and Natali, P. G. (1989) A tumor-associated fibronec-
tin isoform generated by alternative splicing of messenger RNA
precursors. J. Cell Biol. 108, 1139–1148
8. Castellani, P., Viale, G., Dorcaratto, A., Nicolo, G., Kaczmarek,
J., Querze, G., and Zardi, L. (1994) The fibronectin isoform
containing the ED-B oncofetal domain: a marker of angiogen-
esis. Int. J. Cancer 59, 612–618
9. Astrof, S., and Hynes, R. O. (2009) Fibronectins in vascular
morphogenesis. Angiogenesis 12, 165–175
10. Midulla, M., Verma, R., Pignatelli, M., Ritter, M. A., Courtenay-
Luck, N. S., and George, A. J. (2000) Source of oncofetal
ED-B-containing fibronectin: implications of production by
both tumor and endothelial cells. Cancer Res. 60, 164–169
11. Tarli, L., Balza, E., Viti, F., Borsi, L., Castellani, P., Berndorff, D.,
Dinkelborg, L., Neri, D., and Zardi, L. (1999) A high-affinity
human antibody that targets tumoral blood vessels. Blood 94,
192–198
12. Demartis, S., Tarli, L., Borsi, L., Zardi, L., and Neri, D. (2001)
Selective targeting of tumour neovasculature by a radiohaloge-
nated human antibody fragment specific for the ED-B domain
of fibronectin. Eur. J. Nucl. Med. 28, 534–539
13. Halin, C., Rondini, S., Nilsson, F., Berndt, A., Kosmehl, H.,
Zardi, L., and Neri, D. (2002) Enhancement of the antitumor
activity of interleukin-12 by targeted delivery to neovasculature.
Nat. Biotechnol. 20, 264–269
14. Schliemann, C., Palumbo, A., Zuberbuhler, K., Villa, A., Kaspar,
M., Trachsel, E., Klapper, W., Menssen, H. D., and Neri, D.
(2009) Complete eradication of human B-cell lymphoma xeno-
grafts using rituximab in combination with the immunocytokine
L19-IL2. Blood 113, 2275–2283
15. Sauer, S., Erba, P. A., Petrini, M., Menrad, A., Giovannoni, L.,
Grana, C., Hirsch, B., Zardi, L., Paganelli, G., Mariani, G.,
Neri, D., Durkop, H., and Menssen, H. D. (2009) Expression
of the oncofetal ED-B-containing fibronectin isoform in
hematologic tumors enables ED-B-targeted 131I-L19SIP ra-
4543TARGETING TUMOR VESSELS BY THERAPEUTIC VACCINATION
dioimmunotherapy in Hodgkin lymphoma patients. Blood
113, 2265–2274
16. Workman, P., Balmain, A., Hickman, J. A., McNally, N. J., Rohas,
A. M., Mitchison, N. A., Pierrepoint, C. G., Raymond, R.,
Rowlatt, C., Stephens, T. C., and et al. (1988) UKCCCR guide-
lines for the welfare of animals in experimental neoplasia. Lab.
Anim. 22, 195–201
17. Tchougounova, E., Jiang, Y., Brasater, D., Lindberg, N.,
Kastemar, M., Asplund, A., Westermark, B., and Uhrbom, L.
(2009) Sox5 can suppress platelet-derived growth factor
B-induced glioma development in Ink4a-deficient mice
through induction of acute cellular senescence. Oncogene 28,
1537–1548
18. Magnusson, S. E., Pejler, G., Kleinau, S., and Abrink, M. (2009)
Mast cell chymase contributes to the antibody response and the
severity of autoimmune arthritis. FASEB J. 23, 875–882
19. Santimaria, M., Moscatelli, G., Viale, G. L., Giovannoni, L., Neri,
G., Viti, F., Leprini, A., Borsi, L., Castellani, P., Zardi, L., Neri,
D., and Riva, P. (2003) Immunoscintigraphic detection of the
ED-B domain of fibronectin, a marker of angiogenesis, in
patients with cancer. Clin. Cancer Res. 9, 571–579
20. Gundersen, H. J., Bendtsen, T. F., Korbo, L., Marcussen, N.,
Moller, A., Nielsen, K., Nyengaard, J. R., Pakkenberg, B.,
Sorensen, F. B., Vesterby, A., and West, M. J. (1988) Some new,
simple and efficient stereological methods and their use in
pathological research and diagnosis. Apmis 96, 379–394
21. Burton-Wurster, N., Lust, G., and Macleod, J. N. (1997) Carti-
lage fibronectin isoforms: in search of functions for a special
population of matrix glycoproteins. Matrix. Biol. 15, 441–454
22. Peters, J. H., Chen, G. E., and Hynes, R. O. (1996) Fibronectin
isoform distribution in the mouse. II. Differential distribution of
the alternatively spliced EIIIB, EIIIA, and V segments in the
adult mouse. Cell. Adhes. Commun. 4, 127–148
23. Carnemolla, B., Neri, D., Castellani, P., Leprini, A., Neri, G.,
Pini, A., Winter, G., and Zardi, L. (1996) Phage antibodies with
pan-species recognition of the oncofoetal angiogenesis marker
fibronectin ED-B domain. Int. J. Cancer 68, 397–405
24. Castellani, P., Borsi, L., Carnemolla, B., Biro, A., Dorcaratto, A.,
Viale, G. L., Neri, D., and Zardi, L. (2002) Differentiation
between high- and low-grade astrocytoma using a human recom-
binant antibody to the extra domain-B of fibronectin. Am. J.
Pathol. 161, 1695–1700
25. Ma, Y., HayGlass, K. T., Becker, A. B., Fan, Y., Yang, X., Basu, S.,
Srinivasan, G., Simons, F. E., Halayko, A. J., and Peng, Z. (2007)
Novel recombinant interleukin-13 peptide-based vaccine re-
duces airway allergic inflammatory responses in mice. Am. J.
Respir. Crit. Care. Med. 176, 439–445
26. Le Buanec, H., Paturance, S., Couillin, I., Schnyder-Candrian,
S., Larcier, P., Ryffel, B., Bizzini, B., Bensussan, A., Burny, A.,
Gallo, R., Zagury, D., and Peltre, G. (2007) Control of allergic
reactions in mice by an active anti-murine IL-4 immunization.
Vaccine 25, 7206–7216
27. Haller, B. K., Brave, A., Wallgard, E., Roswall, P., Sunkari, V. G.,
Mattson, U., Hallengard, D., Catrina, S. B., Hellstrom, M., and
Pietras, K. (2010) Therapeutic efficacy of a DNA vaccine target-
ing the endothelial tip cell antigen delta-like ligand 4 in
mammary carcinoma. [E-pub ahead of print] Oncogene doi:
10.1038/onc.2010.176
28. Birchler, M. T., Milisavlijevic, D., Pfaltz, M., Neri, D., Odermatt,
B., Schmid, S., and Stoeckli, S. J. (2003) Expression of the extra
domain B of fibronectin, a marker of angiogenesis, in head and
neck tumors. Laryngoscope 113, 1231–1237
29. Hauptmann, S., Zardi, L., Siri, A., Carnemolla, B., Borsi, L.,
Castellucci, M., Klosterhalfen, B., Hartung, P., Weis, J., Stocker,
G., Haubeck, H.-D., and Kirkpatrick, C. J. (1995) Extracellular
matrix proteins in colorectal carcinomas. Expression of tenascin
and fibronectin isoforms. Lab. Invest. 73, 172–182
30. Kaczmarek, J., Castellani, P., Nicolo, G., Spina, B., Allemanni,
G., and Zardi, L. (1994) Distribution of oncofetal fibronectin
isoforms in normal, hyperplastic and neoplastic human breast
tissues. Int. J. Cancer 59, 11–16
31. Ringvall, M., Huijbers, E. J., Ahooghalandari, P., Alekseeva, L.,
Andronova, T., Olsson, A. K., and Hellman, L. (2009) Identifi-
cation of potent biodegradable adjuvants that efficiently break
self-tolerance–a key issue in the development of therapeutic
vaccines. Vaccine 28, 48–52
Received for publication May 11, 2010.
Accepted for publication July 1, 2010.
4544 Vol. 24 November 2010 HUIJBERS ET AL.The FASEB Journal www.fasebj.org
... The splicing dysregulation in tumors can accelerate the development of neoantigen-based immunotherapy, as it provides an expanded candidate pool of antigens for positive selection. For example, fibronectin (FN) encoded by FN1 is a valuable AS-derived antigen resource (Villa et al., 2008;Huijbers et al., 2010;Jailkhani et al., 2019;Xie et al., 2019;Wagner et al., 2021). Through alternative splicing, FN1 can generate three distinct adhesive extracellular matrix isoforms, each with unique structural regions: V (IIICS), EIIIA (EDA), and EIIIB (EDB) (Dubin et al., 1995). ...
... Studies show that fibronectin containing EDA and EDB segments were significantly upregulated during tumor angiogenesis; while displaying low expression levels in normal adult tissues (Khan et al., 2005;Su et al., 2020). Based on this unique AS pattern, the CAR T-cell, mAbs, and cancer vaccine against EDA or EDB have been developed and shown to reduce tumor growth in several solid tumors, including melanoma and lung adenocarcinoma (Villa et al., 2008;Huijbers et al., 2010;Xie et al., 2019;Wagner et al., 2021). It is worth noting that these two splicing derivatives are accumulated in neovasculature, which is present in most solid tumors. ...
Article
Full-text available
Tumor immunotherapy has made great progress in cancer treatment but still faces several challenges, such as a limited number of targetable antigens and varying responses among patients. Alternative splicing (AS) is an essential process for the maturation of nearly all mammalian mRNAs. Recent studies show that AS contributes to expanding cancer-specific antigens and modulating immunogenicity, making it a promising solution to the above challenges. The organoid technology preserves the individual immune microenvironment and reduces the time/economic costs of the experiment model, facilitating the development of splicing-based immunotherapy. Here, we summarize three critical roles of AS in immunotherapy: resources for generating neoantigens, targets for immune-therapeutic modulation, and biomarkers to guide immunotherapy options. Subsequently, we highlight the benefits of adopting organoids to develop AS-based immunotherapies. Finally, we discuss the current challenges in studying AS-based immunotherapy in terms of existing bioinformatics algorithms and biological technologies.
... This approach makes use of conjugation of the target of interest to a foreign protein, such as the truncated form of bacterial thioredoxin (TRXtr) [10]. The conjugate vaccine technology has been shown to efficiently induce an antibody-based immune response against vascular-specific self-proteins [7,8,[10][11][12]. It is important to mention that in addition to an efficient immunization strategy, the co-administration of a potent adjuvant is needed to generate this immune response. ...
... When anti-vimentin antibodies bind their target in the tumor vasculature, the Fc region of the antibody can bind to an activating FcγR on the surface of several types of immune cells, such as macrophages, neutrophils, and NK cells. The activation of immune cells can trigger effector functions, which eventually leads to tumor vessel destruction, as previously suggested for ED-B [11]. When quantifying the number of blood vessels in B16F10 tumor sections of TRXtr-Vim-vaccinated mice, we indeed found a significantly decreased vessel density ( Figure 3D). ...
Article
Full-text available
Simple Summary Vaccination against specific proteins in the tumor vasculature has already shown promising results in several preclinical studies. However, the efficacy of vaccination highly depends on the adjuvant used. This study aimed to assess the potential use of the biodegradable adjuvant Montanide ISA 720 in combination with our vaccine against extracellular vimentin, a protein specifically secreted by the tumor vasculature. Compared to the potent but toxic Freund’s adjuvant, Montanide showed a comparable immune response and tumor growth inhibition in a preclinical vaccination experiment in mice, especially when supplemented with the immune stimulatory molecule CpG. We also observed that vaccination reduced the blood vessel count and increased the infiltration of immune cells. We conclude that Montanide ISA 720 shows potential to be used as an adjuvant for vaccination against extracellular vimentin for future clinical studies in cancer patients. Abstract Extracellular vimentin is a specific marker of the tumor vasculature, where it is secreted by tumor endothelial cells. Vaccination with a conjugate vaccine targeting extracellular vimentin was previously shown to induce a potent humoral immune response and tumor growth inhibition in mice. These data were obtained by vaccination using the toxic Freund’s adjuvant (FA) and are therefore not directly translatable into the clinic. In the present study, we aimed to investigate the potential of the biodegradable Montanide ISA 720 adjuvant. We tested Montanide either alone (MN) or supplemented with CpG 1826 (MN-C). Both adjuvant compositions, as well as FA, resulted in a significant tumor growth inhibition and decreased vessel density in the B16F10 melanoma tumor model. Vaccination of mice with either FA or MN-C resulted in an equally potent humoral immune response towards vimentin, while the antibody titers obtained with MN alone were significantly lower compared to FA. Vaccination coincided with the infiltration of immune cells. The highest number of intratumoral immune cells was seen in tumors from the MN-C group. Therefore, we conclude that Montanide ISA 720 supplemented with CpG allows efficient vaccination against extracellular vimentin, which is a prerequisite for the transfer of the vaccine into the clinic.
... For instance, TCVs containing the Spam1 gene express hyaluronidase, degrading the ECM, enhancing blood flow, reducing tumor hypoxia, and modulating the immune environment, thereby elevating antitumor efficiency and reducing tumor recurrence risk [140]. In addition, TCVs targeting the EDA domain of FN have shown potential in curbing metastatic tumor progression [141], while those targeting the EDB of FN effectively diminish the bulk of solid tumors [142][143][144]. Moreover, an orally administered DNA vaccine targeting FAP-depleted CAFs decreases type I collagen content, restricting the growth and metastasis of primary tumor cells promoting multidrug resistance in mice [145]. ...
Article
Full-text available
The interplay between extracellular matrix (ECM) stiffness and the tumor microenvironment is increasingly recognized as a critical factor in cancer progression and the efficacy of immunotherapy. This review comprehensively discusses the key factors regulating ECM remodeling, including the activation of cancer-associated fibroblasts and the accumulation and crosslinking of ECM proteins. Furthermore, it provides a detailed exploration of how ECM stiffness influences the behaviors of both tumor and immune cells. Significantly, the impact of ECM stiffness on the response to various immunotherapy strategies, such as immune checkpoint blockade, adoptive cell therapy, oncolytic virus therapy, and therapeutic cancer vaccines, is thoroughly examined. The review also addresses the challenges in translating research findings into clinical practice, highlighting the need for more precise biomaterials that accurately mimic the ECM and the development of novel therapeutic strategies. The insights offered aim to guide future research, with the potential to enhance the effectiveness of cancer immunotherapy modalities.
... Antibodies bound to the target in the tumor vasculature induce antibody-dependent cell cytotoxicity (ADCC) in endothelial cells, executed by Fc-receptor-expressing white blood cells. In previous studies to another target, this was shown to happen in the form of 'frustrated phagocytosis' [34]. Also, bound antibodies are expected to induce the complement system, leading to the generation of the membrane attack complex and the subsequent lysis of endothelial cells. ...
Article
Full-text available
Simple Summary In this study, we investigated the safety and usefulness of treatment of dog patients with spontaneous bladder cancer with the CVx1 vaccine. The vaccine is directed against a specific protein, named extracellular vimentin, excreted by the tumor vasculature. Twenty dogs diagnosed at the veterinary clinic with bladder cancer were treated with the CVx1 vaccine in combination with the non-steroidal anti-inflammatory drug (NSAID) meloxicam. All dogs responded to the treatment and developed an immune response towards the tumor vasculature. After treatment with the CVx1 vaccine plus meloxicam, the survival was almost doubled (374 days) compared to the historical control group (196 days) treated with the chemotherapy carboplatin in combination with the NSAID piroxicam. Treatment with the CVx1 vaccine combined with meloxicam was safe and well tolerated. Our results justify further development of the CVx1 vaccine for the treatment of human patients in the clinic. Abstract It was recently shown that targeting extracellular vimentin (eVim) is safe and effective in preclinical models. Here, we report the safety and efficacy in client-owned dogs with spontaneous bladder cancer of CVx1, an iBoost technology-based vaccine targeting eVim in combination with COX-2 inhibition. This was a single-arm prospective phase 1/2 study with CVx1 in 20 client-owned dogs with spontaneous UC which involved four subcutaneous vaccinations with CVx1 at 2-week intervals for induction of antibody titers, followed by maintenance vaccinations at 2-month intervals. Additionally, daily cyclooxygenase (COX)-2 inhibition with meloxicam was given. The response was assessed by antibody titers, physical condition, abdominal ultrasound and thorax X-ray. The primary endpoints were the development of antibody titers, as well as overall survival compared to a historical control group receiving carboplatin and COX-2 inhibition with piroxicam. Kaplan–Meier survival analysis was performed. All dogs developed antibodies against eVim. Titers were adequately maintained for the duration of this study. A median overall survival of 374 days was observed, which was 196 days for the historical control group (p < 0.01). Short-term grade 1–2 toxicity at the injection site and some related systemic symptoms peri-vaccination were observed. No toxicity was observed related to the induced antibody response. A limitation of this study is the single-arm prospective setting. CVx1 plus meloxicam consistently induced efficient antibody titers, was well tolerated and showed prolonged survival. The results obtained merit further development for human clinical care.
... Moreover, Fn-EDA or Fn-EDB can stimulate the production of proinflammatory cytokines by DCs and induce their maturation in vitro and in vivo, which improves the induction of specific T cell immunity against tumors. A recombinant fusion protein using Fn-EDB segments was reported to directly serve as a vaccine for solid tumors, and mice immunized with the fusion protein in Freund's adjuvant showed approximately 70% reduction in subcutaneous fibrosarcoma tumors [92]. In another study, the authors showed that the number of metastases was reduced in a breast cancer mouse model carrying anti-ED-A antibodies [93]. ...
Article
Full-text available
As a major component of the stromal microenvironment of various solid tumors, the ex-tracellular matrix (ECM) has attracted increasing attention in cancer-related studies. ECM in the tumor stroma not only provides an external barrier and framework for tumor cell adhesion and movement, but also acts as an active regulator that modulates the tumor microenvironment, including stromal immunity. Fibronectin (Fn), as a core component of the ECM, plays a key role in the assembly and remodeling of the ECM. Hence, understanding the role of Fn in the modulation of tumor stromal immunity is of great importance for cancer immunotherapy. Hence, in-depth studies on the underlying mechanisms of Fn in tumors are urgently needed to clarify the current understanding and issues and to identify new and specific targets for effective diagnosis and treatment purposes. In this review, we summarize the structure and role of Fn, its potent derivatives in tumor stromal immunity, and their biological effects and mechanisms in tumor development. In addition, we discuss the novel applications of Fn in tumor treatment. Therefore, this review can provide prospective insight into Fn immunotherapeutic applications in tumor treatment.
... Moreover, Fn-EDA or Fn-EDB can stimulate the production of proinflammatory cytokines by DCs and induce their maturation in vitro and in vivo, which improves the induction of specific T cell immunity against tumors. A recombinant fusion protein using Fn-EDB segments was reported to directly serve as a vaccine for solid tumors, and mice immunized with the fusion protein in Freund's adjuvant showed approximately 70% reduction in subcutaneous fibrosarcoma tumors [92]. In another study, the authors showed that the number of metastases was reduced in a breast cancer mouse model carrying anti-ED-A antibodies [93]. ...
Article
As a major component of the stromal microenvironment of various solid tumors, the extracellular matrix (ECM) has attracted increasing attention in cancer-related studies. ECM in the tumor stroma not only provides an external barrier and framework for tumor cell adhesion and movement, but also acts as an active regulator that modulates the tumor microenvironment, including stromal immunity. Fibronectin (Fn), as a core component of the ECM, plays a key role in the assembly and remodeling of the ECM. Hence, understanding the role of Fn in the modulation of tumor stromal immunity is of great importance for cancer immunotherapy. Hence, in-depth studies on the underlying mechanisms of Fn in tumors are urgently needed to clarify the current understanding and issues and to identify new and specific targets for effective diagnosis and treatment purposes. In this review, we summarize the structure and role of Fn, its potent derivatives in tumor stromal immunity, and their biological effects and mechanisms in tumor development. In addition, we discuss the novel applications of Fn in tumor treatment. Therefore, this review can provide prospective insight into Fn immunotherapeutic applications in tumor treatment.
... The current commercial success of targeting the vasculature indirectly-through interference with tumor-derived angiogenic growth factors by antibodies and tyrosine kinase inhibitors-is overshadowed by the occurrence of drug-induced resistance, resulting from the adaptation and alternative growth factor production of tumor cells 6,7 . We have shown that direct targeting of tumor endothelium, by vaccination or antibodies towards tumor endothelial-specific markers, is a highly effective strategy for inhibiting tumor growth and can potentially overcome EC anergy [8][9][10][11] . As such, targeting tumor blood vessels has the capacity to improve immunotherapy and may even act as immunotherapy in itself 5,12 . ...
Article
Full-text available
Anti-angiogenic cancer therapies possess immune-stimulatory properties by counteracting pro-angiogenic molecular mechanisms. We report that tumor endothelial cells ubiquitously overexpress and secrete the intermediate filament protein vimentin through type III unconventional secretion mechanisms. Extracellular vimentin is pro-angiogenic and functionally mimics VEGF action, while concomitantly acting as inhibitor of leukocyte-endothelial interactions. Antibody targeting of extracellular vimentin shows inhibition of angiogenesis in vitro and in vivo. Effective and safe inhibition of angiogenesis and tumor growth in several preclinical and clinical studies is demonstrated using a vaccination strategy against extracellular vimentin. Targeting vimentin induces a pro-inflammatory condition in the tumor, exemplified by induction of the endothelial adhesion molecule ICAM1, suppression of PD-L1, and altered immune cell profiles. Our findings show that extracellular vimentin contributes to immune suppression and functions as a vascular immune checkpoint molecule. Targeting of extracellular vimentin presents therefore an anti-angiogenic immunotherapy strategy against cancer. The pro-tumorigenic effects of vimentin have been attributed to intracellular functions in tumour cells so far. Here, the authors show that tumour endothelial cells can secrete vimentin as a pro-angiogenic factor and that targeting of vimentin can be used as an immunotherapeutic strategy.
Article
Background: CAR-T cell immunotherapy has achieved remarkable success in malignant B-cell malignancies, but progress in solid tumors is slow, and one of the key reasons is the lack of ideal targets. Cancer-specific extra domain B of fibronectin (EDB-FN) is widely upregulated in solid tumors and expressed at low levels in normal tissues. Many imaging and targeted cancer therapies based on EDB-FN targets have been developed and tested in clinical trials, making EDB-FN an ideal target for immunotherapy. Methods: We constructed two EDB-FN-targeted CAR-Ts based on the peptide APT0 and the single-chain antibody CGS2 in a lentiviral infection manner for the first time. Luciferase cytotoxicity assay to assess CAR-T killing of tumor cells. An enzyme-linked immunosorbent assay was used to detect the release of the cytokine IFN-γ. Fluorescence imaging to evaluate the dynamics of CAR-T cell and tumor cell coculture. Knockdown assays were used to validate the target specificity of CAR-T cells. Results: In this research, two CAR-Ts targeting EDB-FN, APT0 CAR-T, and CGS2 CAR-T, were constructed. In vitro, both CAR-T cells produced broad-spectrum killing of multiple EDB-FN-positive solid tumor cell lines and were accompanied by cytokine IFN-γ release. Regarding safety, the two CAR-T cells did not affect T cells' normal growth and proliferation and were not toxic to HEK-293T human embryonic kidney epithelial cells. Conclusion: APT0 CAR-T and CGS2 CAR-T cells are two new CAR-Ts targeting EDB-FN. Both CAR-T cells can successfully identify and specifically kill various EDB-FN-positive solid tumor cells with potential clinical applications.
Chapter
Vascular basement membrane is a critical regulator of angiogenesis. Fibronectin, a component of the vascular matrix, acts as a scaffold between the endothelial and perivascular cells that guide the assembly of other matrix components like the laminins and collagen IV. Fibronectin is unique in being the only extracellular matrix protein present in both soluble and insoluble forms. Fibronectin is secreted as a dimeric glycoprotein that can self-assemble into a network of fibrils. The process of fibronectin fibrillogenesis is carefully tuned according to extracellular cues to regulate the multistep angiogenic processes. This chapter details the regulators of fibronectin remodelling in endothelial cells and the role of fibronectin secretion and fibrillogenesis in guiding physiological and pathophysiological angiogenesis.KeywordsFibronectinAtherosclerosisFibrillogenesisAngiogenesisWound healing
Article
Chimeric antigen receptor (CAR T) cell treatment for solid tumours faces significant challenges. CAR T cells are unable to pass the vascular barrier in tumours due to a lack of endothelial leukocyte adhesion molecules. The invasion, activity, and durability of CAR T cells may be hampered by additional immunosuppressive mechanisms present in the solid tumour environment. The use of CAR T cells to attack cancer vascular endothelial metabolic targets from within the blood may simplify the fight against cancer. These are the principles that govern our examination of CAR T cell treatment for tumor cells, with a specific eye toward tumour venous delivery. CAR T cells may also be designed such that they can be readily, safely, and successfully transferred.
Article
Full-text available
The Notch ligand delta-like ligand 4 (DLL4) is an essential component expressed by endothelial tip cells during angiogenic sprouting. We have described a conceptually novel therapeutic strategy for targeting tumor angiogenesis and endothelial tip cells based on DNA vaccination against DLL4. Immunization with DLL4-encoding plasmid DNA by in vivo electroporation severely retarded the growth of orthotopically implanted mammary carcinomas in mice by induction of a nonproductive angiogenic response. Mechanistically, vaccination brought about a break in tolerance against the self-antigen, DLL4, as evidenced by the production of inhibitory and inherently therapeutic antibodies against mouse DLL4. Importantly, no evidence for a delayed wound healing response, or for toxicity associated with pharmacological blockade of DLL4 signaling, was noted in mice immunized with the DLL4 vaccine. We have thus developed a well-tolerated DNA vaccination strategy targeting the endothelial tip cells and the antigen DLL4 with proven therapeutic efficacy in mouse models of mammary carcinoma; a disease that has been reported to dramatically induce the expression of DLL4. Conceivably, induction of immunity toward principal mediators of pathological angiogenesis could provide protection against recurrent malignant disease in the adjuvant setting.
Article
Full-text available
Fibronectin is an extracellular matrix protein found only in vertebrate organisms containing endothelium-lined vasculature and is required for cardiovascular development in fish and mice. Fibronectin and its splice variants containing EIIIA and EIIIB domains are highly upregulated around newly developing vasculature during embryogenesis and in pathological conditions including atherosclerosis, cardiac hypertrophy, and tumorigenesis. However, their molecular roles in these processes are not entirely understood. We review genetic studies examining functions of fibronectin and its splice variants during embryonic cardiovascular development, and discuss potential roles of fibronectin in vascular disease and tumor angiogenesis.
Article
Purification and amino acid sequence analysis of a proteolytic fragment of fibronectin (FN) from transformed human cells demonstrated that a high percentage of these FN molecules contains an extra amino acid sequence which is present only in a very low percentage of FN molecules from normal fibroblasts and is undetectable in plasma FN. This new amino acid sequence introduces into the FN molecule a site very sensitive to a number of proteolytic enzymes. By analyzing the cellular mRNA and genomic clones, we have demonstrated that this sequence derives from a differential splicing pattern of the FN mRNA precursors, which leads in transformed cells to a high‐level expression of an extra type III homology repeat (ED‐B) coded for by a previously unobserved exon. Here we also report the complete sequence of this new exon. These results demonstrate that in malignant cells the mechanisms regulating the splicing of FN mRNA precursors are altered.
Article
Fibronectin (FN) represents the mixture of a number of structurally different molecules (isoforms) whose make-up varies depending on the FN sources. FN from cultured transformed human cells has a very different isoform composition with respect to its normal counterpart. In fact, SV-40-transformed WI-38VAI3 human fibroblasts produce high levels of a FN isoform (B-FN) which is very poorly expressed in their normal, WI-38, counterpart. We have recently demonstrated that the B-FN isoform derives from a differential splicing pattern of the FN primary transcript which leads, in transformed cells, to a high level expression of the exon ED-B (Zardi, L., B. Carnemolla, A. Siri, T. E. Petersen, G. Paolella, G. Sebastio, and F. E. Baralle. 1987. EMBO (Eur. Mol. Biol. Organ.) J. 6:2337-2342). Here we report on the production and characterization of a monoclonal antibody (BC-1) which recognizes an epitope within the protein sequence coded for by the ED-B exon. This monoclonal antibody makes it possible to carry out immunohistochemical analysis of the distribution of the ED-B-containing FN isoform (B-FN) in human tissues. The results show that while in normal, adult, human tissues total FN has a widespread distribution, the B-FN isoform is restricted only to synovial cells, to some vessels and areas of the interstitium of the ovary, and to the myometrium. On the contrary, the B-FN isoform has a much greater expression in fetal and tumor tissues. These results demonstrate that, in vivo, different FN isoforms have a differential distribution and indicate that the B-FN isoform may play a role in ontogenesis and oncogenetic processes.
Article
Different fibronectin (FN) isoforms are generated by the alternative splicing of 3 regions (ED-A, ED-B and IIICS) of the primary transcript. The FN isoform containing the ED-B sequence, a complete type-III-homology repeat, while having extremely restricted distribution in normal adult tissues, reveals high expression in fetal and tumor tissues. Using the monoclonal antibody (MAb) BC-1, specific for the FN isoform containing the ED-B sequence (B+·FN), we demonstrate here, using immunohistochemical techniques, that while this FN isoform is undetectable in mature vessels, it is highly expressed during angiogenesis both in neoplastic and in normal tissues, as in the case of the functional layer of endometrium during the proliferative phase. B+·FN is thus a marker for the formation of new vessels, and the BC-I MAb may be a useful reagent for evaluating the level of the angiogenetic process in different neoplasms.
Article
Stereology is a set of simple and efficient methods for quantitation of three-dimensional microscopic structures which is specifically tuned to provide reliable data from sections. Within the last few years, a number of new methods has been developed which are of special interest to pathologists. Methods for estimating the volume, surface area and length of any structure are described in this review. The principles on which stereology is based and the necessary sampling procedures are described and illustrated with examples. The necessary equipment, the measurements, and the calculations are invariably simple and easy.
Article
Monoclonal antibodies are used successfully in the treatment of many human disorders. However, these antibodies are expensive and have in many countries put a major strain on the health care economy. Therapeutic vaccines, directed against the same target molecules, may offer a solution to this problem. Vaccines usually involve lower amount of recombinant protein, approximately 10,000-20,000 times less, which is significantly more cost effective. Attempts to develop such therapeutic vaccines have also been made. However, their efficacy has been limited by the lack of potent immunostimulatory compounds, adjuvants, for human use. To address this problem we have conducted a broad screening for adjuvants that can enhance the efficacy of therapeutic vaccines, whilst at the same time being non-toxic and biodegradable. We have now identified adjuvants that show these desired characteristics. A combination of Montanide ISA720 and phosphorothioate stabilized CpG stimulatory DNA, induced similar or even higher anti-self-antibody titers compared to Freund's adjuvant, currently the most potent, but also toxic, adjuvant available. This finding removes one of the major limiting factors in the field and facilitates the development of a broad range of novel therapeutic vaccines.
Article
SOX5 is a member of the high-mobility group superfamily of architectural non-histone proteins involved in gene regulation and maintenance of chromatin structure in a wide variety of developmental processes. Sox5 was identified as a brain tumor locus in a retroviral insertional mutagenesis screen of platelet-derived growth factor B (PDGFB)-induced mouse gliomas. Here we have investigated the role of Sox5 in PDGFB-induced gliomagenesis in mice. We show that Sox5 can suppress PDGFB-induced glioma development predominantly upon Ink4a-loss. In human glioma cell lines and tissues, we found very low levels of SOX5 compared with normal brain. Overexpression of Sox5 in human glioma cells led to a reduction in clone formation and inhibition of proliferation. Combined expression of Sox5 and PDGFB in primary brain cell cultures caused decreased proliferation and an increased number of senescent cells in the Ink4a-/- cells only. Protein analyses showed a reduction in the amount and activation of Akt and increased levels of p27(Kip1) upon Sox5 expression that was dominant to PDGFB signaling and specific to Ink4a-/- cells. Upon inhibition of p27(Kip1), the effects of Sox5 on proliferation and senescence could be reversed. Our data suggest a novel pathway, where Sox5 may suppress the oncogenic effects of PDGFB signaling during glioma development by regulating p27(Kip1) in a p19(Arf)-dependent manner, leading to acute cellular senescence.